51
|
Grippo AJ, McNeal N, Normann MC, Colburn W, Dagner A, Woodbury M. Behavioral and neuroendocrine consequences of disrupting a long-term monogamous social bond in aging prairie voles. Stress 2021; 24:239-250. [PMID: 32820956 PMCID: PMC7914264 DOI: 10.1080/10253890.2020.1812058] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 08/14/2020] [Indexed: 12/13/2022] Open
Abstract
Social support from a spouse, long-term partner, or someone who provides emotional or instrumental support may protect against consequences of aging, including mediating behavioral stress reactivity and altering neurobiological process that underlie short-term stress responses. Therefore, long-term social bonding may have behavioral and neurobiological benefits. The socially monogamous prairie vole provides a valuable experimental model for investigating the benefits of long-term social bonds on short-term stress reactivity in aging animals, given their unique social structure of forming enduring opposite-sex bonds, living in family groups, and bi-parental rearing strategies. Male-female pairs of long-term, cohabitating prairie voles were investigated for short-term behavioral and neuroendocrine stress reactivity following either long-term social pairing (control), or a period of social isolation. In Experiment 1, social isolation was associated with altered behavioral reactivity to an acute swim stressor, and greater neural activation in the hypothalamic paraventricular nucleus, as well as specifically the parvocellular region, following the swim stressor (vs. control). In Experiment 2, social isolation was associated with greater corticosterone reactivity following an acute restraint stressor (vs. control). No sex differences were observed. Exploratory correlation and subgroup analyses revealed systematic relationships among various demographic variables (such as age of the subjects, amount of time the pair cohabitated together, and number of litters the pair reared together) and the behavioral and neuroendocrine outcome measures. These findings may inform our understanding of the benefits of long-term social bonding on modulating short-term behavioral and neuroendocrine responses to stress.LAY SUMMARYReceiving social support from a long-term spouse or partner, or having a strong support network from friends, may have important health benefits as people age. In aging monogamous prairie voles, social isolation from a long-term social partner disrupted behaviors and short-term stress responses, whereas living with a long-term partner protected against these disruptions. This research is important for our understanding of the benefits of social support on stress responses as we age.
Collapse
Affiliation(s)
- Angela J Grippo
- Department of Psychology, Northern Illinois University, DeKalb, IL, USA
| | - Neal McNeal
- Department of Psychology, Northern Illinois University, DeKalb, IL, USA
| | - Marigny C Normann
- Department of Psychology, Northern Illinois University, DeKalb, IL, USA
| | - William Colburn
- Department of Psychology, Northern Illinois University, DeKalb, IL, USA
| | - Ashley Dagner
- Department of Psychology, Northern Illinois University, DeKalb, IL, USA
| | - Matthew Woodbury
- Department of Psychology, Northern Illinois University, DeKalb, IL, USA
| |
Collapse
|
52
|
Zhang WH, Zhang JY, Holmes A, Pan BX. Amygdala Circuit Substrates for Stress Adaptation and Adversity. Biol Psychiatry 2021; 89:847-856. [PMID: 33691931 DOI: 10.1016/j.biopsych.2020.12.026] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/24/2020] [Accepted: 12/18/2020] [Indexed: 12/19/2022]
Abstract
Brain systems that promote maintenance of homeostasis in the face of stress have significant adaptive value. A growing body of work across species demonstrates a critical role for the amygdala in promoting homeostasis by regulating physiological and behavioral responses to stress. This review focuses on an emerging body of evidence that has begun to delineate the contribution of specific long-range amygdala circuits in mediating the effects of stress. After summarizing the major anatomical features of the amygdala and its connectivity to other limbic structures, we discuss recent findings from rodents showing how stress causes structural and functional remodeling of amygdala neuronal outputs to defined cortical and subcortical target regions. We also consider some of the environmental and genetic factors that have been found to moderate how the amygdala responds to stress and relate the emerging preclinical literature to the current understanding of the pathophysiology and treatment of stress-related neuropsychiatric disorders. Future effort to translate these findings to clinics may help to develop valuable tools for prevention, diagnosis, and treatment of these diseases.
Collapse
Affiliation(s)
- Wen-Hua Zhang
- Laboratory of Fear and Anxiety Disorders, Institutes of Life Science, Nanchang University, Nanchang, China
| | - Jun-Yu Zhang
- Laboratory of Fear and Anxiety Disorders, Institutes of Life Science, Nanchang University, Nanchang, China
| | - Andrew Holmes
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institues of Health, Bethesda, Maryland
| | - Bing-Xing Pan
- Laboratory of Fear and Anxiety Disorders, Institutes of Life Science, Nanchang University, Nanchang, China.
| |
Collapse
|
53
|
Verspeek J, Behringer V, Laméris DW, Murtagh R, Salas M, Staes N, Deschner T, Stevens JMG. Time-lag of urinary and salivary cortisol response after a psychological stressor in bonobos (Pan paniscus). Sci Rep 2021; 11:7905. [PMID: 33846457 PMCID: PMC8041877 DOI: 10.1038/s41598-021-87163-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 03/23/2021] [Indexed: 02/01/2023] Open
Abstract
Cortisol is often measured as a marker for stress. Therefore, a profound validation of the time-lag between the stressor and the increase and peak in cortisol levels is needed. No study measured both the urinary and salivary cortisol time-lag after a psychological stressor. In this study, we used a frequent sampling study design to (1) describe the urinary and salivary cortisol pattern during a control day; and (2) characterize the induced excretion pattern of urinary and salivary cortisol after a psychological stressor in six zoo-housed bonobos. Liquid chromatography-tandem mass spectrometry was used to analyze 71 urine and 162 saliva samples collected on a control and a test day. We found that the time-lag between the stressor and the maximal cortisol concentration was similar in urine and saliva (160 min after the stressor). However, salivary cortisol after the stressor did show a faster and steeper increase than urinary cortisol. We also show inter-individual variation in the baseline and stress levels of cortisol, which should be considered in future cortisol studies. Our research highlights the importance of validation studies to confirm relevant sampling windows for cortisol sampling in order to obtain biologically meaningful results.
Collapse
Affiliation(s)
- Jonas Verspeek
- Behavioural Ecology and Ecophysiology Group, Department of Biology, University of Antwerp, Antwerp, Belgium.
- Antwerp ZOO Centre for Research & Conservation (CRC), Royal Zoological Society of Antwerp (RZSA), Antwerp, Belgium.
| | - Verena Behringer
- Interim Group Primatology, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
- Endocrinology Laboratory, German Primate Center, Leibniz Institute for Primate Research, Göttingen, Germany
| | - Daan W Laméris
- Behavioural Ecology and Ecophysiology Group, Department of Biology, University of Antwerp, Antwerp, Belgium
- Antwerp ZOO Centre for Research & Conservation (CRC), Royal Zoological Society of Antwerp (RZSA), Antwerp, Belgium
| | - Róisín Murtagh
- Interim Group Primatology, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Marina Salas
- Antwerp ZOO Centre for Research & Conservation (CRC), Royal Zoological Society of Antwerp (RZSA), Antwerp, Belgium
| | - Nicky Staes
- Behavioural Ecology and Ecophysiology Group, Department of Biology, University of Antwerp, Antwerp, Belgium
- Antwerp ZOO Centre for Research & Conservation (CRC), Royal Zoological Society of Antwerp (RZSA), Antwerp, Belgium
| | - Tobias Deschner
- Interim Group Primatology, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Jeroen M G Stevens
- Behavioural Ecology and Ecophysiology Group, Department of Biology, University of Antwerp, Antwerp, Belgium
- SALTO, Agro- and Biotechnology, Odisee University College, Brussels, Belgium
| |
Collapse
|
54
|
Kang JWM, Mor D, Keay KA. Nerve injury alters restraint-induced activation of the basolateral amygdala in male rats. Brain Struct Funct 2021; 226:1209-1227. [PMID: 33582845 DOI: 10.1007/s00429-021-02235-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 01/28/2021] [Indexed: 01/05/2023]
Abstract
The amygdala is critical for the production of appropriate responses towards emotional or stressful stimuli. It has a characteristic neuronal activation pattern to acute stressors. Chronic pain and acute stress have each been shown to independently modulate the activity of the amygdala. Few studies have investigated the effect of pain or injury, on amygdala activation to acute stress. This study investigated the effects of a neuropathic injury on the activation response of the amygdala to an acute restraint stress. Chronic constriction injury of the right sciatic nerve (CCI) was used to create neuropathic injury and a single brief 15-min acute restraint was used as an emotional/psychological stressor. All rats received cholera toxin B (CTB) retrograde tracer injections into the medial prefrontal cortex (mPFC) to assess if the amygdala to mPFC pathway was specifically regulated by the combination of neuropathic injury and acute stress. To assess differential patterns of activity in amygdala subregions, cFos expression was used as a marker for "acute", restraint triggered neuronal activation, and FosB/ΔFosB expression was used to reveal prolonged neuronal activation/sensitisation triggered by CCI. Restraint resulted in a characteristic increase in cFos expression in the medial amygdala, which was not altered by CCI. Rats with a CCI showed increased cFos expression in the basolateral amygdala (BLA), in response to an acute restraint stress, but not in neurons projecting to the prefrontal cortex. Further, CCI rats showed an increase in FosB/ΔFosB expression which was exclusive to the BLA. This increase likely reflects sensitisation of the BLA as a consequence of nerve injury which may contribute to heightened sensitivity of BLA neurons to acute emotional/ psychological stressors.
Collapse
Affiliation(s)
- James W M Kang
- Faculty of Medicine and Health, School of Medical Sciences (Neuroscience), The University of Sydney, Sydney, NSW, 2006, Australia. .,Brain and Mind Centre (M02G), 100 Mallet Street, Camperdown, NSW, 2050, Australia.
| | - David Mor
- Faculty of Medicine and Health, School of Medical Sciences (Neuroscience), The University of Sydney, Sydney, NSW, 2006, Australia
| | - Kevin A Keay
- Faculty of Medicine and Health, School of Medical Sciences (Neuroscience), The University of Sydney, Sydney, NSW, 2006, Australia.,Brain and Mind Centre (M02G), 100 Mallet Street, Camperdown, NSW, 2050, Australia
| |
Collapse
|
55
|
Henderson LA, Macefield VG. The role of the dorsomedial and ventromedial hypothalamus in regulating behaviorally coupled and resting autonomic drive. HANDBOOK OF CLINICAL NEUROLOGY 2021; 180:187-200. [PMID: 34225929 DOI: 10.1016/b978-0-12-820107-7.00012-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Nearly a century ago it was reported that stimulation of the hypothalamus could evoke profound behavioral state changes coupled with altered autonomic function. Since these initial observations, further studies in animals have revealed that two hypothalamic regions-the dorsomedial and ventromedial hypothalamic nuclei-are critical for numerous behaviors, including those in response to psychological stressors. These behaviors are coupled with changes in autonomic functions, such as altered blood pressure, heart rate, sympathetic nerve activity, resetting of the baroreflex and changes in pituitary function. There is also growing evidence that these two hypothalamic regions play a critical role in thermogenesis, and suggestions they could also be responsible for the hypertension associated with obesity. The aim of this chapter is to review the anatomy, projection patterns, and function of the dorsomedial and ventromedial hypothalamus with a particular focus on their role in autonomic regulation. While most of what is known about these two hypothalamic regions is derived from laboratory animal experiments, recent human studies will also be explored. Finally, we will describe recent human brain imaging studies that provide evidence of a role for these hypothalamic regions in setting resting sympathetic drive and their potential role in conditions such as hypertension.
Collapse
Affiliation(s)
- Luke A Henderson
- Department of Anatomy & Histology, Brain and Mind Centre, University of Sydney, Sydney, NSW, Australia.
| | - Vaughan G Macefield
- Baker Heart & Diabetes Institute, Melbourne, VIC, Australia; Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
56
|
Morphine-Conditioned Placebo Analgesia in Female and Male Rats with Chronic Neuropathic Pain: c-Fos Expression in the Rostral Ventromedial Medulla. Neuroscience 2020; 457:51-73. [PMID: 33285237 DOI: 10.1016/j.neuroscience.2020.11.038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/19/2020] [Accepted: 11/23/2020] [Indexed: 12/17/2022]
Abstract
Placebo analgesia has great potential to overcome the inadequacies of current drug therapies to treat conditions of chronic pain. The rostral ventromedial medulla (RVM) has been implicated as a critical relay in the antinociceptive pathway underpinning placebo analgesia in humans. We developed a model of opiate-conditioned placebo analgesia in rats with neuropathic injury to identify medullary nuclei active during placebo analgesia. Using female and male rats the degree of thermal allodynia was first determined following nerve injury, and a pharmacological conditioning procedure, pairing contextual cues with the experience of morphine-induced analgesia, was used to elicit placebo analgesic reactions. This protocol revealed clear subpopulations of placebo reactors (36% of males, 25% of females) and non-reactors in proportions similar to those reported in human studies. We detected injury-specific c-Fos expression in the gracile nucleus and morphine-specific c-Fos expression in the serotonergic midline raphe nuclei and the caudal nuclei of the solitary tract. However, c-Fos expression did not differ between placebo reactors and non-reactors in either serotonergic or non-serotonergic neurons of the RVM. Despite a subpopulation of rats demonstrating placebo reactions, we found no evidence for enhanced activity in the nuclei from which the classical RVM → spinal cord descending analgesic pathways emerge.
Collapse
|
57
|
Li M, Chen S, Gao Z, Wu W, Xu L. Physiological explicit of delayed psychological stress response induced by extra neural regulation. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2020; 196:105610. [PMID: 32599339 DOI: 10.1016/j.cmpb.2020.105610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 06/12/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND AND OBJECTIVE Acute psychological stress is conducted along a different neural pathway from acute physical stress and causes a conduction delay. This conduction delay reflected on the central nervous system (CNS) as an evaluation process, which allows the human body to make the best response to a stressor based on preceding experiences. However, how this conduction delay reflected on physiological explicit, remains to be studied. METHODS In this work, the variation of nine traditional heart rate variability (HRV) features are studied from twenty-six healthy subjects during acute physical (Cold pressor test (CPT)) and psychological (Stroop color word test (SCWT)) stress on separate occasions. Besides, we discussed ultra-short and short-term HRV analysis and their relationship with acute stress. RESULTS The experimental results suggest that there was a delayed response of HRV features under acute psychological stress compared with acute physical stress. In addition, during the later stage of stress, four features could differentiate between baseline and acute psychological stress (p < 0.05): SDNN, LFpower, HFpower, LF/HF, but these features do not have significant difference during acute physical stress (p > 0.05). CONCLUSIONS Due to the delayed psychological stress response induced by extra neural regulation, a significant change of HRV features was found at the third minute of psychological stress stage. And the variation of these features have linear correlation with time during the remaining three to five minutes stress stage (r = -0.987, 0.996, -0.952, 0.938 for SDNN, LFpower, HFpower, LF/HF).
Collapse
Affiliation(s)
- Ming Li
- College of Electrical Automation and Information Engineering, Tianjin University, Weijin Road 92, Tianjin 300110, China
| | - Shixiong Chen
- Institute of Biomedical and Health Engineering, SIAT, Chinese Academy of Sciences (CAS), Xueyuan Avenue 1068, Shenzhen 518055, China
| | - Zhen Gao
- College of Electrical Automation and Information Engineering, Tianjin University, Weijin Road 92, Tianjin 300110, China.
| | - Wanqing Wu
- School of Biomedical Engineering, Sun Yat-Sen University, West Xingang Road 135, Guangzhou 510275, China.
| | - Lingzheng Xu
- College of Computer Science, Sichuan University, Sichuan 610044, China
| |
Collapse
|
58
|
Herman JP, Nawreen N, Smail MA, Cotella EM. Brain mechanisms of HPA axis regulation: neurocircuitry and feedback in context Richard Kvetnansky lecture. Stress 2020; 23:617-632. [PMID: 33345670 PMCID: PMC8034599 DOI: 10.1080/10253890.2020.1859475] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 11/28/2020] [Indexed: 12/11/2022] Open
Abstract
Regulation of stress reactivity is a fundamental priority of all organisms. Stress responses are critical for survival, yet can also cause physical and psychological damage. This review provides a synopsis of brain mechanisms designed to control physiological responses to stress, focusing primarily on glucocorticoid secretion via the hypothalamo-pituitary-adrenocortical (HPA) axis. The literature provides strong support for multi-faceted control of HPA axis responses, involving both direct and indirect actions at paraventricular nucleus (PVN) corticotropin releasing hormone neurons driving the secretory cascade. The PVN is directly excited by afferents from brainstem and hypothalamic circuits, likely relaying information on homeostatic challenge. Amygdala subnuclei drive HPA axis responses indirectly via disinhibition, mediated by GABAergic relays onto PVN-projecting neurons in the hypothalamus and bed nucleus of the stria terminalis (BST). Inhibition of stressor-evoked HPA axis responses is mediated by an elaborate network of glucocorticoid receptor (GR)-containing circuits, providing a distributed negative feedback signal that inhibits PVN neurons. Prefrontal and hippocampal neurons play a major role in HPA axis inhibition, again mediated by hypothalamic and BST GABAergic relays to the PVN. The complexity of the regulatory process suggests that information on stressors is integrated across functional disparate brain circuits prior to accessing the PVN, with regions such as the BST in prime position to relay contextual information provided by these sources into appropriate HPA activation. Dysregulation of the HPA in disease is likely a product of inappropriate checks and balances between excitatory and inhibitory inputs ultimately impacting PVN output.
Collapse
Affiliation(s)
- James P Herman
- Department of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, OH, USA
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati, Cincinnati, OH, USA
- Cincinnati Veterans Administration Medical Center, Cincinnati, OH, USA
| | - Nawshaba Nawreen
- Department of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, OH, USA
| | - Marissa A Smail
- Department of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, OH, USA
| | - Evelin M Cotella
- Department of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati, OH, USA
- Cincinnati Veterans Administration Medical Center, Cincinnati, OH, USA
| |
Collapse
|
59
|
Perry CJ, Campbell EJ, Drummond KD, Lum JS, Kim JH. Sex differences in the neurochemistry of frontal cortex: Impact of early life stress. J Neurochem 2020; 157:963-981. [PMID: 33025572 DOI: 10.1111/jnc.15208] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 08/02/2020] [Accepted: 09/25/2020] [Indexed: 12/11/2022]
Abstract
Traumatic events during early life have been linked with later life psychopathology. To understand this risk factor, researchers have studied the effects of prenatal and postnatal early life stress on neurochemical changes. Here we review the rodent literature on sex differences and sex-specific impact of early life stress on frontal cortex neurochemistry. This region is implicated in regulating motivation and emotion, which are often disrupted in psychological disorders. The prefrontal cortex (PFC) in particular is one of the last brain regions to develop, and there are sex differences in the rate of this development. To draw direct comparisons between sexes, our review of the literature was restricted to studies where the effects of prenatal or postnatal stress had been described in male and female littermates. This literature included research describing glutamate, γ-amino butyric acid (GABA), corticosteroids, monoamines, and cannabinoids. We found that sex-dependent effects of stress are mediated by the age at which stress is experienced, age at test, and type of stress endured. More research is required, particularly into the effects of adolescent stress on male and female littermates. We hope that a greater understanding of sex-specific susceptibilities in response to stress across development will help to uncover risk factors for psychological disorders in vulnerable populations.
Collapse
Affiliation(s)
- Christina J Perry
- Mental Health Theme, The Florey Institute of Neuroscience and Mental Health, Parkville, Vic, Australia.,Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Vic, Australia
| | - Erin J Campbell
- Mental Health Theme, The Florey Institute of Neuroscience and Mental Health, Parkville, Vic, Australia.,Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Vic, Australia
| | - Katherine D Drummond
- Mental Health Theme, The Florey Institute of Neuroscience and Mental Health, Parkville, Vic, Australia.,Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Vic, Australia
| | - Jeremy S Lum
- Neuropharmacology and Molecular Psychiatry Laboratory, School of Medicine, University of Wollongong, Wollongong, NSW, Australia
| | - Jee Hyun Kim
- Mental Health Theme, The Florey Institute of Neuroscience and Mental Health, Parkville, Vic, Australia.,Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, Vic, Australia.,IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Australia
| |
Collapse
|
60
|
Boorman DC, Brown R, Keay KA. Periaqueductal gray inputs to the paraventricular nucleus of the thalamus: Columnar topography and glucocorticoid (in)sensitivity. Brain Res 2020; 1750:147171. [PMID: 33132167 DOI: 10.1016/j.brainres.2020.147171] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/10/2020] [Accepted: 10/18/2020] [Indexed: 01/01/2023]
Abstract
The ability to cope with a novel acute stressor in the context of ongoing chronic stress is of critical adaptive value. The hypothalamic-pituitary-adrenal (HPA) axis contributes to the integrated physiological and behavioural responses to stressors. Under conditions of chronic stress, the posterior portion of the paraventricular thalamic nucleus (pPVT) mediates the 'habituation' of HPA-axis responses, and also facilitates HPA-axis reactivation to novel acute stressors amidst this habituation. Since pPVT neurons are sensitive to the inhibitory effects of circulating glucocorticoids, a glucocorticoid-insensitive neural pathway to the pPVT is likely essential for this reactivation process. The pPVT receives substantial inputs from neurons of the periaqueductal gray (PAG) region, which is organised into longitudinal columns critical for processing acute and/or chronic stressors. We investigated the columnar organisation of PAG → pPVT projections and for the first time determined their glucocorticoid sensitivity. Retrograde tracer injections were made into different rostro-caudal regions of the pPVT, and their PAG columnar inputs compared. Glucocorticoid receptor immunoreactivity (GR-ir) was quantified in these projection neurons. We found that the dorsolateral PAG projected most strongly to rostral pPVT and the ventrolateral PAG most strongly to the caudal pPVT. Despite abundant GR-ir in the PAG, we report a striking absence of GR-ir in PAG → pPVT neurons. Our data suggests that these pathways, which are insensitive to the direct actions of circulating glucocorticoids, likely play an important role in both the habituation of HPA-axis to chronic stressors and its facilitation to acute stressors in chronically stressed rats.
Collapse
Affiliation(s)
- Damien C Boorman
- School of Medical Sciences (Anatomy & Histology), Faculty of Medicine and Health, University of Sydney, NSW 2006, Australia
| | - Rebecca Brown
- School of Medical Sciences (Anatomy & Histology), Faculty of Medicine and Health, University of Sydney, NSW 2006, Australia
| | - Kevin A Keay
- School of Medical Sciences (Anatomy & Histology), Faculty of Medicine and Health, University of Sydney, NSW 2006, Australia.
| |
Collapse
|
61
|
Moreno-Santos B, Marchi-Coelho C, Costa-Ferreira W, Crestani CC. Angiotensinergic receptors in the medial amygdaloid nucleus differently modulate behavioral responses in the elevated plus-maze and forced swimming test in rats. Behav Brain Res 2020; 397:112947. [PMID: 33011187 DOI: 10.1016/j.bbr.2020.112947] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 09/01/2020] [Accepted: 09/26/2020] [Indexed: 12/16/2022]
Abstract
The brain renin-angiotensin system (RAS) has been implicated in anxiety and depression disorders, but the specific brain sites involved are poorly understood. The medial amygdaloid nucleus (MeA) is involved in expression of behavioral responses. However, despite evidence of the presence of all angiotensinergic receptors in this amygdaloid nucleus, regulation of anxiety- and depressive-like behaviors by angiotensinergic neurotransmissions within the MeA has never been reported. Thus, the present study aimed to investigate the role angiotensin II (AT1 and AT2 receptors) and angiotensin-(1-7) (Mas receptor) receptors present within the MeA in behavioral responses in the elevated plus-maze (EPM) and forced swimming test (FST). For this, male Wistar rats had cannula-guide bilaterally implanted into the MeA, and independent sets of animals received bilateral microinjections of either the selective AT1 receptor antagonist losartan, the selective AT2 receptor antagonist PD123319, the selective Mas receptor antagonist A-779 or vehicle into the MeA before the EPM and FST. Treatment of the MeA with either PD123319 or A-779 decreased the EPM open arms exploration, while losartan did not affect behavioral responses in this apparatus. However, intra-MeA microinjection of losartan decreased immobility in the FST. Administration of either PD123319 or A-779 into the MeA did not affect the immobility during the FST, but changed the pattern of the active behaviors swimming and climbing. Altogether, these results indicate the presence of different angiotensinergic mechanisms within the MeA controlling behavioral responses in the FST and EPM.
Collapse
Affiliation(s)
- Beatriz Moreno-Santos
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, SP, Brazil
| | - Camila Marchi-Coelho
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, SP, Brazil
| | - Willian Costa-Ferreira
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, SP, Brazil; Joint UFSCar-UNESP Graduate Program in Physiological Sciences, São Carlos, SP, Brazil
| | - Carlos C Crestani
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, SP, Brazil; Joint UFSCar-UNESP Graduate Program in Physiological Sciences, São Carlos, SP, Brazil.
| |
Collapse
|
62
|
Predisposition of Neonatal Maternal Separation to Visceral Hypersensitivity via Downregulation of Small-Conductance Calcium-Activated Potassium Channel Subtype 2 (SK2) in Mice. Neural Plast 2020; 2020:8876230. [PMID: 33029124 PMCID: PMC7528131 DOI: 10.1155/2020/8876230] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/31/2020] [Accepted: 09/06/2020] [Indexed: 12/13/2022] Open
Abstract
Background Visceral hypersensitivity is a common occurrence of gastrointestinal diseases such as irritable bowel syndrome (IBS), wherein early-life stress (ELS) may have a high predisposition to the development of visceral hypersensitivity in adulthood, with the specific underlying mechanism still elusive. Herein, we assessed the potential effect of small-conductance calcium-activated potassium channel subtype 2 (SK2) in the spinal dorsal horn (DH) on the pathogenesis of visceral hypersensitivity induced by maternal separation (MS) in mice. Methods Neonatal mice were subjected to the MS paradigm, an established ELS model. In adulthood, the visceral pain threshold and the abdominal withdrawal reflex (AWR) were measured with an inflatable balloon. The elevated plus maze, open field test, sucrose preference test, and forced swim test were employed to evaluate the anxiety- and depression-like behaviors. The expression levels of SK2 in the spinal DH were determined by immunofluorescence and western blotting. The mRNA of SK2 and membrane palmitoylated protein 2 (MPP2) were determined by quantitative real-time polymerase chain reaction (qRT-PCR). Electrophysiology was applied to evaluate the neuronal firing rates and SK2 channel-mediated afterhyperpolarization current (I AHP). The interaction between MPP2 and SK2 was validated by coimmunoprecipitation. Results In contrast to the naïve mice, ethological findings in MS mice revealed lowered visceral pain threshold, more evident anxiety- and depression-like behaviors, and downregulated expression of membrane SK2 protein and MPP2 protein. Moreover, electrophysiological results indicated increased neuronal firing rates and decreased I AHP in the spinal DH neurons. Nonetheless, intrathecal injection of the SK2 channel activator 1-ethyl-2-benzimidazolinone (1-EBIO) in MS mice could reverse the electrophysiological alterations and elevate the visceral pain threshold. In the naïve mice, administration of the SK2 channel blocker apamin abated I AHP and elevated spontaneous neuronal firing rates in the spinal DH neurons, reducing the visceral pain threshold. Finally, disruption of the MPP2 expression by small interfering RNA (siRNA) could amplify visceral hypersensitivity in naïve mice. Conclusions ELS-induced visceral pain and visceral hypersensitivity are associated with the underfunction of SK2 channels in the spinal DH.
Collapse
|
63
|
Meknatkhah S, Dashti PS, Raminfard S, Rad HS, Mousavi MS, Riazi GH. The Changes in 1H-MRS Metabolites in Cuprizone-Induced Model of Multiple Sclerosis: Effects of Prior Psychological Stress. J Mol Neurosci 2020; 71:804-809. [PMID: 32915417 DOI: 10.1007/s12031-020-01702-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 09/07/2020] [Indexed: 11/25/2022]
Abstract
Stress is considered as an important risk factor in the progression and the onset of many disorders such as multiple sclerosis. However, metabolite changes as a result of demyelination under the detrimental effects of stress are not well understood. Thus, 36 female Wistar rats (i.e., groups (1) no-cuprizone (Cont), (2) no-stress + cuprizone-treated (Cup), (3) physical stress + cuprizone-treated (P-Cup), (4) psychological stress + cuprizone-treated (Psy-Cup), (5) physical stress + no-cuprizone-treated (P), (6) psychological stress + no-cuprizone-treated (Psy)) were used in this study. Following induction of repetitive stress, cuprizone treatment was carried out for 6 weeks to instigate demyelination in all groups except the control animal. Relative metabolite concentrations of the brain were investigated by single-voxel proton magnetic resonance spectroscopy (reporting N-acetyl-aspartate (NAA), glycerophosphocholine with phosphocholine (tCho) relative to total creatine (tCr)). According to 1H-MRS, rats in the Cup group indicated a reduction in NAA/ tCr (p < 0.001) as well as tCho/ tCr (p < 0.05) compared with that in the Cont group. In contrast, in both stress + cuprizone-treated groups, NAA/tCr and tCho/tCr ratios remarkably increased versus the Cup group (p < 0.001) and the Cont group (p < 0.001 for the Psy-Cup group and p < 0.05 for the P-Cup group). Both P and Psy groups revealed normal metabolite concentrations similar to the Cont group 6 weeks post stress. Seemingly, in the case of cuprizone alone, decreased level of metabolites is mainly relevant to neuronal cell impairments. Meanwhile, as a result of oxidative stress enhancement due to stress exposure, oligodendrocyte becomes the main victim indicating the increased level of metabolite ratios.
Collapse
Affiliation(s)
- Sogol Meknatkhah
- Laboratory of Neuro-Organic Chemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Pouya Sharif Dashti
- College of Engineering, Faculty of Chemical Engineering, University of Tehran, Tehran, Iran
| | - Samira Raminfard
- Department of Neuroscience, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamidreza Saligheh Rad
- Quantitative Medical Imaging Systems Group (QMISG) Research Center for Molecular and Cellular Imaging, Tehran University of Medical Sciences, Tehran, Iran
- Department of Medical Physics and Biomedical Engineering, Tehran University of Medical Sciences, Tehran, Iran
| | - Monireh-Sadat Mousavi
- Laboratory of Neuro-Organic Chemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Gholam Hossein Riazi
- Laboratory of Neuro-Organic Chemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran.
| |
Collapse
|
64
|
Sher LD, Geddie H, Olivier L, Cairns M, Truter N, Beselaar L, Essop MF. Chronic stress and endothelial dysfunction: mechanisms, experimental challenges, and the way ahead. Am J Physiol Heart Circ Physiol 2020; 319:H488-H506. [PMID: 32618516 DOI: 10.1152/ajpheart.00244.2020] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Although chronic stress is an important risk factor for cardiovascular diseases (CVD) onset, the underlying mechanisms driving such pathophysiological complications remain relatively unknown. Here, dysregulation of innate stress response systems and the effects of downstream mediators are strongly implicated, with the vascular endothelium emerging as a primary target of excessive glucocorticoid and catecholamine action. Therefore, this review article explores the development of stress-related endothelial dysfunction by focusing on the following: 1) assessing the phenomenon of stress and complexities surrounding this notion, 2) discussing mechanistic links between chronic stress and endothelial dysfunction, and 3) evaluating the utility of various preclinical models currently employed to study mechanisms underlying the onset of stress-mediated complications such as endothelial dysfunction. The data reveal that preclinical models play an important role in our efforts to gain an increased understanding of mechanisms underlying stress-mediated endothelial dysfunction. It is our understanding that this provides a good foundation going forward, and we propose that further efforts should be made to 1) more clearly define the concept of stress and 2) standardize protocols of animal models with specific guidelines to better indicate the mental complications that are simulated.
Collapse
Affiliation(s)
- Lucien Derek Sher
- Centre for Cardio-Metabolic Research in Africa, Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Hannah Geddie
- Centre for Cardio-Metabolic Research in Africa, Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Lukas Olivier
- Centre for Cardio-Metabolic Research in Africa, Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Megan Cairns
- Centre for Cardio-Metabolic Research in Africa, Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Nina Truter
- Centre for Cardio-Metabolic Research in Africa, Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Leandrie Beselaar
- Centre for Cardio-Metabolic Research in Africa, Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - M Faadiel Essop
- Centre for Cardio-Metabolic Research in Africa, Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
65
|
Zhang F, Chen H, Zhang R, Liu Y, Kong N, Guo Y, Xu M. 5-Fluorouracil induced dysregulation of the microbiome-gut-brain axis manifesting as depressive like behaviors in rats. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165884. [PMID: 32574836 DOI: 10.1016/j.bbadis.2020.165884] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 04/08/2020] [Accepted: 06/17/2020] [Indexed: 02/08/2023]
Abstract
Disturbances of the gut microbiome have been widely suggested to be associated with 5-fluorouracil (5-Fu) induced digestive pathologies. Furthermore, it has been elucidated that the gut microbiome may play a key role in the pathogenesis of depressive disorders via the microbiota-gut-brain axis. Despite the speculation, there exists no direct evidence proving the causality between disturbances in the gut microbiome induced by 5-Fu and depressive mood dysregulation. Herein, behavioral testing was used to evaluate depressive-like behaviors in 5-Fu treated rats. Subsequently, the gut microbiota and prefrontal cortex (PFC) metabolic were analyzed by 16S rRNA sequencing and 1H nuclear magnetic resonance (1H NMR). To clarify the association between the gut microbiota and their role on depressive-like behaviors caused by 5-Fu, a fecal microbiota transplantation (FMT) experiment was carried out. The results suggested that 5-Fu could significantly alter the diversity and abundance of the gut microbiome, and induce PFC metabolic disorders, as well as depressive behaviors in rats. Transplantation of fecal microbiota from healthy control into 5-Fu treated rats significantly alleviated the PFC metabolic disorder and depressive-like behaviors. In conclusion, this study demonstrated that the gut microbiome was actively involved in the occurrence of 5-Fu induced depressive-like behaviors, and manipulation of specific gut microbiome parameters may serve as a promising novel target for side effects of 5-Fu treatment.
Collapse
Affiliation(s)
- Fan Zhang
- The First Clinical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China; Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310003, China
| | - Haitao Chen
- The First Clinical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Ruixin Zhang
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310003, China
| | - Yu Liu
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310000, China
| | - Ning Kong
- The First Clinical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Yong Guo
- Department of Oncology, First Affiliated Hospital of Zhejiang Traditional Medical University, Hangzhou, Zhejiang 310003, China.
| | - Maosheng Xu
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310003, China.
| |
Collapse
|
66
|
A limbic circuitry involved in emotional stress-induced grooming. Nat Commun 2020; 11:2261. [PMID: 32385304 PMCID: PMC7210270 DOI: 10.1038/s41467-020-16203-x] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 04/16/2020] [Indexed: 01/08/2023] Open
Abstract
Prolonged exposure to negative stressors could be harmful if a subject cannot respond appropriately. Strategies evolved to respond to stress, including repetitive displacement behaviours, are important in maintaining behavioural homoeostasis. In rodents, self-grooming is a frequently observed repetitive behaviour believed to contribute to post-stress de-arousal with adaptive value. Here we identified a rat limbic di-synaptic circuit that regulates stress-induced self-grooming with positive affective valence. This circuit links hippocampal ventral subiculum to ventral lateral septum (LSv) and then lateral hypothalamus tuberal nucleus. Optogenetic activation of this circuit triggers delayed but robust excessive grooming with patterns closely resembling those evoked by emotional stress. Consistently, the neural activity of LSv reaches a peak before emotional stress-induced grooming while inhibition of this circuit significantly suppresses grooming triggered by emotional stress. Our results uncover a previously unknown limbic circuitry involved in regulating stress-induced self-grooming and pinpoint a critical role of LSv in this ethologically important behaviour. Self-grooming is a frequently observed repetitive behaviour in rodents that is believed to contribute to post-stress de-arousal. The authors identified a previously unknown limbic circuit that includes the ventral lateral septum in rats and is involved in regulating stress-induced self-grooming.
Collapse
|
67
|
Matuska R, Zelena D, Könczöl K, Papp RS, Durst M, Guba D, Török B, Varnai P, Tóth ZE. Colocalized neurotransmitters in the hindbrain cooperate in adaptation to chronic hypernatremia. Brain Struct Funct 2020; 225:969-984. [PMID: 32200401 PMCID: PMC7166202 DOI: 10.1007/s00429-020-02049-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 02/13/2020] [Indexed: 11/29/2022]
Abstract
Chronic hypernatremia activates the central osmoregulatory mechanisms and inhibits the function of the hypothalamic-pituitary-adrenal (HPA) axis. Noradrenaline (NE) release into the periventricular anteroventral third ventricle region (AV3V), the supraoptic (SON) and hypothalamic paraventricular nuclei (PVN) from efferents of the caudal ventrolateral (cVLM) and dorsomedial (cDMM) medulla has been shown to be essential for the hypernatremia-evoked responses and for the HPA response to acute restraint. Notably, the medullary NE cell groups highly coexpress prolactin-releasing peptide (PrRP) and nesfatin-1/NUCB2 (nesfatin), therefore, we assumed they contributed to the reactions to chronic hypernatremia. To investigate this, we compared two models: homozygous Brattleboro rats with hereditary diabetes insipidus (DI) and Wistar rats subjected to chronic high salt solution (HS) intake. HS rats had higher plasma osmolality than DI rats. PrRP and nesfatin mRNA levels were higher in both models, in both medullary regions compared to controls. Elevated basal tyrosine hydroxylase (TH) expression and impaired restraint-induced TH, PrRP and nesfatin expression elevations in the cVLM were, however, detected only in HS, but not in DI rats. Simultaneously, only HS rats exhibited classical signs of chronic stress and severely blunted hormonal reactions to acute restraint. Data suggest that HPA axis responsiveness to restraint depends on the type of hypernatremia, and on NE capacity in the cVLM. Additionally, NE and PrRP signalization primarily of medullary origin is increased in the SON, PVN and AV3V in HS rats. This suggests a cooperative action in the adaptation responses and designates the AV3V as a new site for PrRP's action in hypernatremia.
Collapse
Affiliation(s)
- Rita Matuska
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Dóra Zelena
- Behavioral Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
- Centre for Neuroscience, Szentágothai Research Centre, Institute of Physiology, Medical School, University of Pécs, Pécs, Hungary
| | - Katalin Könczöl
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Rege Sugárka Papp
- Human Brain Tissue Bank and Microdissection Laboratory, Semmelweis University, Budapest, Hungary
| | - Máté Durst
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Dorina Guba
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Bibiana Török
- Behavioral Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
- Janos Szentagothai School of Neurosciences, Semmelweis University, Budapest, Hungary
| | - Peter Varnai
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Zsuzsanna E Tóth
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
68
|
Varlinskaya EI, Johnson JM, Przybysz KR, Deak T, Diaz MR. Adolescent forced swim stress increases social anxiety-like behaviors and alters kappa opioid receptor function in the basolateral amygdala of male rats. Prog Neuropsychopharmacol Biol Psychiatry 2020; 98:109812. [PMID: 31707090 PMCID: PMC6920550 DOI: 10.1016/j.pnpbp.2019.109812] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 11/05/2019] [Accepted: 11/06/2019] [Indexed: 12/15/2022]
Abstract
Adolescence is a developmental period marked by robust neural alterations and heightened vulnerability to stress, a factor that is highly associated with increased risk for emotional processing deficits, such as anxiety. Stress-induced upregulation of the dynorphin/kappa opioid receptor (DYN/KOP) system is thought to, in part, underlie the negative affect associated with stress. The basolateral amygdala (BLA) is a key structure involved in anxiety, and neuromodulatory systems, such as the DYN/KOP system, can 1) regulate BLA neural activity in an age-dependent manner in stress-naïve animals and 2) underlie stress-induced anxiety in adults. However, the role of the DYN/KOP system in modulating stress-induced anxiety in adolescents is unknown. To test this, we examined the impact of an acute, 2-day forced swim stress (FSS - 10 min each day) on adolescent (~postnatal day (P) 35) and adult Sprague-Dawley rats (~P70), followed by behavioral, molecular and electrophysiological assessment 24 h following FSS. Adolescent males, but not adult males or females of either age, demonstrated social anxiety-like behavioral alterations indexed via significantly reduced social investigation and preference when tested 24 h following FSS. Conversely, adult males exhibited increased social preference. While there were no FSS-induced changes in expression of genes related to the DYN/KOP system in the BLA, these behavioral alterations were associated with alterations in BLA KOP function. Specifically, while GABA transmission in BLA pyramidal neurons from non-stressed adolescent males responded variably (potentiated, suppressed, or was unchanged) to the KOP agonist, U69593, U69593 significantly inhibited BLA GABA transmission in the majority of neurons from stressed adolescent males, consistent with the observed anxiogenic phenotype in stressed adolescent males. This is the first study to demonstrate stress-induced alterations in BLA KOP function that may contribute to stress-induced social anxiety in adolescent males. Importantly, these findings provide evidence for potential KOP-dependent mechanisms that may contribute to pathophysiological interactions with subsequent stress challenges.
Collapse
Affiliation(s)
- E I Varlinskaya
- Department of Psychology, Center for Development and Behavioral Neuroscience, Developmental Exposure Alcohol Research Center, Binghamton University, Binghamton, NY 13902, United States
| | - J M Johnson
- Department of Psychology, Center for Development and Behavioral Neuroscience, Developmental Exposure Alcohol Research Center, Binghamton University, Binghamton, NY 13902, United States
| | - K R Przybysz
- Department of Psychology, Center for Development and Behavioral Neuroscience, Developmental Exposure Alcohol Research Center, Binghamton University, Binghamton, NY 13902, United States
| | - T Deak
- Department of Psychology, Center for Development and Behavioral Neuroscience, Developmental Exposure Alcohol Research Center, Binghamton University, Binghamton, NY 13902, United States
| | - M R Diaz
- Department of Psychology, Center for Development and Behavioral Neuroscience, Developmental Exposure Alcohol Research Center, Binghamton University, Binghamton, NY 13902, United States.
| |
Collapse
|
69
|
Mirogabalin prevents repeated restraint stress-induced dysfunction in mice. Behav Brain Res 2020; 383:112506. [PMID: 31982462 DOI: 10.1016/j.bbr.2020.112506] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 01/15/2020] [Accepted: 01/23/2020] [Indexed: 12/18/2022]
Abstract
Gabapentinoids, which are the common analgesics, are also thought to be an effective treatment for anxiety disorder, which is one of several psychiatric disorders triggered and exacerbated by stress. The aim of the present study was to investigate whether mirogabalin, a recently launched gabapentinoid, protects multiple brain functions against repeated restraint stress. Adult male ddY mice were restrained for 7 days (repeated restraint stress: 2 h/day) or for 30 min (single restraint stress). Mirogabalin (intraperitoneal, intracerebroventricular or intrahippocampal injection) was administered prior to the restraint stress. Y-maze, elevated-plus maze and c-Fos immunohistochemistry were performed to evaluate learning function, anxiety levels and hippocampal neuronal activities, respectively, after the 7th day of the repeated restraint stress. Intestinal function was evaluated in terms of defecation, which was scored after the 5th day of repeated restraint stress and by the number of fecal pellets excreted after a single session of restraint stress. Repeated restraint stress induced memory dysfunction, anxiety-like behavior, an abnormal defecation score and increased hippocampal c-Fos expression. These changes were prevented by systemic administration of mirogabalin. Abnormal defecation was also induced by single restraint stress, and was inhibited by both systemic and central administration of mirogabalin, suggesting that the effect on the intestinal function was also mediated via the central nervous system. Enhancement of c-Fos expression by repeated stress was decreased by intrahippocampal injection of mirogabalin. Together, these observations suggest that mirogabalin protects multiple brain functions from repeated stress, which may be mediated by inhibition of hippocampal neuron hyperactivation.
Collapse
|
70
|
Ebertowska A, Ludkiewicz B, Melka N, Klejbor I, Moryś J. The influence of early postnatal chronic mild stress stimulation on the activation of amygdala in adult rat. J Chem Neuroanat 2020; 104:101743. [PMID: 31926296 DOI: 10.1016/j.jchemneu.2020.101743] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 12/30/2019] [Accepted: 01/07/2020] [Indexed: 11/18/2022]
Abstract
Amygdala is a limbic structure involved in the stress response. The immunohistochemical and morphometric methods were used to examine whether the chronic mild psychological stress during the early postnatal period would change activation of amygdaloid nuclei in response to the same stressor in adult. In the study we focused on the role of neurons containing calbindin (CB), calretinin (CR), parvalbumin (PV) and nitric oxide synthase (NOS). The rats were divided into three groups: control non-stressed animals and two experimental: EI consisted of animals that were exposed to acute stress in the high-light, open-field test (HL-OF) at P90 (P - postnatal day) and EII consisted of rats that were exposed to chronic stress in HL-OF, daily during the first 21 postnatal days and then once at P90. The scheme of activation of amygdaloid nuclei under stress in EI and EII group was similar. The highest density of c-Fos-ir cells (c-Fos - a marker of neuronal activation) was demonstrated by the medial nucleus (Me) and bed nucleus of the accessory olfactory tract (BAOT). The amygdaloid nuclei diversity after HL-OF was determined by the high activation of the NOS-ir cells in the Me and NOS- and CR-ir cells in the BAOT. These are probably projection neurons involved in modulation of defensive, reproductive and autonomic behavior in stress response and creation/storage of aversive memory. However, in comparison with EI group, significant decrease in density of c-Fos-ir cells, in almost all amygdaloid nuclei of EII group was revealed. Particularly in BAOT and Me the strong decrease of activity of NOS- and CR-ir neurons was observed. It probably results in attenuation of stress responses what, depending on the circumstances, can be adaptive or maladaptive.
Collapse
Affiliation(s)
- A Ebertowska
- Department of Anatomy and Neurobiology Medical University of Gdańsk, Poland.
| | - B Ludkiewicz
- Department of Anatomy and Neurobiology Medical University of Gdańsk, Poland
| | - N Melka
- Department of Anatomy and Neurobiology Medical University of Gdańsk, Poland
| | - I Klejbor
- Department of Anatomy and Neurobiology Medical University of Gdańsk, Poland
| | - J Moryś
- Department of Anatomy and Neurobiology Medical University of Gdańsk, Poland
| |
Collapse
|
71
|
Nariya M, Joshi D, Ranpariya N. Adaptogenic and immunomodulatory activity of Virgozest Avaleha – An ayurvedic proprietary formulation. Ayu 2020; 41:255-261. [PMID: 35813363 PMCID: PMC9261994 DOI: 10.4103/ayu.ayu_57_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 12/06/2021] [Accepted: 03/11/2022] [Indexed: 11/09/2022] Open
Abstract
Introduction: Rasayana (rejuvenator) or adaptogenic drugs have been proved to produce the complete potential to prevent diseases and degenerative changes that leads to diseases and promote longevity by providing strength and immunity. Virgozest Avaleha is a poly-herbal formulation claimed to serve as adaptogenic, and immunomodulatory, as a health tonic, enriched with dry fruits, and ingredients containing natural supplements of Vitamin E and proteins. Aim: To evaluate the adaptogenic activity and humoral immune activity of virgozest Avaleha in Wistar albino rats. Materials and methods: Virgozest Avaleha was evaluated for adaptogenic activity against swimming stress-induced changes and hypothermia in albino rats. The humoral immune activity of virgozest Avaleha was evaluated against sheep red blood cells (SRBCs)-induced response in albino rats with the inclusion of cyclophosphamide as immune suppressant agent. Results: In adaptogenic activity, virgozest Avaleha (450 and 900 mg/kg) exhibited an increase in physical activity, decrease in stress-induced hypothermia, and serum cortisol level when compared to the stress control group of albino rats. In humoral immune activity, virgozest Avaleha reversed the effects of cyclophosphamide-induced adverse changes on spleen and lymph node, and produced a significant increase in serum antibody titer in SRBCs-sensitized rats. Conclusion: The present study concluded that virgozest Avaleha has adaptogenic and humoral immune activity in Wistar albino rats, which may suggest the Rasayana like properties of Ayurvedic formulation.
Collapse
|
72
|
Kinlein SA, Karatsoreos IN. The hypothalamic-pituitary-adrenal axis as a substrate for stress resilience: Interactions with the circadian clock. Front Neuroendocrinol 2020; 56:100819. [PMID: 31863788 PMCID: PMC7643247 DOI: 10.1016/j.yfrne.2019.100819] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 10/29/2019] [Accepted: 12/17/2019] [Indexed: 12/22/2022]
Abstract
Stress, primarily processed via the hypothalamic-pituitary-adrenal (HPA) axis, engages biological pathways throughout the brain and body which promote adaptation and survival to changing environmental demands. Adaptation to environmental challenges is compromised when these pathways are no longer functioning optimally. The physiological and behavioral mechanisms through which HPA axis function influences stress adaptation and resilience are not fully elucidated. Our understanding of stress biology and disease must take into account the complex interactions between the endocrine system, neural circuits, and behavioral coping strategies. In addition, further consideration must be taken concerning influences of other aspects of physiology, including the circadian clock which is critical for regulation of daily changes in HPA activity. While adding a layer of complexity, it also offers targets for intervention. Understanding the role of HPA function in mediating these diverse biological responses will lead to important insights about how to bolster successful stress adaptation and promote stress resilience.
Collapse
Affiliation(s)
- Scott A Kinlein
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, United States
| | - Ilia N Karatsoreos
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA 99164, United States; Department of Psychological and Brain Sciences, University of Massachusetts, Amherst, MA 01003, United States.
| |
Collapse
|
73
|
Reinebrant HE, Wixey JA, Buller KM. Hypoxia-ischemia in the immature rodent brain impairs serotonergic neuronal function in certain dorsal raphé nuclei. Neural Regen Res 2020; 15:457-463. [PMID: 31571657 PMCID: PMC6921336 DOI: 10.4103/1673-5374.266067] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Neonatal hypoxia-ischemia (HI) results in losses of serotonergic neurons in specific dorsal raphé nuclei. However, not all serotonergic raphé neurons are lost and it is therefore important to assess the function of remaining neurons in order to understand their potential to contribute to neurological disorders in the HI-affected neonate. The main objective of this study was to determine how serotonergic neurons, remaining in the dorsal raphé nuclei after neonatal HI, respond to an external stimulus (restraint stress). On postnatal day 3 (P3), male rat pups were randomly allocated to one of the following groups: (i) control + no restraint (n = 5), (ii) control + restraint (n = 6), (iii) P3 HI + no restraint (n = 5) or (iv) P3 HI + restraint (n = 7). In the two HI groups, rat pups underwent surgery to ligate the common carotid artery and were then exposed to 6% O2 for 30 minutes. Six weeks after P3 HI, on P45, rats were subjected to restraint stress for 30 minutes. Using dual immunolabeling for Fos protein, a marker for neuronal activity, and serotonin (5-hydroxytrypamine; 5-HT), numbers of Fos-positive 5-HT neurons were determined in five dorsal raphé nuclei. We found that restraint stress alone increased numbers of Fos-positive 5-HT neurons in all five dorsal raphé nuclei compared to control animals. However, following P3 HI, the number of stress-induced Fos-positive 5-HT neurons was decreased significantly in the dorsal raphé ventrolateral, interfascicular and ventral nuclei compared with control animals exposed to restraint stress. In contrast, numbers of stress-induced Fos-positive 5-HT neurons in the dorsal raphé dorsal and caudal nuclei were not affected by P3 HI. These data indicate that not only are dorsal raphé serotonergic neurons lost after neonatal HI, but also remaining dorsal raphé serotonergic neurons have reduced differential functional viability in response to an external stimulus. Procedures were approved by the University of Queensland Animal Ethics Committee (UQCCR958/08/NHMRC) on February 27, 2009.
Collapse
Affiliation(s)
- Hanna E Reinebrant
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, Queensland, Australia
| | - Julie A Wixey
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, Queensland, Australia
| | - Kathryn M Buller
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, Queensland, Australia
| |
Collapse
|
74
|
Sharma VK, Singh TG. Chronic Stress and Diabetes Mellitus: Interwoven Pathologies. Curr Diabetes Rev 2020; 16:546-556. [PMID: 31713487 DOI: 10.2174/1573399815666191111152248] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 09/25/2019] [Accepted: 10/27/2019] [Indexed: 12/15/2022]
Abstract
Stress threatens the homeostasis and mobilizes a plethora of adaptive physiological and behavioral changes via the hypothalamic-pituitary-adrenal (HPA) axis and the sympathetic nervous system. The HPA axis influences the pituitary gland, hypothalamus and adrenal gland via a complex set of positive and negative feedback system. The feedback system operates in a well regulated neuroendocrine manner to reestablish the threatened body equilibrium. The HPA axis secreted major product is a glucocorticoid (cortisol) which is kept within a physiologically optimal range and serves to accomplish the various physiological functions crucial for survival. In chronically stressed individuals dishabituation of HPA axis is followed by increased release of glucocorticoids and catecholamines. Higher secretion of glucocorticoids influences glucose metabolism by promoting gluconeogenesis in the liver, suppressing glucose uptake (adipocytes and skeletal muscles), promoting lipolysis in adipocytes, suppressing insulin secretion, inflicting insulin resistance and inflammation. These biological changes alter neuroendocrine mechanisms and lead to maladaptive congregation of events that form the underlying cause of development of Type 2 diabetes (T2D). The currently reviewed evidences advocate that targeting stress mediated hypersecretion of glucocorticoids may be a viable approach for the treatment of T2D and to reinstate glucose homeostasis.
Collapse
Affiliation(s)
- Vivek Kumar Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
- Department of Pharmacology, Government College of Pharmacy, Rohru, Distt. Shimla-171207, Himachal Pradesh, India
| | | |
Collapse
|
75
|
Thorsdottir D, Cruickshank NC, Einwag Z, Hennig GW, Erdos B. BDNF downregulates β-adrenergic receptor-mediated hypotensive mechanisms in the paraventricular nucleus of the hypothalamus. Am J Physiol Heart Circ Physiol 2019; 317:H1258-H1271. [PMID: 31603352 DOI: 10.1152/ajpheart.00478.2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) is upregulated in the paraventricular nucleus of the hypothalamus (PVN) in response to hypertensive stimuli such as stress and hyperosmolality, and BDNF acting in the PVN plays a key role in elevating sympathetic activity and blood pressure. However, downstream mechanisms mediating these effects remain unclear. We tested the hypothesis that BDNF increases blood pressure, in part by diminishing inhibitory hypotensive input from nucleus of the solitary tract (NTS) catecholaminergic neurons projecting to the PVN. Male Sprague-Dawley rats received bilateral PVN injections of viral vectors expressing either green fluorescent protein (GFP) or BDNF and bilateral NTS injections of vehicle or anti-dopamine-β-hydroxylase-conjugated saporin (DSAP), a neurotoxin that selectively lesions noradrenergic and adrenergic neurons. BDNF overexpression in the PVN without NTS lesioning significantly increased mean arterial pressure (MAP) in awake animals by 18.7 ± 1.8 mmHg. DSAP treatment also increased MAP in the GFP group, by 9.8 ± 3.2 mmHg, but failed to affect MAP in the BDNF group, indicating a BDNF-induced loss of NTS catecholaminergic hypotensive effects. In addition, in α-chloralose-urethane-anesthetized rats, hypotensive responses to PVN injections of the β-adrenergic agonist isoprenaline were significantly attenuated by BDNF overexpression, whereas PVN injections of phenylephrine had no effect on blood pressure. BDNF treatment was also found to significantly reduce β1-adrenergic receptor mRNA expression in the PVN, whereas expression of other adrenergic receptors was unaffected. In summary, increased BDNF expression in the PVN elevates blood pressure, in part by downregulating β-receptor signaling and diminishing hypotensive catecholaminergic input from the NTS to the PVN.NEW & NOTEWORTHY We have shown that BDNF, a key hypothalamic regulator of blood pressure, disrupts catecholaminergic signaling between the NTS and the PVN by reducing the responsiveness of PVN neurons to inhibitory hypotensive β-adrenergic input from the NTS. This may be occurring partly via BDNF-mediated downregulation of β1-adrenergic receptor expression in the PVN and results in an increase in blood pressure.
Collapse
Affiliation(s)
| | | | - Zachary Einwag
- Department of Pharmacology, University of Vermont, Burlington, Vermont
| | - Grant W Hennig
- Department of Pharmacology, University of Vermont, Burlington, Vermont
| | - Benedek Erdos
- Department of Pharmacology, University of Vermont, Burlington, Vermont
| |
Collapse
|
76
|
Noble DJ, Hochman S. Hypothesis: Pulmonary Afferent Activity Patterns During Slow, Deep Breathing Contribute to the Neural Induction of Physiological Relaxation. Front Physiol 2019; 10:1176. [PMID: 31572221 PMCID: PMC6753868 DOI: 10.3389/fphys.2019.01176] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 08/30/2019] [Indexed: 12/26/2022] Open
Abstract
Control of respiration provides a powerful voluntary portal to entrain and modulate central autonomic networks. Slowing and deepening breathing as a relaxation technique has shown promise in a variety of cardiorespiratory and stress-related disorders, but few studies have investigated the physiological mechanisms conferring its benefits. Recent evidence suggests that breathing at a frequency near 0.1 Hz (6 breaths per minute) promotes behavioral relaxation and baroreflex resonance effects that maximize heart rate variability. Breathing around this frequency appears to elicit resonant and coherent features in neuro-mechanical interactions that optimize physiological function. Here we explore the neurophysiology of slow, deep breathing and propose that coincident features of respiratory and baroreceptor afferent activity cycling at 0.1 Hz entrain central autonomic networks. An important role is assigned to the preferential recruitment of slowly-adapting pulmonary afferents (SARs) during prolonged inhalations. These afferents project to discrete areas in the brainstem within the nucleus of the solitary tract (NTS) and initiate inhibitory actions on downstream targets. Conversely, deep exhalations terminate SAR activity and activate arterial baroreceptors via increases in blood pressure to stimulate, through NTS projections, parasympathetic outflow to the heart. Reciprocal SAR and baroreceptor afferent-evoked actions combine to enhance sympathetic activity during inhalation and parasympathetic activity during exhalation, respectively. This leads to pronounced heart rate variability in phase with the respiratory cycle (respiratory sinus arrhythmia) and improved ventilation-perfusion matching. NTS relay neurons project extensively to areas of the central autonomic network to encode important features of the breathing pattern that may modulate anxiety, arousal, and attention. In our model, pronounced respiratory rhythms during slow, deep breathing also support expression of slow cortical rhythms to induce a functional state of alert relaxation, and, via nasal respiration-based actions on olfactory signaling, recruit hippocampal pathways to boost memory consolidation. Collectively, we assert that the neurophysiological processes recruited during slow, deep breathing enhance the cognitive and behavioral therapeutic outcomes obtained through various mind-body practices. Future studies are required to better understand the physio-behavioral processes involved, including in animal models that control for confounding factors such as expectancy biases.
Collapse
Affiliation(s)
- Donald J. Noble
- Department of Physiology, Emory University School of Medicine, Atlanta, GA, United States
| | | |
Collapse
|
77
|
Varodayan FP, Minnig MA, Steinman MQ, Oleata CS, Riley MW, Sabino V, Roberto M. PACAP regulation of central amygdala GABAergic synapses is altered by restraint stress. Neuropharmacology 2019; 168:107752. [PMID: 31476352 DOI: 10.1016/j.neuropharm.2019.107752] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 08/27/2019] [Accepted: 08/29/2019] [Indexed: 12/25/2022]
Abstract
The pituitary adenylate cyclase-activating polypeptide (PACAP) system plays a central role in the brain's emotional response to psychological stress by activating cellular processes and circuits associated with threat exposure. The neuropeptide PACAP and its main receptor PAC1 are expressed in the rodent central amygdala (CeA), a brain region critical in negative emotional processing, and CeA PACAPergic signaling drives anxiogenic and stress coping behaviors. Despite this behavioral evidence, PACAP's effects on neuronal activity within the medial subdivision of the CeA (CeM, the major output nucleus for the entire amygdala complex) during basal conditions and after psychological stress remain unknown. Therefore, in the present study, male Wistar rats were subjected to either restraint stress or control conditions, and PACAPergic regulation of CeM cellular function was assessed using immunohistochemistry and whole-cell patch-clamp electrophysiology. Our results demonstrate that PACAP-38 potentiates GABA release in the CeM of naïve rats, via its actions at presynaptic PAC1. Basal PAC1 activity also enhances GABA release in an action potential-dependent manner. Notably, PACAP-38's facilitation of CeM GABA release was attenuated after a single restraint stress session, but after repeated sessions returned to the level observed in naïve animals. A single restraint session also significantly decreased PAC1 levels in the CeM, with repeated restraint sessions producing a slight recovery. Collectively our data reveal that PACAP/PAC1 signaling enhances inhibitory control of the CeM and that psychological stress can modulate this influence to potentially disinhibit downstream effector regions that mediate anxiety and stress-related behaviors. This article is part of the special issue on 'Neuropeptides'.
Collapse
Affiliation(s)
- F P Varodayan
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA.
| | - M A Minnig
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University, School of Medicine, Boston, MA, 02118, USA
| | - M Q Steinman
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - C S Oleata
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - M W Riley
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University, School of Medicine, Boston, MA, 02118, USA
| | - V Sabino
- Laboratory of Addictive Disorders, Departments of Pharmacology and Psychiatry, Boston University, School of Medicine, Boston, MA, 02118, USA
| | - M Roberto
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| |
Collapse
|
78
|
Abstract
The innate immune system plays a critical role in the ethanol-induced neuroimmune response in the brain. Ethanol initiates the innate immune response via activation of the innate immune receptors Toll-like receptors (TLRs, e.g., TLR4, TLR3, TLR7) and NOD-like receptors (inflammasome NLRs) leading to a release of a plethora of chemokines and cytokines and development of the innate immune response. Cytokines and chemokines can have pro- or anti-inflammatory properties through which they regulate the immune response. In this chapter, we will focus on key cytokines (e.g., IL-1, IL-6, TNF-α) and chemokines (e.g., MCP-1/CCL2) that mediate the ethanol-induced neuroimmune responses. In this regard, we will use IL-1β, as an example cytokine, to discuss the neuromodulatory properties of cytokines on cellular properties and synaptic transmission. We will discuss their involvement through a set of evidence: (1) changes in gene and protein expression following ethanol exposure, (2) association of gene polymorphisms (humans) and alterations in gene expression (animal models) with increased alcohol intake, and (3) modulation of alcohol-related behaviors by transgenic or pharmacological manipulations of chemokine and cytokine systems. Over the last years, our understanding of the molecular mechanisms mediating cytokine- and chemokine-dependent regulation of immune responses has advanced tremendously, and we review evidence pointing to cytokines and chemokines serving as neuromodulators and regulators of neurotransmission.
Collapse
Affiliation(s)
- Marisa Roberto
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA.
| | - Reesha R Patel
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| | - Michal Bajo
- Department of Neuroscience, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
79
|
Buharin VE, Shinohara M. Corticospinal excitability for flexor carpi radialis decreases with baroreceptor unloading during intentional co-contraction with opposing forearm muscles. Exp Brain Res 2019; 237:1947-1958. [PMID: 31129694 DOI: 10.1007/s00221-019-05563-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 05/17/2019] [Indexed: 11/29/2022]
Abstract
Concurrent activation of antagonistic muscles (co-contraction) is used for stiffening a joint, whereas its neural control under hemodynamic stress (e.g., posture change, high gravity, and hemorrhage) is unknown. Corticospinal excitability during co-contraction may be altered with baroreceptor unloading due to potential modulations in spinal and/or inhibitory pathways (e.g., disynaptic group I inhibition and GABA-mediated intracortical inhibition). The purpose of this study was to understand the effect of baroreceptor unloading on corticospinal excitability during co-contraction in humans. Motor evoked potential and cortical silent period in a wrist flexor muscle were examined using transcranial magnetic stimulation in two groups of healthy young adults. All subjects performed isometric contraction of the wrist flexors (flexion) and co-contraction of the wrist flexors and extensors (co-contraction). Spinal disynaptic inhibition was also assessed with the ratio of H-reflex responses to unconditioned and conditioned electrical stimulations of the peripheral nerves for the muscles. In one of the groups, baroreflex unloading was induced by applying lower body negative pressure. There was no significant effect of baroreflex unloading on cortical silent period or H-reflex measure of disynaptic inhibition. With baroreflex unloading, motor evoked potential area in the flexor carpi radialis was decreased during co-contraction but not during flexion. The results indicated that baroreceptor unloading decreases corticospinal excitability during co-contraction of antagonistic muscles, apparently by influencing neural pathways that were not probed with cortical silent period or spinal disynaptic inhibition.
Collapse
Affiliation(s)
- Vasiliy E Buharin
- School of Biological Sciences, Georgia Institute of Technology, 555 14th Street NW, Atlanta, GA, 30332-0356, USA.,Activ Surgical, 840 Summer Street, Suite 108, Boston, MA, 02127, USA
| | - Minoru Shinohara
- School of Biological Sciences, Georgia Institute of Technology, 555 14th Street NW, Atlanta, GA, 30332-0356, USA.
| |
Collapse
|
80
|
Niu X, Wu X, Ying A, Shao B, Li X, Zhang W, Lin C, Lin Y. Maternal high fat diet programs hypothalamic-pituitary-adrenal function in adult rat offspring. Psychoneuroendocrinology 2019; 102:128-138. [PMID: 30544004 DOI: 10.1016/j.psyneuen.2018.12.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 11/29/2018] [Accepted: 12/04/2018] [Indexed: 02/04/2023]
Abstract
Maternal environmental factors such as diet have profound effects on offspring development and later health. The hypothalamic-pituitary-adrenal (HPA) axis is an important stress neuroendocrine system that is subject to programming by early life challenges. The present study was further to investigate whether maternal high fat diet (HFD) exposure during rat pregnancy and lactation can alter the HPA axis activity in adult male offspring. We observed that maternal HFD consumption exerted long-term effects on the basal activity of the HPA axis in adult offspring, with increased mean plasma corticosterone levels that result from elevated steroid pulse frequence and pulse amplitude. More importantly, maternal HFD offspring displayed enhanced corticosterone responses to restraint (1 h) and lipopolysaccharide (25 μg/kg, iv) but not insulin-induced hypoglycemia (0.3U/kg, iv) stress, suggesting a stressor-specific effect of maternal diet on the hyperresponsiveness of the HPA axis to stress. Additionally, maternal HFD exposure markedly attenuated the habituation of HPA responses to repeated restraint stress. These findings demonstrate that perinatal HFD exposure has a potent and long-lasting influence on development of neuroendocrine regulatory mechanisms. Maternal HFD consumption significantly increased basal corticotropin-releasing factor (CRF) mRNA expression in the paraventricular nucleus; nevertheless, similar increments in CRF mRNA levels following restraint were observed between maternal HFD offspring and control rats. Furthermore, the medial and central nuclei of amygdala played a pivotal role in maternal HFD-induced sensitization of the HPA response to psychological and systemic stress, respectively, suggesting that different neural pathways may mediate maternal HFD-induced HPA hyperresponsivity to different types of stressors. Take together, the long-term effects of maternal HFD challenge on the central regulation of the HPA axis, therefore, expose the adult offspring to greater HPA function throughout lifespan, in stressor-specific and region-specific manner.
Collapse
Affiliation(s)
- XiaoTing Niu
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - XiaoYun Wu
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - AnNa Ying
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Bei Shao
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - XiaoFeng Li
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - WanLi Zhang
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - ChengCheng Lin
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| | - YuanShao Lin
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| |
Collapse
|
81
|
Mousavi MS, Riazi G, Imani A, Meknatkhah S, Fakhraei N, Pooyan S, Tofigh N. Comparative evaluation of adolescent repeated psychological or physical stress effects on adult cognitive performance, oxidative stress, and heart rate in female rats. Stress 2019; 22:123-132. [PMID: 30345860 DOI: 10.1080/10253890.2018.1507021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Accepted: 07/29/2018] [Indexed: 01/13/2023] Open
Abstract
Multiple adult health problems are associated with adolescent stress. As the brain discriminates physical and psychological stressors by activation of different neural networks, we hypothesized that behavioral and physiological performance would be modulated differently based on the nature of the stressors. Thus, we studied the comparative effects of adolescent repeated physical and psychological stresses on adult cognitive performance, pro-oxidant-antioxidant balance (PAB) and heart rate in female rats. The aim was to differentiate disparate potency of chronic psychological and physical stresses leading to long-term behavioral and physiological alterations. Twenty-one female rats were divided randomly into three groups of seven rats each; control, physical, and psychological stress. Experimental rats were exposed to the stressors for five consecutive days (10 min daily) via a two-communication box. After verifying stress induction by serum corticosterone measurement, the rats were returned to their home cage for 6 weeks, until adulthood, elevated plus maze (EPM), forced swimming test (FST), Y-maze, object recognition task (ORT), and passive avoidance test (PAT) were used as five different behavioral tests to evaluate cognitive performance of each group. Serum PAB and heart rate were measured to assess long-term stress-induced physiological disorders. The results showed exposure to adolescent psychological stress resulted in a larger set of significant changes (in behavioral variation, oxidative stress, and elevated heart rate) 6 weeks post-stress compared to adolescent physical stress. Hence, mental health care in adolescence and therapies targeting PAB and heart rate could be prevention and treatment approaches to confront persistent adolescent stress-induced disorders. Lay summaryThe aim of our study on female laboratory rats was to differentiate disparate potency of chronic psychological and physical stresses in adolescence leading to long-term behavioral and physiological alterations. The results suggest that psychological stresses result in a greater extent of changes compared to physical stress. Adolescent chronic psychological stress may reveal itself in the form of certain behavioral and physiological variations in adulthood. Therefore, mental health care in adolescence could be a valuable prevention approach to confront a variety of adult stress-induced disorders.
Collapse
Affiliation(s)
- Monireh-Sadat Mousavi
- a Laboratory of Neuro-Organic Chemistry , Institute of Biochemistry and Biophysics (IBB), University of Tehran , Tehran , Iran
| | - Gholamhossein Riazi
- a Laboratory of Neuro-Organic Chemistry , Institute of Biochemistry and Biophysics (IBB), University of Tehran , Tehran , Iran
| | - Alireza Imani
- b Department of Physiology, School of Medicine , Tehran University of Medical Sciences , Tehran , Iran
| | - Sogol Meknatkhah
- a Laboratory of Neuro-Organic Chemistry , Institute of Biochemistry and Biophysics (IBB), University of Tehran , Tehran , Iran
| | - Nahid Fakhraei
- c Brain and Spinal Cord Injury Research Center , Neurosciences Institute, Tehran University of Medical Sciences , Tehran , Iran
| | - Shahriar Pooyan
- a Laboratory of Neuro-Organic Chemistry , Institute of Biochemistry and Biophysics (IBB), University of Tehran , Tehran , Iran
- d Rooyan Darou Pharmaceutical Company , Tehran , Iran
| | - Nahid Tofigh
- a Laboratory of Neuro-Organic Chemistry , Institute of Biochemistry and Biophysics (IBB), University of Tehran , Tehran , Iran
| |
Collapse
|
82
|
Hassell JE, Nguyen KT, Gates CA, Lowry CA. The Impact of Stressor Exposure and Glucocorticoids on Anxiety and Fear. Curr Top Behav Neurosci 2019; 43:271-321. [PMID: 30357573 DOI: 10.1007/7854_2018_63] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Anxiety disorders and trauma- and stressor-related disorders, such as posttraumatic stress disorder (PTSD), are common and are associated with significant economic and social burdens. Although trauma and stressor exposure are recognized as a risk factors for development of anxiety disorders and trauma or stressor exposure is recognized as essential for diagnosis of PTSD, the mechanisms through which trauma and stressor exposure lead to these disorders are not well characterized. An improved understanding of the mechanisms through which trauma or stressor exposure leads to development and persistence of anxiety disorders or PTSD may result in novel therapeutic approaches for the treatment of these disorders. Here, we review the current state-of-the-art theories, with respect to mechanisms through which stressor exposure leads to acute or chronic exaggeration of avoidance or anxiety-like defensive behavioral responses and fear, endophenotypes in both anxiety disorders and trauma- and stressor-related psychiatric disorders. In this chapter, we will explore physiological responses and neural circuits involved in the development of acute and chronic exaggeration of anxiety-like defensive behavioral responses and fear states, focusing on the role of the hypothalamic-pituitary-adrenal (HPA) axis and glucocorticoid hormones.
Collapse
Affiliation(s)
- J E Hassell
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
- Center for Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - K T Nguyen
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - C A Gates
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - C A Lowry
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA.
- Center for Neuroscience, University of Colorado Boulder, Boulder, CO, USA.
- Department of Physical Medicine and Rehabilitation, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Center for Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center, Denver Veterans Affairs Medical Center (VAMC), Denver, CO, USA.
- Military and Veteran Microbiome Consortium for Research and Education (MVM-CoRE), Denver, CO, USA.
| |
Collapse
|
83
|
Meknatkhah S, Sharif Dashti P, Mousavi MS, Zeynali A, Ahmadian S, Karima S, Saboury AA, Riazi GH. Psychological stress effects on myelin degradation in the cuprizone-induced model of demyelination. Neuropathology 2018; 39:14-21. [PMID: 30536911 DOI: 10.1111/neup.12522] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 09/28/2018] [Accepted: 09/29/2018] [Indexed: 01/02/2023]
Abstract
Multiple sclerosis (MS) is known as the most common demyelinating disease worldwide in which previous studies have shown that stress is a risk factor for the disease's onset and progression. Nevertheless, further studies are needed to investigate the consequences of stress in MS pathology. In this study, after 5 days of exposure to psychological and physical stress as a repetitive distress modality, rats were treated with cuprizone. The demyelination degree was compared in animal groups using Luxol fast blue staining, immunohistochemical staining for myelin basic protein and transmission electron microscopy. Outcomes revealed that animals exposed to stress prior to cuprizone ingestion, elicit more intense demyelination. Continuous psychological distress has more severe effects on myelin sheath destruction in the preclinical stage.
Collapse
Affiliation(s)
- Sogol Meknatkhah
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Pouya Sharif Dashti
- Department of Chemical Engineering, Faculty of Engineering, University of Tehran, Tehran, Iran
| | | | - Amirbahador Zeynali
- Department of Physics, Iran University of Science and Technology, Tehran, Iran
| | - Shahin Ahmadian
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Saeed Karima
- Clinical Biochemistry Department, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Akbar Saboury
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | | |
Collapse
|
84
|
Role of C1 neurons in anti-inflammatory reflex: Mediation between afferents and efferents. Neurosci Res 2018; 136:6-12. [DOI: 10.1016/j.neures.2018.05.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 04/24/2018] [Accepted: 05/07/2018] [Indexed: 12/21/2022]
|
85
|
Walker LC, Cornish LC, Lawrence AJ, Campbell EJ. The effect of acute or repeated stress on the corticotropin releasing factor system in the CRH-IRES-Cre mouse: A validation study. Neuropharmacology 2018; 154:96-106. [PMID: 30266597 DOI: 10.1016/j.neuropharm.2018.09.037] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Revised: 08/31/2018] [Accepted: 09/22/2018] [Indexed: 12/18/2022]
Abstract
Corticotropin releasing factor (CRF) is a key component of stress responsivity, modulating related behaviors including anxiety and reward. Difficulties identifying CRF neurons, using traditional approaches including immunohistochemistry, has led to the development of a number of transgenic CRF reporter mice. The Crh-IRES-Cre::Ai14 (tdTomato) reporter mouse is increasing in popularity as a useful tool to assess the localization, connectivity and function of CRF neurons in various stress-related behaviors. However, without proper characterization of reporter expression, the in vivo and in vitro manifestations resulting from the manipulation of these cells must be interpreted with caution. Here we mapped the distribution of tdTomato-expressing CRF cells throughout the rostro-caudal extent of the Crh-IRES-Cre::Ai14 mouse brain. To determine if reporter expression faithfully reproduced native CRF expression, we assessed the colocalization of CRF expression with tdTomato reporter expression across several brain regions. Good concordance was observed in the extended amygdala and paraventricular nucleus of the hypothalamus (PVN), while discrepancies were observed within the lateral hypothalamus and hippocampus. Finally, we examined the activation of CRF neurons in Crh-IRES-Cre::Ai14 mice in response to different types of stressors using Fos immunohistochemistry. Acute psychological (swim) and pharmacological (yohimbine) stress stimulated Fos-protein expression in PVN CRF neurons. Interestingly though, exposure to four daily restraint stress sessions followed by a novel acute stressor did not further recruit CRF neurons across any brain region examined. Our results highlight the importance of thoroughly characterizing reporter mice before use and suggest that acute versus repeated stress may differentially impact the CRF system. This article is part of the Special Issue entitled 'Hypothalamic Control of Homeostasis'.
Collapse
Affiliation(s)
- Leigh C Walker
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, 3052, Australia; Florey Department of Neuroscience and Mental Health, The University of Melbourne, Victoria, 3010, Australia
| | - Lara C Cornish
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, 3052, Australia
| | - Andrew J Lawrence
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, 3052, Australia; Florey Department of Neuroscience and Mental Health, The University of Melbourne, Victoria, 3010, Australia
| | - Erin J Campbell
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, 3052, Australia; Florey Department of Neuroscience and Mental Health, The University of Melbourne, Victoria, 3010, Australia.
| |
Collapse
|
86
|
Nosjean A, de Chaumont F, Olivo-Marin JC, Granon S. Stress-induced brain activation: buffering role of social behavior and neuronal nicotinic receptors. Brain Struct Funct 2018; 223:4259-4274. [DOI: 10.1007/s00429-018-1745-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 08/30/2018] [Indexed: 11/28/2022]
|
87
|
Common and differential transcriptional responses to different models of traumatic stress exposure in rats. Transl Psychiatry 2018; 8:165. [PMID: 30139969 PMCID: PMC6107654 DOI: 10.1038/s41398-018-0223-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 07/14/2018] [Indexed: 11/22/2022] Open
Abstract
The effect of six different traumatic stress protocols on the transcriptome of the rat adrenal gland was examined using RNA sequencing. These protocols included chronic variable stress, chronic shock, social defeat and social isolation. The response of the transcriptome to stress suggested that there are genes that respond in a universal or stress modality-independent manner, as well as genes that respond in a stress modality-specific manner. Using a small number of the genes selected from the modality-independent set of stress-sensitive genes, a sensitive and robust measure of chronic stress exposure was developed. This stress-sensitive gene expression (SSGE) index could detect chronic traumatic stress exposure in a wide range of different stress models in a manner that was relatively independent of the modality of stress exposure and that paralleled the intensity of stress exposure in a dose-dependent manner. This measure could reliably distinguish control and stressed individuals in the case of animals exposed to the most intense stress protocols. The response of a subset of the modality-specific genes could also distinguish some types of stress exposure, based solely on changes in the pattern of gene expression. The results suggest that it is possible to develop diagnostic measures of traumatic stress exposure based solely on changes in the level of expression of a relatively small number of genes.
Collapse
|
88
|
Kovács LÁ, Schiessl JA, Nafz AE, Csernus V, Gaszner B. Both Basal and Acute Restraint Stress-Induced c-Fos Expression Is Influenced by Age in the Extended Amygdala and Brainstem Stress Centers in Male Rats. Front Aging Neurosci 2018; 10:248. [PMID: 30186150 PMCID: PMC6113579 DOI: 10.3389/fnagi.2018.00248] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 07/30/2018] [Indexed: 01/03/2023] Open
Abstract
The hypothalamus-pituitary-adrenal axis (HPA) is the main regulator of the stress response. The key of the HPA is the parvocellular paraventricular nucleus of the hypothalamus (pPVN) controlled by higher-order limbic stress centers. The reactivity of the HPA axis is considered to be a function of age, but to date, little is known about the background of this age-dependency. Sporadic literature data suggest that the stress sensitivity as assessed by semi-quantitation of the neuronal activity marker c-Fos may also be influenced by age. Here, we aimed at investigating the HPA activity and c-Fos immunoreactivity 2 h after the beginning of a single 60 min acute restraint stress in eight age groups of male Wistar rats. We hypothesized that the function of the HPA axis (i.e., pPVN c-Fos and blood corticosterone (CORT) level), the neuronal activity of nine stress-related limbic areas (i.e., magnocellular PVN (mPVN), medial (MeA), central (CeA), basolateral nuclei of the amygdala, the oval (ovBNST), dorsolateral (dlBNST), dorsomedial (dmBNST), ventral and fusiform (fuBNST) divisions of the bed nucleus of the stria terminalis (BNST)), and two brainstem stress centers such as the centrally projecting Edinger-Westphal nucleus (cpEW) and dorsal raphe nucleus (DR) show age dependency in their c-Fos response. The somatosensory barrel cortex area (S1) was evaluated to test whether the age dependency is specific for stress-centers. Our results indicate that the stress-induced rise in blood CORT titer was lower in young age reflecting relatively low HPA activity. All 12 stress-related brain areas showed c-Fos response that peaked at 2 months of age. The magnitude of c-Fos immunoreactivity correlated negatively with age in seven regions (MeA, CeA, ovBNST, dlBNST, dmBNST, fuBNST and pPVN). Unexpectedly, the CeA, ovBNST and cpEW showed a considerable basal c-Fos expression in 1-month-old rats which decreased with age. The S1 showed a U-shaped age-related dynamics in contrast to the decline observed in stress centers. We conclude that the age- and brain area dependent dynamics in stress-induced neuronal activity pattern may contribute to the age dependance of the stress reactivity. Further studies are in progress to determine the neurochemical identity of neurons showing age-dependent basal and/or stress-induced c-Fos expression.
Collapse
Affiliation(s)
- László Ákos Kovács
- Department of Anatomy, Medical School, University of Pécs, Pécs, Hungary.,Center for Neuroscience, Pécs University, Pécs, Hungary
| | | | | | - Valér Csernus
- Department of Anatomy, Medical School, University of Pécs, Pécs, Hungary
| | - Balázs Gaszner
- Department of Anatomy, Medical School, University of Pécs, Pécs, Hungary.,Center for Neuroscience, Pécs University, Pécs, Hungary
| |
Collapse
|
89
|
Godoy LD, Rossignoli MT, Delfino-Pereira P, Garcia-Cairasco N, de Lima Umeoka EH. A Comprehensive Overview on Stress Neurobiology: Basic Concepts and Clinical Implications. Front Behav Neurosci 2018; 12:127. [PMID: 30034327 PMCID: PMC6043787 DOI: 10.3389/fnbeh.2018.00127] [Citation(s) in RCA: 420] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 06/06/2018] [Indexed: 12/20/2022] Open
Abstract
Stress is recognized as an important issue in basic and clinical neuroscience research, based upon the founding historical studies by Walter Canon and Hans Selye in the past century, when the concept of stress emerged in a biological and adaptive perspective. A lot of research after that period has expanded the knowledge in the stress field. Since then, it was discovered that the response to stressful stimuli is elaborated and triggered by the, now known, stress system, which integrates a wide diversity of brain structures that, collectively, are able to detect events and interpret them as real or potential threats. However, different types of stressors engage different brain networks, requiring a fine-tuned functional neuroanatomical processing. This integration of information from the stressor itself may result in a rapid activation of the Sympathetic-Adreno-Medullar (SAM) axis and the Hypothalamus-Pituitary-Adrenal (HPA) axis, the two major components involved in the stress response. The complexity of the stress response is not restricted to neuroanatomy or to SAM and HPA axes mediators, but also diverge according to timing and duration of stressor exposure, as well as its short- and/or long-term consequences. The identification of neuronal circuits of stress, as well as their interaction with mediator molecules over time is critical, not only for understanding the physiological stress responses, but also to understand their implications on mental health.
Collapse
Affiliation(s)
- Lívea Dornela Godoy
- Physiology Department, Ribeirão Preto School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Matheus Teixeira Rossignoli
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Polianna Delfino-Pereira
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Norberto Garcia-Cairasco
- Physiology Department, Ribeirão Preto School of Medicine, University of São Paulo, São Paulo, Brazil
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Eduardo Henrique de Lima Umeoka
- Department of Neuroscience and Behavioral Sciences, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
90
|
Mork R, Falkenberg HK, Fostervold KI, Thorud HMS. Visual and psychological stress during computer work in healthy, young females-physiological responses. Int Arch Occup Environ Health 2018; 91:811-830. [PMID: 29850947 PMCID: PMC6132651 DOI: 10.1007/s00420-018-1324-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 05/22/2018] [Indexed: 11/26/2022]
Abstract
Purpose Among computer workers, visual complaints, and neck pain are highly prevalent. This study explores how occupational simulated stressors during computer work, like glare and psychosocial stress, affect physiological responses in young females with normal vision. Methods The study was a within-subject laboratory experiment with a counterbalanced, repeated design. Forty-three females performed four 10-min computer-work sessions with different stress exposures: (1) minimal stress; (2) visual stress (direct glare); (3) psychological stress; and (4) combined visual and psychological stress. Muscle activity and muscle blood flow in trapezius, muscle blood flow in orbicularis oculi, heart rate, blood pressure, blink rate and postural angles were continuously recorded. Immediately after each computer-work session, fixation disparity was measured and a questionnaire regarding perceived workstation lighting and stress was completed. Results Exposure to direct glare resulted in increased trapezius muscle blood flow, increased blink rate, and forward bending of the head. Psychological stress induced a transient increase in trapezius muscle activity and a more forward-bent posture. Bending forward towards the computer screen was correlated with higher productivity (reading speed), indicating a concentration or stress response. Forward bent posture was also associated with changes in fixation disparity. Furthermore, during computer work per se, trapezius muscle activity and blood flow, orbicularis oculi muscle blood flow, and heart rate were increased compared to rest. Conclusions Exposure to glare and psychological stress during computer work were shown to influence the trapezius muscle, posture, and blink rate in young, healthy females with normal binocular vision, but in different ways. Accordingly, both visual and psychological factors must be taken into account when optimizing computer workstations to reduce physiological responses that may cause excessive eyestrain and musculoskeletal load.
Collapse
Affiliation(s)
- Randi Mork
- Department of Public Health Science, Norwegian University of Life Sciences, Ås, Norway.
- Department of Optometry, Radiography and Lighting Design, University of South-Eastern Norway, National Centre for Optics, Vision and Eye Care, P.O. Box 235, 3603, Kongsberg, Norway.
| | - Helle K Falkenberg
- Department of Optometry, Radiography and Lighting Design, University of South-Eastern Norway, National Centre for Optics, Vision and Eye Care, P.O. Box 235, 3603, Kongsberg, Norway
| | | | - Hanne Mari S Thorud
- Department of Optometry, Radiography and Lighting Design, University of South-Eastern Norway, National Centre for Optics, Vision and Eye Care, P.O. Box 235, 3603, Kongsberg, Norway
| |
Collapse
|
91
|
Godoy LD, Umeoka EHL, Ribeiro DE, Santos VR, Antunes-Rodrigues J, Joca SRL, Garcia-Cairasco N. Multimodal early-life stress induces biological changes associated to psychopathologies. Horm Behav 2018; 100:69-80. [PMID: 29548783 DOI: 10.1016/j.yhbeh.2018.03.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 03/07/2018] [Accepted: 03/11/2018] [Indexed: 01/21/2023]
Abstract
Evidences suggest the contributive role of early-life stress (ELS) to affective and anxiety disorders. Chronic exposure to the same stressor may generate habituation, while the exposure to different and repeated stressors gradually promotes maladaptive plasticity. Therefore, to further understand the effects of heterotypic stressors during early life period, male Wistar rat pups (P1-P21) were exposed to Multimodal ELS paradigm. Results indicate pups did not habituate to multimodal ELS and neonates respond to both physical and psychogenic stressors. Adult rats that underwent ELS protocol showed significant lower sucrose intake, decreased latency to immobility in the forced swim test and increased latency to light compartment in the light-dark test when compared to control group. Although it has been shown that ELS-induced changes in hippocampus can be used as biomarkers, multimodal ELS did not significantly alter BDNF, Tyrosine Kinase B (TrkB) receptor expression or neurogenesis in the hippocampus. Taken together, these findings indicate that multimodal ELS protocol can be an interesting experimental model for understanding long-term psychiatric disorders associated with stress. Indeed, our data with neurogenesis, BDNF and TrkB, and conflicting data from the literature, suggest that additional studies on synaptic plasticity/intracellular cascades would help to detect the underlying mechanisms.
Collapse
Affiliation(s)
- Lívea Dornela Godoy
- Physiology Department, Ribeirão Preto School of Medicine, University of São Paulo, Brazil
| | - Eduardo H L Umeoka
- Neurosciences and Behavioral Sciences Department, Ribeirão Preto School of Medicine, University of São Paulo, Brazil
| | - Deidiane Elisa Ribeiro
- Pharmacology Department, Ribeirão Preto School of Medicine, University of São Paulo, Brazil
| | | | - José Antunes-Rodrigues
- Physiology Department, Ribeirão Preto School of Medicine, University of São Paulo, Brazil
| | - Samia Regiane Lourenço Joca
- Physics and Chemistry Department, Ribeirão Preto School of Pharmacy, University of São Paulo, Brazil; Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Denmark
| | - Norberto Garcia-Cairasco
- Physiology Department, Ribeirão Preto School of Medicine, University of São Paulo, Brazil; Neurosciences and Behavioral Sciences Department, Ribeirão Preto School of Medicine, University of São Paulo, Brazil.
| |
Collapse
|
92
|
Spencer RL, Chun LE, Hartsock MJ, Woodruff ER. Glucocorticoid hormones are both a major circadian signal and major stress signal: How this shared signal contributes to a dynamic relationship between the circadian and stress systems. Front Neuroendocrinol 2018; 49:52-71. [PMID: 29288075 DOI: 10.1016/j.yfrne.2017.12.005] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 12/23/2017] [Accepted: 12/23/2017] [Indexed: 12/13/2022]
Abstract
Glucocorticoid hormones are a powerful mammalian systemic hormonal signal that exerts regulatory effects on almost every cell and system of the body. Glucocorticoids act in a circadian and stress-directed manner to aid in adaptation to an ever-changing environment. Circadian glucocorticoid secretion provides for a daily waxing and waning influence on target cell function. In addition, the daily circadian peak of glucocorticoid secretion serves as a timing signal that helps entrain intrinsic molecular clock phase in tissue cells distributed throughout the body. Stress-induced glucocorticoid secretion also modulates the state of these same cells in response to both physiological and psychological stressors. We review the strong functional interrelationships between glucocorticoids and the circadian system, and discuss how these interactions optimize the appropriate cellular and systems response to stress throughout the day. We also discuss clinical implications of this dual aspect of glucocorticoid signaling, especially for conditions of circadian and HPA axis dysregulation.
Collapse
Affiliation(s)
- Robert L Spencer
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Lauren E Chun
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Matthew J Hartsock
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA.
| | - Elizabeth R Woodruff
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO 80309, USA.
| |
Collapse
|
93
|
Anteroventral bed nuclei of the stria terminalis neurocircuitry: Towards an integration of HPA axis modulation with coping behaviors - Curt Richter Award Paper 2017. Psychoneuroendocrinology 2018; 89:239-249. [PMID: 29395488 PMCID: PMC5878723 DOI: 10.1016/j.psyneuen.2017.12.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 11/19/2017] [Accepted: 12/11/2017] [Indexed: 12/15/2022]
Abstract
A network of interconnected cell groups in the limbic forebrain regulates hypothalamic-pituitary-adrenal (HPA) axis activation and behavioral responses to emotionally stressful experiences, and chronic disruption of these systems chronically is implicated in the pathogenesis of psychiatric illnesses. A significant challenge has been to unravel the circuitry and mechanisms providing for regulation of HPA activity, as these limbic forebrain regions do not provide any direct innervation of HPA effector cell groups in the paraventricular hypothalamus (PVH). Moreover, information regarding how endocrine and behavioral responses are integrated has remained obscure. Here we summarize work from our laboratory showing that anteroventral (av) bed nuclei of the stria terminalis (BST) acts as a point of convergence between the limbic forebrain and PVH, receiving and coordinating upstream influences, and restraining HPA axis output in response to inescapable stressors. Recent studies highlight a more expansive modulatory role for avBST as one that coordinates HPA-inhibitory influences while concurrently suppressing passive behavioral responses via divergent pathways. avBST is uniquely positioned to convey endocrine and behavioral alterations resulting from chronic stress exposure, such as HPA axis hyperactivity and increased passive coping strategies, that may result from synaptic reorganization in upstream limbic cortical regions. We discuss how these studies give new insights into understanding the systems-level organization of stress response circuitry, the neurobiology of coping styles, and BST circuit dysfunction in stress-related psychiatric disorders.
Collapse
|
94
|
Úbeda-Contreras J, Marín-Blasco I, Nadal R, Armario A. Brain c-fos expression patterns induced by emotional stressors differing in nature and intensity. Brain Struct Funct 2018; 223:2213-2227. [PMID: 29450645 DOI: 10.1007/s00429-018-1624-2] [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: 03/22/2017] [Accepted: 02/06/2018] [Indexed: 01/09/2023]
Abstract
Regardless of its particular nature, emotional stressors appear to elicit a widespread and roughly similar brain activation pattern as evaluated by c-fos expression. However, their behavioral and physiological consequences may strongly differ. Here we addressed in adult male rats the contribution of the intensity and the particular nature of stressors by comparing, in a set of brain areas, the number of c-fos expressing neurons in response to open-field, cat odor or immobilization on boards (IMO). These are qualitatively different stressors that are known to differ in terms of intensity, as evaluated by biological markers. In the present study, plasma levels of the adrenocorticotropic hormone (ACTH) demonstrated that intensity increases in the following order: open-field, cat odor and IMO. Four different c-fos activation patterns emerged among all areas studied: (i) positive relationship with intensity (posterior-dorsal medial amygdala, dorsomedial hypothalamus, lateral septum ventral and paraventricular nucleus of the hypothalamus), (ii) negative relationship with intensity (cingulate cortex 1, posterior insular cortex, dorsal striatum, nucleus accumbens and some subdivisions of the hippocampal formation); (iii) activation not dependent on the intensity of the stressor (prelimbic and infralimbic cortex and lateral and basolateral amygdala); and (iv) activation specifically associated with cat odor (ventromedial amygdala and ventromedial hypothalamus). Histone 3 phosphorylation at serine 10, another neuronal activation marker, corroborated c-fos results. Summarizing, deepest analysis of the brain activation pattern elicit by emotional stressor indicated that, in spite of activating similar areas, each stressor possess their own brain activation signature, mediated mainly by qualitative aspects but also by intensity.
Collapse
Affiliation(s)
- Jesús Úbeda-Contreras
- Institut de Neurociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193, Barcelona, Spain.,CIBERSAM, Instituto de Salud Carlos III, Madrid, Spain.,Animal Physiology Unit, School of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193, Barcelona, Spain
| | - Ignacio Marín-Blasco
- Institut de Neurociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193, Barcelona, Spain.,Animal Physiology Unit, School of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193, Barcelona, Spain.,Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Roser Nadal
- Institut de Neurociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193, Barcelona, Spain.,CIBERSAM, Instituto de Salud Carlos III, Madrid, Spain.,Psychobiology Unit, School of Psychology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193, Barcelona, Spain
| | - Antonio Armario
- Institut de Neurociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193, Barcelona, Spain. .,CIBERSAM, Instituto de Salud Carlos III, Madrid, Spain. .,Animal Physiology Unit, School of Biosciences, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, 08193, Barcelona, Spain.
| |
Collapse
|
95
|
Estrada CM, Ghisays V, Nguyen ET, Caldwell JL, Streicher J, Solomon MB. Estrogen signaling in the medial amygdala decreases emotional stress responses and obesity in ovariectomized rats. Horm Behav 2018; 98:33-44. [PMID: 29248436 DOI: 10.1016/j.yhbeh.2017.12.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Revised: 12/01/2017] [Accepted: 12/09/2017] [Indexed: 12/15/2022]
Abstract
Declining estradiol (E2), as occurs during menopause, increases risk for obesity and psychopathology (i.e., depression, anxiety). E2 modulates mood and energy homeostasis via binding to estrogen receptors (ER) in the brain. The often comorbid and bidirectional relationship between mood and metabolic disorders suggests shared hormonal and/or brain networks. The medial amygdala (MeA) is abundant in ERs and regulates mood, endocrine, and metabolic stress responses; therefore we tested the hypothesis that E2 in the MeA mitigates emotional and metabolic dysfunction in a rodent model of surgical menopause. Adult female rats were ovariectomized (OVX) and received bilateral implants of E2 or cholesterol micropellets aimed at the MeA. E2-MeA decreased anxiety-like (center entries, center time) and depression-like (immobility) behaviors in the open field and forced swim tests (FST), respectively in ovariectomized rats. E2-MeA also prevented hyperphagia, body weight gain, increased visceral adiposity, and glucose intolerance in ovariectomized rats. E2-MeA decreased caloric efficiency, suggestive of increased energy expenditure. E2-MeA also modulated c-Fos neural activity in amygdalar (central and medial) and hypothalamic (paraventricular and arcuate) brain regions that regulate mood and energy homeostasis in response to the FST, a physically demanding task. Given the shared neural circuitry between mood and body weight regulation, c-Fos expression in discrete brain regions in response to the FST may be due to the psychologically stressful and/or metabolic demands of the task. Together, these findings suggest that the MeA is a critical node for mediating estrogenic effects on mood and energy homeostasis.
Collapse
Affiliation(s)
- Christina M Estrada
- Department of Psychology Experimental Psychology Program, University of Cincinnati, Cincinnati, OH 45237, United States
| | - Valentina Ghisays
- Department of Psychology Experimental Psychology Program, University of Cincinnati, Cincinnati, OH 45237, United States
| | - Elizabeth T Nguyen
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH 45237, United States
| | - Jody L Caldwell
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH 45237, United States
| | - Joshua Streicher
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH 45237, United States
| | - Matia B Solomon
- Department of Psychology Experimental Psychology Program, University of Cincinnati, Cincinnati, OH 45237, United States; Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH 45237, United States; Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH 45237, United States.
| |
Collapse
|
96
|
Liu L, Zhou X, Zhang Y, Pu J, Yang L, Yuan S, Zhao L, Zhou C, Zhang H, Xie P. Hippocampal metabolic differences implicate distinctions between physical and psychological stress in four rat models of depression. Transl Psychiatry 2018; 8:4. [PMID: 29317595 PMCID: PMC5802536 DOI: 10.1038/s41398-017-0018-1] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 08/15/2017] [Accepted: 08/20/2017] [Indexed: 12/28/2022] Open
Abstract
Major depressive disorder (MDD) is a heterogeneous and multi-factorial disorder, and the underlying molecular mechanisms remain largely unknown. However, many studies have indicated that the molecular mechanisms underlying depression in response to different stress may differ. After screening, 28-30 rats were included in each model of depression (chronic unpredictable mild stress (CUMS); learned helplessness (LH); chronic restraint stress (CRS); or social defeat (SD)). Non-targeted gas chromatography-mass spectrometry was used to profile the metabolic changes in the hippocampus. As a result, all four models exhibited significant depression-like behavior. A total of 30, 24, 19, and 25 differential metabolites were identified in the CUMS, LH, CRS, and SD models, respectively. Interestingly, the hierarchical clustering results revealed two patterns of metabolic changes that are characteristic of the response to cluster 1 (CUMS, LH) and cluster 2 (CRS, SD) stress, which represent physical and psychological stress, respectively. Bioinformatic analysis suggested that physical stress was mainly associated with lipid metabolism and glutamate metabolism, whereas psychological stress was related to cell signaling, cellular proliferation, and neurodevelopment, suggesting the molecular changes induced by physical and psychological stress were different. Nine shared metabolites were opposite in the directions of change between physical and psychological models, and these metabolites were associated with cellular proliferation and neurodevelopment functions, indicating the response to physical and psychological stress was different in the activation and deactivation of the final common pathway to depression. Our results provide a further understanding of the heterogeneity in the molecular mechanisms of MDD that could facilitate the development of personalized medicine for this disorder.
Collapse
Affiliation(s)
- Lanxiang Liu
- grid.452206.7Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China ,0000 0000 8653 0555grid.203458.8Institute of Neuroscience and The Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
| | - Xinyu Zhou
- 0000 0000 8653 0555grid.203458.8Institute of Neuroscience and The Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China ,grid.452206.7Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuqing Zhang
- grid.452206.7Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China ,0000 0000 8653 0555grid.203458.8Institute of Neuroscience and The Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
| | - Juncai Pu
- grid.452206.7Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China ,0000 0000 8653 0555grid.203458.8Institute of Neuroscience and The Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
| | - Lining Yang
- grid.452206.7Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China ,0000 0000 8653 0555grid.203458.8Institute of Neuroscience and The Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
| | - Shuai Yuan
- 0000 0000 8653 0555grid.203458.8Institute of Neuroscience and The Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
| | - Libo Zhao
- 0000 0000 8653 0555grid.203458.8Institute of Neuroscience and The Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China ,0000 0000 8653 0555grid.203458.8Department of Neurology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Chanjun Zhou
- 0000 0000 8653 0555grid.203458.8Institute of Neuroscience and The Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China ,0000 0000 8653 0555grid.203458.8Department of Neurology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Hanping Zhang
- grid.452206.7Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China ,0000 0000 8653 0555grid.203458.8Institute of Neuroscience and The Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
| | - Peng Xie
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China. .,Institute of Neuroscience and The Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
97
|
Sominsky L, Hodgson DM, McLaughlin EA, Smith R, Wall HM, Spencer SJ. Linking Stress and Infertility: A Novel Role for Ghrelin. Endocr Rev 2017; 38:432-467. [PMID: 28938425 DOI: 10.1210/er.2016-1133] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 07/24/2017] [Indexed: 12/23/2022]
Abstract
Infertility affects a remarkable one in four couples in developing countries. Psychological stress is a ubiquitous facet of life, and although stress affects us all at some point, prolonged or unmanageable stress may become harmful for some individuals, negatively impacting on their health, including fertility. For instance, women who struggle to conceive are twice as likely to suffer from emotional distress than fertile women. Assisted reproductive technology treatments place an additional physical, emotional, and financial burden of stress, particularly on women, who are often exposed to invasive techniques associated with treatment. Stress-reduction interventions can reduce negative affect and in some cases to improve in vitro fertilization outcomes. Although it has been well-established that stress negatively affects fertility in animal models, human research remains inconsistent due to individual differences and methodological flaws. Attempts to isolate single causal links between stress and infertility have not yet been successful due to their multifaceted etiologies. In this review, we will discuss the current literature in the field of stress-induced reproductive dysfunction based on animal and human models, and introduce a recently unexplored link between stress and infertility, the gut-derived hormone, ghrelin. We also present evidence from recent seminal studies demonstrating that ghrelin has a principal role in the stress response and reward processing, as well as in regulating reproductive function, and that these roles are tightly interlinked. Collectively, these data support the hypothesis that stress may negatively impact upon fertility at least in part by stimulating a dysregulation in ghrelin signaling.
Collapse
Affiliation(s)
- Luba Sominsky
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria 3083, Australia
| | - Deborah M Hodgson
- School of Psychology, Faculty of Science and IT, The University of Newcastle, New South Wales 2308, Australia
| | - Eileen A McLaughlin
- School of Biological Sciences, Faculty of Science, The University of Auckland, Auckland 1010, New Zealand.,School of Environmental & Life Sciences, Faculty of Science and IT, The University of Newcastle, New South Wales 2308, Australia
| | - Roger Smith
- Mothers and Babies Research Centre, Hunter Medical Research Institute, Lookout Road, New Lambton Heights, New South Wales 2305, Australia.,Priority Research Centre in Reproductive Science, The University of Newcastle, New South Wales 2308, Australia
| | - Hannah M Wall
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria 3083, Australia
| | - Sarah J Spencer
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria 3083, Australia
| |
Collapse
|
98
|
Carter DA, Choong YT, Connelly AA, Bassi JK, Hunter NO, Thongsepee N, Llewellyn-Smith IJ, Fong AY, McDougall SJ, Allen AM. Functional and neurochemical characterization of angiotensin type 1A receptor-expressing neurons in the nucleus of the solitary tract of the mouse. Am J Physiol Regul Integr Comp Physiol 2017; 313:R438-R449. [PMID: 28701322 DOI: 10.1152/ajpregu.00168.2017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 07/10/2017] [Accepted: 07/10/2017] [Indexed: 11/22/2022]
Abstract
Angiotensin II acts via two main receptors within the central nervous system, with the type 1A receptor (AT1AR) most widely expressed in adult neurons. Activation of the AT1R in the nucleus of the solitary tract (NTS), the principal nucleus receiving central synapses of viscerosensory afferents, modulates cardiovascular reflexes. Expression of the AT1R occurs in high density within the NTS of most mammals, including humans, but the fundamental electrophysiological and neurochemical characteristics of the AT1AR-expressing NTS neurons are not known. To address this, we have used a transgenic mouse, in which the AT1AR promoter drives expression of green fluorescent protein (GFP). Approximately one-third of AT1AR-expressing neurons express the catecholamine-synthetic enzyme tyrosine hydroxylase (TH), and a subpopulation of these stained for the transcription factor paired-like homeobox 2b (Phox2b). A third group, comprising approximately two-thirds of the AT1AR-expressing NTS neurons, showed Phox2b immunoreactivity alone. A fourth group in the ventral subnucleus expressed neither TH nor Phox2b. In whole cell recordings from slices in vitro, AT1AR-GFP neurons exhibited voltage-activated potassium currents, including the transient outward current and the M-type potassium current. In two different mouse strains, both AT1AR-GFP neurons and TH-GFP neurons showed similar AT1AR-mediated depolarizing responses to superfusion with angiotensin II. These data provide a comprehensive description of AT1AR-expressing neurons in the NTS and increase our understanding of the complex actions of this neuropeptide in the modulation of viscerosensory processing.
Collapse
Affiliation(s)
- D A Carter
- Department of Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Y-T Choong
- Department of Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - A A Connelly
- Department of Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - J K Bassi
- Department of Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - N O Hunter
- Department of Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - N Thongsepee
- Department of Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - I J Llewellyn-Smith
- Cardiovascular Medicine and Human Physiology, School of Medicine, Flinders University, Bedford Park, South Australia, Australia; and
| | - A Y Fong
- Department of Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - S J McDougall
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - A M Allen
- Department of Physiology, The University of Melbourne, Melbourne, Victoria, Australia; .,Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
99
|
Randich A, DeWitte C, DeBerry JJ, Robbins MT, Ness TJ. Lesions of the central amygdala and ventromedial medulla reduce bladder hypersensitivity produced by acute but not chronic foot shock. Brain Res 2017; 1675:1-7. [PMID: 28867481 DOI: 10.1016/j.brainres.2017.08.032] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Revised: 08/11/2017] [Accepted: 08/28/2017] [Indexed: 02/07/2023]
Abstract
Both acute and chronic stress has been shown to exacerbate symptoms of chronic visceral pain conditions such as interstitial cystitis. Studies using animal models support these findings in that both acute and chronic exposure to foot shock-induced stress (FS) augment nociceptive reflex responses to urinary bladder distension (UBD). Only a few studies have examined the neural substrates mediating these phenomena and it is not clear whether acute and chronic stress engage the same or different substrates to produce bladder hypersensitivity. The present studies examined the role of two important central nervous system structures - the amygdala (AMG) and the ventromedial medulla (VMM) - in mediating/modulating hypersensitivity evoked by acute versus chronic FS using responses to graded UBD in adult, female Sprague-Dawley rats. Bladder hypersensitivity produced by acute FS was significantly reduced by either bilateral central AMG or VMM lesions using measures generated by graded UBD, but these lesions had no significant effects using the same measures on bladder hyperalgesia produced by chronic FS. Our findings provide evidence that neural substrates underlying bladder hypersensitivity produced by chronic stress differ from those produced by acute stress. These findings suggest that while the AMG and VMM participate in pain processing during periods of limited exposure to stress, prolonged stress may recruit a new set of neural substrates not initially activated by acute exposure to stress.
Collapse
Affiliation(s)
- Alan Randich
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Cary DeWitte
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jennifer J DeBerry
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Meredith T Robbins
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Timothy J Ness
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL, United States.
| |
Collapse
|
100
|
Pham L, Baker MR, Shahanoor Z, Romeo RD. Adolescent changes in hindbrain noradrenergic A2 neurons in male rats. Brain Res 2017; 1666:11-16. [DOI: 10.1016/j.brainres.2017.04.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 04/04/2017] [Accepted: 04/15/2017] [Indexed: 12/19/2022]
|