701
|
Cortisol levels and seizures in adults with epilepsy: A systematic review. Neurosci Biobehav Rev 2019; 103:216-229. [PMID: 31129236 DOI: 10.1016/j.neubiorev.2019.05.023] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 05/21/2019] [Accepted: 05/21/2019] [Indexed: 12/11/2022]
Abstract
Stress has been suggested as a trigger factor for seizures in epilepsy patients, but little is known about cortisol levels, as indicators of stress, in adults with epilepsy. This systematic review summarizes the evidence on this topic. Following PRISMA guidelines, 38 articles were selected: 14 analyzing basal cortisol levels, eight examining antiepileptic drugs (AEDs) effects, 13 focused on seizure effects, and three examining stress. Higher basal cortisol levels were found in patients than in healthy people in studies with the most homogeneous samples (45% of 38 total studies). Despite heterogeneous results associated with AEDs, seizures were related to increases in cortisol levels in 77% of 38 total studies. The only study with acute stress administration found higher cortisol reactivity in epilepsy than in healthy controls. In studies using self-reported stress, high seizure frequency was related to increased cortisol levels and lower functional brain connectivity. Findings suggest that epilepsy could be considered a chronic stress model. The potential sensitizing role of accumulative seizures and issues for future research are discussed.
Collapse
|
702
|
Davis EP, Hankin BL, Glynn LM, Head K, Kim DJ, Sandman CA. Prenatal Maternal Stress, Child Cortical Thickness, and Adolescent Depressive Symptoms. Child Dev 2019; 91:e432-e450. [PMID: 31073997 DOI: 10.1111/cdev.13252] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Prenatal maternal stress predicts subsequent elevations in youth depressive symptoms, but the neural processes associated with these links are unclear. This study evaluated whether prenatal maternal stress is associated with child brain development, and adolescent depressive symptoms using a prospective design with 74 mother child pairs (40 boys). Maternal stress was assessed during pregnancy, child cortical thickness at age 7, and depressive symptoms at age 12. Prenatal maternal stress was associated with less cortical thickness primarily in frontal and temporal regions and with elevated depressive symptoms; child cortical thickness additionally correlated with adolescent depressive symptoms. The observed associations are consistent with the possibility that cortical thickness in superior frontal regions links associations between prenatal maternal stress and adolescent depressive symptoms.
Collapse
|
703
|
Reich B, Zhou Y, Goldstein E, Srivats SS, Contoreggi NH, Kogan JF, McEwen BS, Kreek MJ, Milner TA, Gray JD. Chronic immobilization stress primes the hippocampal opioid system for oxycodone-associated learning in female but not male rats. Synapse 2019; 73:e22088. [PMID: 30632204 PMCID: PMC11548942 DOI: 10.1002/syn.22088] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 12/27/2018] [Accepted: 01/06/2019] [Indexed: 12/21/2022]
Abstract
In adult female, but not male, Sprague Dawley rats, chronic immobilization stress (CIS) increases mossy fiber (MF) Leu-Enkephalin levels and redistributes delta- and mu-opioid receptors (DORs and MORs) in hippocampal CA3 pyramidal cells and GABAergic interneurons to promote excitation and learning processes following subsequent opioid exposure. Here, we demonstrate that CIS females, but not males, acquire conditioned place preference (CPP) to oxycodone and that CIS "primes" the hippocampal opioid system in females for oxycodone-associated learning. In CA3b, oxycodone-injected (Oxy) CIS females relative to saline-injected (Sal) CIS females exhibited an increase in the cytoplasmic and total densities of DORs in pyramidal cell dendrites so that they were similar to Sal- and Oxy-CIS males. Consistent with our earlier studies, Sal- and Oxy-CIS females but not CIS males had elevated DOR densities in MF-CA3 dendritic spines, which we have previously shown are important for opioid-mediated long-term potentiation. In the dentate gyrus, Oxy-CIS females had more DOR-labeled interneurons than Sal-CIS females. Moreover, Sal- and Oxy-CIS females compared to both groups of CIS males had elevated levels of DORs and MORs in GABAergic interneuron dendrites, suggesting capacity for greater synthesis or storage of these receptors in circuits important for opioid-mediated disinhibition. However, more plasmalemmal MORs were on large parvalbumin-containing dendrites of Oxy-CIS males compared to Sal-CIS males, suggesting a limited ability for increased granule cell disinhibition. These results suggest that low levels of DORs in MF-CA3 synapses and hilar GABAergic interneurons may contribute to the attenuation of oxycodone CPP in males exposed to CIS.
Collapse
MESH Headings
- Analgesics, Opioid/pharmacology
- Animals
- CA3 Region, Hippocampal/cytology
- CA3 Region, Hippocampal/drug effects
- CA3 Region, Hippocampal/metabolism
- Conditioning, Classical
- Dendrites/metabolism
- Dentate Gyrus/cytology
- Dentate Gyrus/drug effects
- Dentate Gyrus/metabolism
- Female
- Interneurons/metabolism
- Male
- Oxycodone/pharmacology
- Rats
- Rats, Sprague-Dawley
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, mu/metabolism
- Repetition Priming
- Restraint, Physical
- Stress, Psychological/metabolism
- Stress, Psychological/physiopathology
Collapse
Affiliation(s)
- Batsheva Reich
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065
| | - Yan Zhou
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065
| | - Ellen Goldstein
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065
| | - Sudarshan S. Srivats
- Weill Cornell Medicine in Qatar, Qatar Foundation, Education City, P.O. Box 24144 - Doha, Qatar
| | - Natalina H. Contoreggi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065
| | - Joshua F. Kogan
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065
| | - Bruce S. McEwen
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065
| | - Mary Jeanne Kreek
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065
| | - Teresa A. Milner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065
| | - Jason D. Gray
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY 10065
| |
Collapse
|
704
|
Monteiro C, Cardoso-Cruz H, Galhardo V. Animal models of congenital hypoalgesia: Untapped potential for assessing pain-related plasticity. Neurosci Lett 2019; 702:51-60. [DOI: 10.1016/j.neulet.2018.11.045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
705
|
Vogel S, Schwabe L. Stress, aggression, and the balance of approach and avoidance. Psychoneuroendocrinology 2019; 103:137-146. [PMID: 30685681 DOI: 10.1016/j.psyneuen.2019.01.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 01/17/2019] [Accepted: 01/17/2019] [Indexed: 11/17/2022]
Abstract
Stress is a well-established risk factor for many mental disorders including anxiety disorders or substance abuse. A hallmark of these disorders is an imbalance between behavioral approach and avoidance in situations with approach-avoidance conflicts and unclear outcomes. However, if and how stress affects human behavior in approach-avoidance conflicts is largely unknown. To investigate the effects of stress on approach-avoidance behavior, 80 participants underwent a stress or control manipulation before performing an approach-avoidance conflict task. Stress markedly increased behavioral inhibition when threats were distant and accelerated responses when threats were close; suggesting that stress amplifies the importance of threat distance. However, participants high in trait aggression showed increased approach behavior, particularly when stressed. These findings indicate that stress generally leads to enhanced avoidance, but induces approach in individuals prone to aggression, with important implications for stress-related psychopathologies.
Collapse
Affiliation(s)
- Susanne Vogel
- Department of Cognitive Psychology, Institute of Psychology, University of Hamburg, Von-Melle-Park 5, 20146, Hamburg, Germany.
| | - Lars Schwabe
- Department of Cognitive Psychology, Institute of Psychology, University of Hamburg, Von-Melle-Park 5, 20146, Hamburg, Germany.
| |
Collapse
|
706
|
Nikkar E, Ghoshooni H, Hadipour MM, Sahraei H. Effect of Nitric Oxide on Basolateral Amygdala on Persistence of Anxiety and Depression in Stressed Male Rats. Basic Clin Neurosci 2019; 10:13-22. [PMID: 31031890 PMCID: PMC6484182 DOI: 10.32598/bcn.9.10.100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 03/10/2018] [Accepted: 07/17/2018] [Indexed: 01/22/2023] Open
Abstract
Introduction: The current study aimed at investigating the role of Nitric Oxide (NO) in the maintenance of anxiety and depression induced by stress in male Wistar rats using intra-Basolateral Amygdala (BLA) injection of NO precursor, L-arginine, Nitric Oxide Synthase (NOS) inhibitor, and L-NAME. Methods: Two 23-gauge stainless steel cannulas were placed in the BLA, stereotaxically. Seven days later, animals experienced electro foot shock stress based on the following protocol: animals experienced four sessions of stress for 60 minutes in four consecutive days. Five minutes before each stress session, the animals received different doses of L-arginine or L-NAME (1, 5 and, 10 μg/rat) or saline (0.5 μL/rat) intra-BLA. Six days after the stress termination, animals were tested for maintenance of anxiety-like behavior (elevated plus maze; EPM) and eight days after the stress they were examined for depression (forced swimming test; FST). Results: Stress reduced the time and number of open arms and decreased motor activity on EPM. Stress-induced anxiety was inhibited by L-arginine and L-NAME (1, 5, and 10 μg/rat). L-Arginine and L-NAME induced anxiety in non-stressed rats. Stress also increased the immobility time in animals in FST paradigm. Interestingly, both L-arginine and L-NAME, in all doses reduced the stress effect. Conclusion: BLA nitric oxide may play a pivotal role in anxiety and depression induced by stress in rats. Since the effects of both L-arginine and L-NAME were similar, NO might have a modulatory role in the BLA.
Collapse
Affiliation(s)
- Esmaeil Nikkar
- Department of Physiology and Biophysics, School of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Hassan Ghoshooni
- Department of Physiology and Biophysics, School of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran.,Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | | - Hedayat Sahraei
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
707
|
Interventions after acute stress prevent its delayed effects on the amygdala. Neurobiol Stress 2019; 10:100168. [PMID: 31193585 PMCID: PMC6535648 DOI: 10.1016/j.ynstr.2019.100168] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 04/26/2019] [Accepted: 04/29/2019] [Indexed: 11/23/2022] Open
Abstract
Stress is known to elicit contrasting patterns of plasticity in the amygdala and hippocampus. While chronic stress leads to neuronal atrophy in the rodent hippocampus, it has the opposite effect in the basolateral amygdala (BLA). Further, even a single episode of acute stress is known to elicit delayed effects in the amygdala. For example, 2 h of immobilisation stress has been shown to cause a delayed increase in dendritic spine density on BLA principal neurons 10 days later in young rats. This is paralleled by higher anxiety-like behaviour at the same delayed time point. This temporal build-up of morphological and behavioural effects 10 days later, in turn, provides a stress-free time window of intervention after exposure to acute stress. Here, we explore this possibility by specifically testing the efficacy of an anxiolytic drug in reversing the delayed effects of acute immobilisation stress. Oral gavage of diazepam 1 h after immobilisation stress prevented the increase in anxiety-like behaviour on the elevated plus-maze 10 days later. The same post-stress intervention also prevented delayed spinogenesis in the BLA 10 days after acute stress. Surprisingly, gavage of only the vehicle also had a protective effect on both the behavioural and synaptic effects of stress 10 days later. Vehicle gavage was found to trigger a significant rise in corticosterone levels that was comparable to that elicited by acute stress. This suggests that a surge in corticosterone levels, caused by the vehicle gavage 1 h after acute stress, was capable of reversing the delayed enhancing effects of stress on anxiety-like behaviour and BLA synaptic connectivity. These findings are consistent with clinical reports on the protective effects of glucocorticoids against the development of symptoms of post-traumatic stress disorder. Taken together, these results reveal strategies, targeted 1 h after stress, which can prevent the delayed effects of a brief exposure to a severe physical stressor. Acute immobilisation stress increases anxiety and BLA spinogenesis 10 days later. Oral gavage of diazepam 1 h after stress prevents both these delayed effects. Oral gavage of vehicle also has a similar protective effect on anxiety and spines. Vehicle-gavage administration leads to an increase in levels of corticosterone. This post-stress corticosterone surge may have prevented stress-effects 10 days later.
Collapse
|
708
|
Raber J, Arzy S, Bertolus JB, Depue B, Haas HE, Hofmann SG, Kangas M, Kensinger E, Lowry CA, Marusak HA, Minnier J, Mouly AM, Mühlberger A, Norrholm SD, Peltonen K, Pinna G, Rabinak C, Shiban Y, Soreq H, van der Kooij MA, Lowe L, Weingast LT, Yamashita P, Boutros SW. Current understanding of fear learning and memory in humans and animal models and the value of a linguistic approach for analyzing fear learning and memory in humans. Neurosci Biobehav Rev 2019; 105:136-177. [PMID: 30970272 DOI: 10.1016/j.neubiorev.2019.03.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 01/30/2019] [Accepted: 03/18/2019] [Indexed: 01/04/2023]
Abstract
Fear is an emotion that serves as a driving factor in how organisms move through the world. In this review, we discuss the current understandings of the subjective experience of fear and the related biological processes involved in fear learning and memory. We first provide an overview of fear learning and memory in humans and animal models, encompassing the neurocircuitry and molecular mechanisms, the influence of genetic and environmental factors, and how fear learning paradigms have contributed to treatments for fear-related disorders, such as posttraumatic stress disorder. Current treatments as well as novel strategies, such as targeting the perisynaptic environment and use of virtual reality, are addressed. We review research on the subjective experience of fear and the role of autobiographical memory in fear-related disorders. We also discuss the gaps in our understanding of fear learning and memory, and the degree of consensus in the field. Lastly, the development of linguistic tools for assessments and treatment of fear learning and memory disorders is discussed.
Collapse
Affiliation(s)
- Jacob Raber
- Department of Behavioral Neuroscience, ONPRC, Oregon Health & Science University, Portland, OR, USA; Departments of Neurology and Radiation Medicine, and Division of Neuroscience, ONPRC, Oregon Health & Science University, Portland, OR, USA.
| | - Shahar Arzy
- Department of Medical Neurobiology, Hebrew University, Jerusalem 91904, Israel
| | | | - Brendan Depue
- Departments of Psychological and Brain Sciences and Anatomical Sciences and Neurobiology, University of Louisville, Louisville, KY, USA
| | - Haley E Haas
- Department of Psychiatry and Behavioral Science, Emory University School of Medicine, Atlanta, GA, USA
| | - Stefan G Hofmann
- Department of Psychological and Brain Sciences, Boston University, Boston, MA, USA
| | - Maria Kangas
- Department of Psychology, Macquarie University, Sydney, Australia
| | | | - Christopher A Lowry
- Department of Integrative Physiology and Center for Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Hilary A Marusak
- Department of Pharmacy Practice, Wayne State University, Detroit, MI, USA
| | - Jessica Minnier
- School of Public Health, Oregon Health & Science University, Portland, OR, USA
| | - Anne-Marie Mouly
- Lyon Neuroscience Research Center, CNRS-UMR 5292, INSERM U1028, Université Lyon, Lyon, France
| | - Andreas Mühlberger
- Department of Psychology (Clinical Psychology and Psychotherapy), University of Regensburg, Regensburg, Germany; PFH - Private University of Applied Sciences, Department of Psychology (Clinical Psychology and Psychotherapy Research), Göttingen, Germany
| | - Seth Davin Norrholm
- Department of Psychiatry and Behavioral Science, Emory University School of Medicine, Atlanta, GA, USA
| | - Kirsi Peltonen
- Faculty of Social Sciences/Psychology, Tampere University, Tampere, Finland
| | - Graziano Pinna
- The Psychiatric Institute, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
| | - Christine Rabinak
- Department of Pharmacy Practice, Wayne State University, Detroit, MI, USA
| | - Youssef Shiban
- Department of Psychology (Clinical Psychology and Psychotherapy), University of Regensburg, Regensburg, Germany; PFH - Private University of Applied Sciences, Department of Psychology (Clinical Psychology and Psychotherapy Research), Göttingen, Germany
| | - Hermona Soreq
- Department of Biological Chemistry, Edmond and Lily Safra Center of Brain Science and The Institute of Life Sciences, Hebrew University, Jerusalem 91904, Israel
| | - Michael A van der Kooij
- Translational Psychiatry, Department of Psychiatry and Psychotherapy, Universitatsmedizin der Johannes Guttenberg University Medical Center, Mainz, Germany
| | | | - Leah T Weingast
- Department of Psychiatry and Behavioral Science, Emory University School of Medicine, Atlanta, GA, USA
| | - Paula Yamashita
- School of Public Health, Oregon Health & Science University, Portland, OR, USA
| | - Sydney Weber Boutros
- Department of Behavioral Neuroscience, ONPRC, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
709
|
Duman RS, Sanacora G, Krystal JH. Altered Connectivity in Depression: GABA and Glutamate Neurotransmitter Deficits and Reversal by Novel Treatments. Neuron 2019; 102:75-90. [PMID: 30946828 PMCID: PMC6450409 DOI: 10.1016/j.neuron.2019.03.013] [Citation(s) in RCA: 567] [Impact Index Per Article: 94.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 03/04/2019] [Accepted: 03/07/2019] [Indexed: 12/12/2022]
Abstract
The mechanisms underlying the pathophysiology and treatment of depression and stress-related disorders remain unclear, but studies in depressed patients and rodent models are beginning to yield promising insights. These studies demonstrate that depression and chronic stress exposure cause atrophy of neurons in cortical and limbic brain regions implicated in depression, and brain imaging studies demonstrate altered connectivity and network function in the brains of depressed patients. Studies of the neurobiological basis of the these alterations have focused on both the principle, excitatory glutamate neurons, as well as inhibitory GABA interneurons. They demonstrate structural, functional, and neurochemical deficits in both major neuronal types that could lead to degradation of signal integrity in cortical and hippocampal regions. The molecular mechanisms underlying these changes have not been identified but are thought to be related to stress induced excitotoxic effects in combination with elevated adrenal glucocorticoids and inflammatory cytokines as well as other environmental factors. Transcriptomic studies are beginning to demonstrate important sex differences and, together with genomic studies, are starting to reveal mechanistic domains of overlap and uniqueness with regards to risk and pathophysiological mechanisms with schizophrenia and bipolar disorder. These studies also implicate GABA and glutamate dysfunction as well as immunologic mechanisms. While current antidepressants have significant time lag and efficacy limitations, new rapid-acting agents that target the glutamate and GABA systems address these issues and offer superior therapeutic interventions for this widespread and debilitating disorder.
Collapse
Affiliation(s)
- Ronald S Duman
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street, New Haven, CT 06508, USA.
| | - Gerard Sanacora
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street, New Haven, CT 06508, USA
| | - John H Krystal
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street, New Haven, CT 06508, USA
| |
Collapse
|
710
|
Mampay M, Sheridan GK. REST: An epigenetic regulator of neuronal stress responses in the young and ageing brain. Front Neuroendocrinol 2019; 53:100744. [PMID: 31004616 DOI: 10.1016/j.yfrne.2019.04.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 04/03/2019] [Accepted: 04/11/2019] [Indexed: 12/27/2022]
Abstract
The transcriptional repressor REST (Repressor Element-1 Silencing Transcription factor) is a key modulator of the neuronal epigenome and targets genes involved in neuronal differentiation, axonal growth, vesicular transport, ion channel conductance and synaptic plasticity. Whilst its gene expression-modifying properties have been examined extensively in neuronal development, REST's response towards stress-induced neuronal insults has only recently been explored. Overall, REST appears to be an ideal candidate to fine-tune neuronal gene expression following different forms of cellular, neuropathological, psychological and physical stressors. Upregulation of REST is reportedly protective against premature neural stem cell depletion, neuronal hyperexcitability, oxidative stress, neuroendocrine system dysfunction and neuropathology. In contrast, neuronal REST activation has also been linked to neuronal dysfunction and neurodegeneration. Here, we highlight key findings and discrepancies surrounding our current understanding of REST's function in neuronal adaptation to stress and explore its potential role in neuronal stress resilience in the young and ageing brain.
Collapse
Affiliation(s)
- Myrthe Mampay
- Neuroimmunology & Neurotherapeutics Laboratory, School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton BN2 4GJ, UK
| | - Graham K Sheridan
- Neuroimmunology & Neurotherapeutics Laboratory, School of Pharmacy and Biomolecular Sciences, University of Brighton, Brighton BN2 4GJ, UK.
| |
Collapse
|
711
|
Begdache L, Kianmehr H, Sabounchi N, Marszalek A, Dolma N. Principal component regression of academic performance, substance use and sleep quality in relation to risk of anxiety and depression in young adults. Trends Neurosci Educ 2019; 15:29-37. [PMID: 31176469 DOI: 10.1016/j.tine.2019.03.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 03/08/2019] [Accepted: 03/27/2019] [Indexed: 12/14/2022]
Abstract
Adverse lifestyle factors increase risk of anxiety and depression in young adults. Consequently, neurochemical and neuroanatomical alterations ensue, and may initiate a vicious cycle of mental distress, poor lifestyle choices and academic performance. A total of 558 students from different US colleges completed an anonymous survey on academic performance, daytime sleepiness, substance use and mental distress. Low mental distress in college students positively associated with good academic efforts and limited daytime sleepiness. Mild mental distress correlated with borderline work neglect and with a marginal negative association with Grade-point average (GPA). Severe mental distress correlated with excessive daytime sleepiness and poor academic performance. A System Dynamic model was developed to reflect the integration of these variables with mental distress and academic performance. Our results demonstrate that manageable lifestyle factors contribute to mental health in college students, which become potentially cyclic events that may impact academic performance.
Collapse
Affiliation(s)
- Lina Begdache
- Binghamton University, Department of Health and Wellness Studies, USA.
| | - Hamed Kianmehr
- Binghamton University Thomas J. Watson School of Engineering and Applied Science, USA
| | - Nasim Sabounchi
- Binghamton University Thomas J. Watson School of Engineering and Applied Science, USA
| | - Anna Marszalek
- Binghamton University, Department of Biological Sciences, USA
| | - Ngawang Dolma
- Binghamton University, Department of Biological Sciences, USA
| |
Collapse
|
712
|
Ullmann E, Perry SW, Licinio J, Wong ML, Dremencov E, Zavjalov EL, Shevelev OB, Khotskin NV, Koncevaya GV, Khotshkina AS, Moshkin MP, Lapshin MS, Komelkova MV, Feklicheva IV, Tseilikman OB, Cherkasova OP, Bhui KS, Jones E, Kirschbaum C, Bornstein SR, Tseilikman V. From Allostatic Load to Allostatic State-An Endogenous Sympathetic Strategy to Deal With Chronic Anxiety and Stress? Front Behav Neurosci 2019; 13:47. [PMID: 30967764 PMCID: PMC6442703 DOI: 10.3389/fnbeh.2019.00047] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 02/22/2019] [Indexed: 01/10/2023] Open
Abstract
The concepts of allostatic load and overload, i. e., a dramatic increase in the allostatic load that predisposes to disease, have been extensively described in the literature. Here, we show that rats engaging in active offensive response (AOR) behavioral strategies to chronic predator scent stress (PSS) display less anxiety behavior and lower plasma cortisol levels vs. rats engaging in passive defensive response (PDR) behavioral strategies to chronic PSS. In the same chronic PSS paradigm, AOR rats also have higher lactate and lower glutamate levels in amygdala but not in control-region hippocampus vs. PDR rats. The implications of these findings for regulation of allostatic and stress responses, and post-traumatic stress disorder (PTSD) are discussed.
Collapse
Affiliation(s)
- Enrico Ullmann
- Department of Medicine, Carl Gustav Carus, Technical University of Dresden, Dresden, Germany.,Department of Child and Adolescent Psychiatry, Psychotherapy, and Psychosomatics, University of Leipzig, Leipzig, Germany.,School of Medical Biology, South Ural State University, Chelyabinsk, Russia
| | - Seth W Perry
- College of Medicine, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Julio Licinio
- College of Medicine, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Ma-Li Wong
- College of Medicine, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Eliyahu Dremencov
- School of Medical Biology, South Ural State University, Chelyabinsk, Russia.,Institute of Molecular Physiology and Genetics, Centre for Biosciences, Slovak Academy of Sciences, Bratislava, Slovakia.,Biomedical Research Center, Institute of Experimental Endocrinology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Evgenii L Zavjalov
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Science (RAS), Novosibirsk, Russia
| | - Oleg B Shevelev
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Science (RAS), Novosibirsk, Russia
| | - Nikita V Khotskin
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Science (RAS), Novosibirsk, Russia
| | - Galina V Koncevaya
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Science (RAS), Novosibirsk, Russia
| | - Anna S Khotshkina
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Science (RAS), Novosibirsk, Russia
| | - Mikhail P Moshkin
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Science (RAS), Novosibirsk, Russia
| | - Maxim S Lapshin
- School of Medical Biology, South Ural State University, Chelyabinsk, Russia
| | - Maria V Komelkova
- School of Medical Biology, South Ural State University, Chelyabinsk, Russia
| | - Inna V Feklicheva
- School of Medical Biology, South Ural State University, Chelyabinsk, Russia
| | - Olga B Tseilikman
- School of Medical Biology, South Ural State University, Chelyabinsk, Russia
| | - Olga P Cherkasova
- Biophysics Laboratory, Institute of Laser Physics, Siberian Branch of the Russian Academy of Science, Novosibirsk, Russia
| | - Kamaldeep S Bhui
- Centre for Psychiatry, Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, United Kingdom
| | - Edgar Jones
- Institute of Psychiatry Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Clemens Kirschbaum
- Department of Psychology, Biopsychology, Technical University of Dresden, Dresden, Germany
| | - Stefan R Bornstein
- Department of Medicine, Carl Gustav Carus, Technical University of Dresden, Dresden, Germany.,Faculty of Life Sciences & Medicine, Endocrinology and Diabetes, Kings College London, London, United Kingdom
| | - Vadim Tseilikman
- School of Medical Biology, South Ural State University, Chelyabinsk, Russia
| |
Collapse
|
713
|
Kharabian Masouleh S, Eickhoff SB, Hoffstaedter F, Genon S. Empirical examination of the replicability of associations between brain structure and psychological variables. eLife 2019; 8:e43464. [PMID: 30864950 PMCID: PMC6483597 DOI: 10.7554/elife.43464] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 03/08/2019] [Indexed: 02/01/2023] Open
Abstract
Linking interindividual differences in psychological phenotype to variations in brain structure is an old dream for psychology and a crucial question for cognitive neurosciences. Yet, replicability of the previously-reported 'structural brain behavior' (SBB)-associations has been questioned, recently. Here, we conducted an empirical investigation, assessing replicability of SBB among heathy adults. For a wide range of psychological measures, the replicability of associations with gray matter volume was assessed. Our results revealed that among healthy individuals 1) finding an association between performance at standard psychological tests and brain morphology is relatively unlikely 2) significant associations, found using an exploratory approach, have overestimated effect sizes and 3) can hardly be replicated in an independent sample. After considering factors such as sample size and comparing our findings with more replicable SBB-associations in a clinical cohort and replicable associations between brain structure and non-psychological phenotype, we discuss the potential causes and consequences of these findings.
Collapse
Affiliation(s)
- Shahrzad Kharabian Masouleh
- Institute of Neuroscience and Medicine (INM-7: Brain and Behaviour)Research Centre JülichJülichGermany
- Institute of Systems NeuroscienceHeinrich Heine University DüsseldorfDüsseldorfGermany
| | - Simon B Eickhoff
- Institute of Neuroscience and Medicine (INM-7: Brain and Behaviour)Research Centre JülichJülichGermany
- Institute of Systems NeuroscienceHeinrich Heine University DüsseldorfDüsseldorfGermany
| | - Felix Hoffstaedter
- Institute of Neuroscience and Medicine (INM-7: Brain and Behaviour)Research Centre JülichJülichGermany
- Institute of Systems NeuroscienceHeinrich Heine University DüsseldorfDüsseldorfGermany
| | - Sarah Genon
- Institute of Neuroscience and Medicine (INM-7: Brain and Behaviour)Research Centre JülichJülichGermany
| | - Alzheimer's Disease Neuroimaging Initiative
- Institute of Neuroscience and Medicine (INM-7: Brain and Behaviour)Research Centre JülichJülichGermany
- Institute of Systems NeuroscienceHeinrich Heine University DüsseldorfDüsseldorfGermany
| |
Collapse
|
714
|
Rincón-Cortés M, Herman JP, Lupien S, Maguire J, Shansky RM. Stress: Influence of sex, reproductive status and gender. Neurobiol Stress 2019; 10:100155. [PMID: 30949564 PMCID: PMC6430637 DOI: 10.1016/j.ynstr.2019.100155] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 02/25/2019] [Accepted: 03/05/2019] [Indexed: 11/17/2022] Open
Abstract
Emerging evidence from the preclinical and human research suggests sex differences in response to different types of stress exposure, and that developmental timing, reproductive status, and biological sex are important factors influencing the degree of HPA activation/function. Here we review data regarding: i) sex differences in behavioral and neural responses to uncontrollable and controllable stressors; ii) distinct trajectories of behavioral development and HPA-axis function in male and female rats following adolescent stress exposure; iii) normative changes in behavior and dopamine function in early postpartum rats; iv) aberrant HPA-axis function and its link to abnormal behaviors in two independent, preclinical mouse models of postpartum depression; and, v) data indicating that gender, in addition to sex, is an important determinant of stress reactivity in humans. Based on these findings, we conclude it will be important for future studies to investigate the short and long-term effects of a wide variety of stressors, how these effects may differ according to developmental timing and in relation to gonadal function, the relationship between aberrant HPA-axis activity during the postpartum and mood disorders, and influences of both sex and gender on stress reactivity in humans.
Collapse
Affiliation(s)
- Millie Rincón-Cortés
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
- Corresponding author. Department of Neuroscience, A210 Langley Hall, University of Pittsburgh, Pittsburgh, PA, 15260, USA.
| | - James P. Herman
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, USA
| | - Sonia Lupien
- Department of Psychiatry, Université de Montréal, Montréal, Québec, Canada
| | - Jamie Maguire
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | | |
Collapse
|
715
|
Harris BN, Hohman ZP, Campbell CM, King KS, Tucker CA. FAAH genotype, CRFR1 genotype, and cortisol interact to predict anxiety in an aging, rural Hispanic population: A Project FRONTIER study. Neurobiol Stress 2019; 10:100154. [PMID: 30949563 PMCID: PMC6430712 DOI: 10.1016/j.ynstr.2019.100154] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 01/21/2019] [Accepted: 03/04/2019] [Indexed: 12/18/2022] Open
Abstract
The neurophysiological underpinnings involved in susceptibility to and maintenance of anxiety are not entirely known. However, two stress-responsive systems, the hypothalamic-pituitary-adrenal axis and the endocannabinoid system, may interact in anxiety. Here, we examine the relationship between FAAH genotype, CRFR1 genotype, baseline cortisol, and state anxiety in a rural adult population using data from Project FRONTIER. We predicted that FAAH A (AA and AC vs CC; rs324420) and three CRFR1 SNP minor alleles (rs7209436 C→ T [minor allele]; rs110402, G → A [minor]; and rs242924 G→ T [minor]), would interact to predict low baseline cortisol and low state anxiety scores. We found partial support for our prediction. In CRFR1 minor carriers, the FAAH AA or AC (vs. CC) genotype was associated with higher cortisol and with lower anxiety. In CRFR1 non-minors, those with FAAH AA or AC (vs. CC) showed decreased cortisol and higher anxiety. These results suggest that FAAH CC genotype only conveys risk for anxiety in individuals who are also carriers of the CRFR1 minor combination. FAAH genotype was significantly associated with baseline cortisol but was not independently associated with anxiety. Contrary to our predictions, baseline cortisol was negatively associated with anxiety. Lastly, we did not find any independent relationships between any of our SNPs and baseline cortisol or anxiety. These data suggest FAAH and cortisol interact to predict state anxiety, but that the relationship depends on CRFR1 genotype. The Project FRONTIER dataset is supported by Texas Tech University Health Sciences Center Garrison Institute on Aging.
Collapse
Affiliation(s)
- Breanna N Harris
- Department of Biological Sciences, Texas Tech University, Lubbock, TX, USA
| | - Zachary P Hohman
- Department of Psychological Sciences, Texas Tech University, Lubbock, TX, USA
| | - Callie M Campbell
- Department of Biological Sciences, Texas Tech University, Lubbock, TX, USA
| | - Kaleb S King
- Department of Biological Sciences, Texas Tech University, Lubbock, TX, USA
| | - Cody A Tucker
- Department of Biological Sciences, Texas Tech University, Lubbock, TX, USA
| | | |
Collapse
|
716
|
Ewing-Cobbs L, DeMaster D, Watson CG, Prasad MR, Cox CS, Kramer LA, Fischer JT, Duque G, Swank PR. Post-Traumatic Stress Symptoms after Pediatric Injury: Relation to Pre-Frontal Limbic Circuitry. J Neurotrauma 2019; 36:1738-1751. [PMID: 30672379 DOI: 10.1089/neu.2018.6071] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Pre-frontal limbic circuitry is vulnerable to effects of stress and injury. We examined microstructure of pre-frontal limbic circuitry after traumatic brain injury (TBI) or extracranial injury (EI) and its relation to post-traumatic stress symptoms (PTSS). Participants aged 8 to 15 years who sustained mild to severe TBI (n = 53) or EI (n = 26) in motor vehicle incidents were compared with healthy children (n = 38) in a prospective longitudinal study. At the seven-week follow-up, diffusion tensor imaging was obtained in all groups; injured children completed PTSS ratings using a validated scale. Using probabilistic diffusion tensor tractography, pathways were seeded from bilateral amygdalae and hippocampi to estimate the trajectory of white matter connecting them to each other and to targeted pre-frontal cortical (PFC) regions. Microstructure was estimated using fractional anisotropy (FA) in white matter and mean diffusivity (MD) in gray matter. Pre-frontal limbic microstructure was similar across groups, except for reduced FA in the right hippocampus to orbital PFC pathway in the injured versus healthy group. We examined microstructure of components of pre-frontal limbic circuitry with concurrently obtained PTSS cluster scores in the injured children. Neither microstructure nor PTSS scores differed significantly in the TBI and EI groups. Across PTSS factors, specific symptom clusters were related positively to higher FA and MD. Higher hyperarousal, avoidance, and re-experiencing symptoms were associated with higher FA in amygdala to pre-frontal and hippocampus to amygdala pathways. Higher hippocampal MD had a central role in hyperarousal and emotional numbing symptoms. Age moderated the relation of white and gray matter microstructure with hyperarousal scores. Our findings are consistent with models of traumatic stress that implicate disrupted top-down PFC and hippocampal moderation of overreactive subcortical threat arousal systems. Alterations in limbic pre-frontal circuitry and PTSS place children with either brain or body injuries at elevated risk for both current and future psychological health problems.
Collapse
Affiliation(s)
- Linda Ewing-Cobbs
- 1 Children's Learning Institute and Department of Pediatrics, University of Texas Health Science Center at Houston, Houston, Texas
| | - Dana DeMaster
- 1 Children's Learning Institute and Department of Pediatrics, University of Texas Health Science Center at Houston, Houston, Texas
| | - Christopher G Watson
- 1 Children's Learning Institute and Department of Pediatrics, University of Texas Health Science Center at Houston, Houston, Texas
| | - Mary R Prasad
- 1 Children's Learning Institute and Department of Pediatrics, University of Texas Health Science Center at Houston, Houston, Texas
| | - Charles S Cox
- 2 Department of Pediatric Surgery, University of Texas Health Science Center at Houston, Houston, Texas
| | - Larry A Kramer
- 4 Department of Interventional Radiology, University of Texas Health Science Center at Houston, Houston, Texas
| | - Jesse T Fischer
- 5 Department of Psychology, University of Houston, Houston, Texas
| | - Gerardo Duque
- 1 Children's Learning Institute and Department of Pediatrics, University of Texas Health Science Center at Houston, Houston, Texas
| | - Paul R Swank
- 3 School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas
| |
Collapse
|
717
|
Neonatal Dexamethasone Treatment Suppresses Hippocampal Estrogen Receptor α Expression in Adolescent Female Rats. Mol Neurobiol 2019; 56:2224-2233. [DOI: 10.1007/s12035-018-1214-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 06/27/2018] [Indexed: 11/24/2022]
|
718
|
Riem MME, van IJzendoorn MH, Bakermans-Kranenburg MJ. Hippocampal volume modulates salivary oxytocin level increases after intranasal oxytocin administration. Psychoneuroendocrinology 2019; 101:182-185. [PMID: 30469085 DOI: 10.1016/j.psyneuen.2018.11.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 10/10/2018] [Accepted: 11/09/2018] [Indexed: 11/26/2022]
Abstract
Adverse childhood experiences have been shown to affect sensitivity to intranasal oxytocin administration, but the neural mechanisms underlying this altered sensitivity are unclear. The aim of the current study was to examine whether hippocampal abnormalities underlie the effects of adversity on the response to oxytocin administration. In a sample of healthy women (N = 54, age M = 19.63), we examined 1) the association between hippocampal volume and experiences of emotional maltreatment and 2) whether hippocampal volume reductions influence the effect of intranasal oxytocin administration on salivary oxytocin levels. There was no association between hippocampal volume and experiences of emotional maltreatment in the current study. However, we found that larger hippocampal volume was related to a stronger increase in salivary oxytocin level after intranasal oxytocin administration. The hippocampus may be a neural substrate underlying individual differences in sensitivity to oxytocin administration.
Collapse
Affiliation(s)
- Madelon M E Riem
- Department of Medical and Clinical Psychology, Tilburg University, Tilburg, the Netherlands.
| | - Marinus H van IJzendoorn
- Department of Psychology, Education and Child Studies, Erasmus University, Rotterdam, the Netherlands; Primary Care Unit School of Clinical Medicine, University of Cambridge, UK
| | - Marian J Bakermans-Kranenburg
- Primary Care Unit School of Clinical Medicine, University of Cambridge, UK; Clinical Child and Family Studies, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
719
|
The Role of Stressful Parenting and Mineralocorticoid Receptor Haplotypes on Social Development During Adolescence and Young Adulthood. J Youth Adolesc 2019; 48:1082-1099. [PMID: 30805852 PMCID: PMC6525128 DOI: 10.1007/s10964-019-00988-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 01/31/2019] [Indexed: 12/04/2022]
Abstract
The development of social behavior could be affected by stressful parenting. The mineralocorticoid receptor, one of the two main receptors for the stress hormone cortisol, plays a vital role in adequate responses to stress. Therefore, the effects of stressful parenting on social development (i.e., empathic concern, perspective taking and prosocial behavior) may be moderated by functional genetic variation in mineralocorticoid receptor haplotypes (a combination of alleles). A group of 343 adolescents (44.3% females) was followed from the age of 13 until 24 years. Growth curve analyses showed lower levels of prosocial behaviors and a slower increase in empathic concern and perspective taking in adolescents who reported more stressful parenting. In contrast, relatively higher levels of prosocial behavior, empathic concern and perspective taking were present in combination with stress resilient mineralocorticoid receptor haplotypes. Despite sex differences in social development with earlier social development for girls, no consistent sex differences were found with regard to mineralocorticoid receptor haplotypes. The current study showed that genetic variation in mineralocorticoid receptor impacts the social development during adolescence and young adulthood.
Collapse
|
720
|
Bolton JL, Short AK, Simeone KA, Daglian J, Baram TZ. Programming of Stress-Sensitive Neurons and Circuits by Early-Life Experiences. Front Behav Neurosci 2019; 13:30. [PMID: 30833892 PMCID: PMC6387907 DOI: 10.3389/fnbeh.2019.00030] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 02/04/2019] [Indexed: 12/22/2022] Open
Abstract
Early-life experiences influence brain structure and function long-term, contributing to resilience or vulnerability to stress and stress-related disorders. Therefore, understanding the mechanisms by which early-life experiences program specific brain cells and circuits to shape life-long cognitive and emotional functions is crucial. We identify the population of corticotropin-releasing hormone (CRH)-expressing neurons in the hypothalamic paraventricular nucleus (PVN) as a key, early target of early-life experiences. Adverse experiences increase excitatory neurotransmission onto PVN CRH cells, whereas optimal experiences, such as augmented and predictable maternal care, reduce the number and function of glutamatergic inputs onto this cell population. Altered synaptic neurotransmission is sufficient to initiate large-scale, enduring epigenetic re-programming within CRH-expressing neurons, associated with stress resilience and additional cognitive and emotional outcomes. Thus, the mechanisms by which early-life experiences influence the brain provide tractable targets for intervention.
Collapse
Affiliation(s)
- Jessica L Bolton
- Departments of Pediatrics, Anatomy/Neurobiology, Neurology, University of California, Irvine, Irvine, CA, United States
| | - Annabel Katherine Short
- Departments of Pediatrics, Anatomy/Neurobiology, Neurology, University of California, Irvine, Irvine, CA, United States
| | - Kristina A Simeone
- Departments of Pediatrics, Anatomy/Neurobiology, Neurology, University of California, Irvine, Irvine, CA, United States
| | - Jennifer Daglian
- Departments of Pediatrics, Anatomy/Neurobiology, Neurology, University of California, Irvine, Irvine, CA, United States
| | - Tallie Z Baram
- Departments of Pediatrics, Anatomy/Neurobiology, Neurology, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
721
|
Exploring the involvement of Tac2 in the mouse hippocampal stress response through gene networking. Gene 2019; 696:176-185. [PMID: 30769143 DOI: 10.1016/j.gene.2019.02.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 01/05/2019] [Accepted: 02/01/2019] [Indexed: 01/15/2023]
Abstract
Tachykinin 2 (Tac2) is expressed in a number of areas throughout the brain, including the hippocampus. However, knowledge about its function has been only well explored in the hypothalamus in the context of reproductive health. In this study, we identified and validated increased hippocampal Tac2 mRNA expression in response to chronic mild stress in mice. Expression quantitative trait locus (eQTL) analysis showed Tac2 is cis-regulated in the hippocampus. Using a systems genetics approach, we constructed a Tac2 co-expression network to better understand the relationship between Tac2 and the hippocampal stress response. Our network identified 69 total genes associated with Tac2, several of which encode major neuropeptides involved in hippocampal stress signaling as well as critical genes for producing neural plasticity, indicating that Tac2 is involved in these processes. Pathway analysis for the member of Tac2 gene network revealed a strong connection between Tac2 and neuroactive ligand-receptor interaction, calcium signaling pathway, as well as cardiac muscle contraction. In addition, we also identified 46 stress-related phenotypes, specifically fear conditioning response, that were significantly correlated with Tac2 expression. Our results provide evidence for Tac2 as a strong candidate gene who likely plays a role in hippocampal stress processing and neural plasticity.
Collapse
|
722
|
Kalvas LB. The Life Course Health Development Model: A theoretical research framework for paediatric delirium. J Clin Nurs 2019; 28:2351-2360. [PMID: 30653772 DOI: 10.1111/jocn.14776] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 12/05/2018] [Accepted: 01/07/2019] [Indexed: 12/31/2022]
Abstract
AIMS AND OBJECTIVES To create a framework for future research through application and critique of the Life Course Health Development Model to the phenomenon of paediatric delirium. BACKGROUND Delirium in the paediatric intensive care unit is associated with increased duration of mechanical ventilation, length of stay and mortality. Nurses are uniquely positioned at the bedside to identify, prevent and treat delirium. An understanding of the potential long-term consequences of paediatric delirium is necessary to provide impetus for nursing research and practice change. The Life Course Health Development Model is a valuable tool when considering the multiple mechanisms and processes through which the experience of delirium could affect a child's life trajectory. DESIGN Critical review of the literature through application and critique of the Life Course Health Development Model in the context of paediatric delirium. Gaps in the current understanding of paediatric delirium, as well as future directions for research and practice, are discussed. METHODS The seven core principles of the model are considered in the context of paediatric delirium. Each of the principles has the potential to further understanding of paediatric delirium and identify areas for future inquiry. This discussion leads to a critique of the ability of the model to lead future research and practice change. CONCLUSIONS The Life Course Health Development Model depicts a process in which the acute and severe stress of critical illness leads to maladaptive neurologic changes that contribute to the development of delirium and impair a child's life trajectory. RELEVANCE TO CLINICAL PRACTICE By emphasising the potential lifelong consequences for critically ill children who experience delirium, this application of the Life Course Health Development Model will stimulate discussion, research and practice change among paediatric clinicians and researchers.
Collapse
|
723
|
Zhang JY, Liu TH, He Y, Pan HQ, Zhang WH, Yin XP, Tian XL, Li BM, Wang XD, Holmes A, Yuan TF, Pan BX. Chronic Stress Remodels Synapses in an Amygdala Circuit-Specific Manner. Biol Psychiatry 2019; 85:189-201. [PMID: 30060908 PMCID: PMC6747699 DOI: 10.1016/j.biopsych.2018.06.019] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 06/20/2018] [Accepted: 06/22/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND Chronic stress exposure increases the risk of developing various neuropsychiatric illnesses. The behavioral sequelae of stress correlate with dendritic hypertrophy and glutamate-related synaptic remodeling at basolateral amygdala projection neurons (BLA PNs). Yet, though BLA PNs are functionally heterogeneous with diverse corticolimbic targets, it remains unclear whether stress differentially impacts specific output circuits. METHODS Confocal imaging was used to reconstruct the morphology of mouse BLA PNs with the aid of retrograde tracing and biocytin staining. The synaptic activity in these neurons was measured with in vitro electrophysiology, and anxiety-like behavior of the mice was assessed with the elevated plus maze and open field test. RESULTS Chronic restraint stress (CRS) produced dendritic hypertrophy across mouse BLA PNs, regardless of whether they did (BLA→dorsomedial prefrontal cortex [dmPFC]) or did not (BLA↛dmPFC) target dmPFC. However, CRS increased the size of dendritic spine heads and the number of mature, mushroom-shaped spines only in BLA↛dmPFC PNs, sparing neighboring BLA→dmPFC PNs. Moreover, the excitatory glutamatergic transmission was also selectively increased in BLA↛dmPFC PNs, and this effect correlated with CRS-induced increases in anxiety-like behavior. Segregating BLA↛dmPFC PNs based on their targeting of ventral hippocampus (BLA→ventral hippocampus) or nucleus accumbens (BLA→nucleus accumbens) revealed that CRS increased spine density and glutamatergic signaling in BLA→ventral hippocampus PNs in a manner that correlated with anxiety-like behavior. CONCLUSIONS Chronic stress caused BLA PN neuronal remodeling with a previously unrecognized degree of circuit specificity, offering new insight into the pathophysiological basis of depression, anxiety disorders, and other stress-related conditions.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Bing-Xing Pan
- Laboratory of Fear and Anxiety Disorders, Institute of Life Science, Nanchang University, Nanchang, China; Department of Neurology, the 2nd Affiliated Hospital, Nanchang University, Nanchang, China; Human Aging Research Institute, School of Life Science, Nanchang University, Nanchang, China.
| |
Collapse
|
724
|
Xu B, Lian S, Guo JR, Wang JF, Zhang LP, Li SZ, Yang HM. Activation of the MAPK signaling pathway induces upregulation of pro-apoptotic proteins in the hippocampi of cold stressed adolescent mice. Neurosci Lett 2019; 699:97-102. [PMID: 30711527 DOI: 10.1016/j.neulet.2018.12.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 12/17/2018] [Accepted: 12/19/2018] [Indexed: 11/30/2022]
Abstract
Stress induces many non-specific responses in the hippocampus, especially during adolescence. Low environmental temperature is known to induce stress, but its influence on the hippocampus, especially in adolescent mice is not clear. We compared apoptotic-related protein levels and MAPK signaling pathway activation in hippocampal neurons of adolescent mice under low temperature conditions (4 °C for 12 h) with western blotting and immunohistochemistry. Western bolt results demonstrated that the levels of phospho-JNK, phospho-p38, and cleaved-caspase 3 significantly increased, while the ratio of Bcl-XL/Bax decreased, in the cold stress group. The results of immunohistochemistry (IHC) and Nissl staining demonstrated that the protein optical density of caspase 3 increased and Nissl bodies decreased in the cold stress group compared with controls. Thus, we conclude that cold exposure initiates activation of the MAPK signaling pathway and subsequently induces the upregulation of pro-apoptotic proteins in the hippocampi of adolescent mice. Overall our study reveals the relationship between cold stress and apoptosis in adolescent mice.
Collapse
Affiliation(s)
- Bin Xu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, PR China
| | - Shuai Lian
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, PR China
| | - Jing-Ru Guo
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, PR China
| | - Jian-Fa Wang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, PR China
| | - Li-Ping Zhang
- College of Food Science, Heilongjiang Bayi Agricultural University, Daqing, 163319, PR China
| | - Shi-Ze Li
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, PR China.
| | - Huan-Min Yang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, PR China.
| |
Collapse
|
725
|
Beneficial Effects of Physical Activity and Crocin Against Adolescent Stress Induced Anxiety or Depressive-Like Symptoms and Dendritic Morphology Remodeling in Prefrontal Cortex in Adult Male Rats. Neurochem Res 2019; 44:917-929. [DOI: 10.1007/s11064-019-02727-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 01/09/2019] [Indexed: 01/16/2023]
|
726
|
Childhood trauma and insulin resistance in patients suffering from depressive disorders. Exp Neurol 2019; 315:15-20. [PMID: 30639184 DOI: 10.1016/j.expneurol.2019.01.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 12/18/2018] [Accepted: 01/09/2019] [Indexed: 01/06/2023]
Abstract
OBJECTIVE Insulin resistance (IR) is a metabolic dysfunction often co-morbid with major depressive disorder (MDD). The paths to development of MDD remain largely unspecified, highlighting a need for identification of risk factors. Here, we tested whether specific subscales of childhood trauma as well as family history of type-2 diabetes (Fam-Hx-Dm2) are risk factors for development of metabolic dysfunction and severity of depressive symptoms. RESEARCH DESIGN AND METHODS We used a sample of 45 adults suffering from MDD that was well-characterized for insulin resistance and sensitivity as assessed by measures of fasting plasma glucose (FPG) plasma insulin (FPI) levels, body mass index (BMI), weight, homeostasis model assessment of insulin sensitivity (HOMA), Matsuda index as well as both glucose and insulin responses to oral glucose challenges. Severity of depressive symptoms was assessed with the Hamilton Depression Rating Scale (HDRS-21). Physical, sexual and emotional abuse as well as physical and emotional neglect were assessed with the Childhood Trauma Questionnaire. First- or second-degree relatives with type-2 diabetes defined fam-Hx-DM2. RESULTS Individuals reporting higher rates of emotional abuse were more likely to have greater IR as showed by elevated FPI levels and HOMA. No association was found with any of the other subscales of childhood trauma (e.g., physical abuse). Similarly, Fam-Hx-DM2 was associated with greater degree of IR as shown by elevated FPI, HOMA, but also FPG, weight and BMI. Moreover, we report a relationship and interaction between Fam-Hx-DM2 and emotional abuse on severity of depressive symptoms. Specifically, emotional abuse and Fam-HX-DM2 predicted severity of depressive symptoms at HDRS-21. Also, severity of depressive symptoms was greater with higher reported rates of emotional abuse but only in patients with negative Fam-Hx-Dm2. Individuals reporting higher emotional abuse and negative Fam-Hx-Dm2 also showed higher FPG levels. Conversely, individuals reporting higher emotional abuse and positive Fam-Hx-Dm2 showed higher FPI levels. This data suggest that Fam-Hx-Dm2 may define two different metabolic endophenotypes. CONCLUSIONS Our findings suggest that Fam-HX-DM2 and emotional abuse represent separate risk factors for developing metabolic dysfunction (i.e.: IR) in patients suffering from MDD, and that the effects of emotional abuse on psychiatric illness may depend upon the personal characteristics, including Fam-Hx-DM2.
Collapse
|
727
|
Watson CG, DeMaster D, Ewing-Cobbs L. Graph theory analysis of DTI tractography in children with traumatic injury. Neuroimage Clin 2019; 21:101673. [PMID: 30660661 PMCID: PMC6412099 DOI: 10.1016/j.nicl.2019.101673] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 12/13/2018] [Accepted: 01/07/2019] [Indexed: 01/02/2023]
Abstract
OBJECTIVE To evaluate brai structural connectivity in children with traumatic injury (TI) following a motor vehicle accident using graph theory analysis of DTI tractography data. METHODS DTI scans were acquired on a 3 T Philips scanner from children aged 8-15 years approximately 2 months post-injury. The TI group consisted of children with traumatic brain injury (TBI; n = 44) or extracranial injury (EI; n = 23). Healthy control children (n = 36) were included as an age-matched comparison group. A graph theory approach was applied to DTI tractography data to investigate injury-related differences in connectivity network characteristics. Group differences in structural connectivity evidenced by graph metrics including efficiency, strength, and modularity were assessed using the multi-threshold permutation correction (MTPC) and network-based statistic (NBS) methods. RESULTS At the global network level, global efficiency and mean network strength were lower, and modularity was higher, in the TBI than in the control group. Similarly, strength was lower and modularity higher when comparing the EI to the control group. At the vertex level, nodal efficiency, vertex strength, and average shortest path length were different between all pairwise comparisons of the three groups. Both nodal efficiency and vertex strength were higher in the control than in the EI group, which in turn were higher than in the TBI group. The opposite between-group relationships were seen with path length. These between-group differences were distributed throughout the brain, in both hemispheres. NBS analysis resulted in a cluster of 22 regions and 21 edges with significantly lower connectivity in the TBI group compared to controls. This cluster predominantly involves the frontal lobe and subcortical gray matter structures in both hemispheres. CONCLUSIONS Graph theory analysis of DTI tractography showed diffuse differences in structural brain network connectivity in children 2 months post-TI. Network differences were consistent with lower network integration and higher segregation in the injured groups compared to healthy controls. Findings suggest that inclusion of trauma-exposed comparison groups in studies of TBI outcome is warranted to better characterize the indirect effect of stress on brain networks.
Collapse
Affiliation(s)
- Christopher G Watson
- Dept. of Pediatrics, Children's Learning Institute, University of Texas Health Science Center at Houston, United States.
| | - Dana DeMaster
- Dept. of Pediatrics, Children's Learning Institute, University of Texas Health Science Center at Houston, United States
| | - Linda Ewing-Cobbs
- Dept. of Pediatrics, Children's Learning Institute, University of Texas Health Science Center at Houston, United States
| |
Collapse
|
728
|
Abstract
For decades, symptoms of depression have been treated primarily with medications that directly target the monoaminergic brain systems, which typically take weeks to exert measurable effects and months to exert remission of symptoms. Low, subanesthetic doses of ( R,S)-ketamine (ketamine) result in the rapid improvement of core depressive symptoms, including mood, anhedonia, and suicidal ideation, occurring within hours following a single administration, with relief from symptoms typically lasting up to a week. The discovery of these actions of ketamine has resulted in a reconceptualization of how depression could be more effectively treated in the future. In this review, we discuss clinical data pertaining to ketamine and other rapid-acting antidepressant drugs, as well as the current state of pharmacological knowledge regarding their mechanism of action. Additionally, we discuss the neurobiological circuits that are engaged by this drug class and that may be targeted by a future generation of medications, for example, hydroxynorketamine; metabotropic glutamate receptor 2/3 antagonists; and N-methyl-d-aspartate, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid, and γ-aminobutyric acid receptor modulators.
Collapse
Affiliation(s)
- Todd D Gould
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA;
- Departments of Pharmacology and Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, USA 20892
| | - Scott M Thompson
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA;
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| |
Collapse
|
729
|
Sex differences and the neurobiology of affective disorders. Neuropsychopharmacology 2019; 44:111-128. [PMID: 30061743 PMCID: PMC6235863 DOI: 10.1038/s41386-018-0148-z] [Citation(s) in RCA: 159] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 05/14/2018] [Accepted: 06/25/2018] [Indexed: 12/11/2022]
Abstract
Observations of the disproportionate incidence of depression in women compared with men have long preceded the recent explosion of interest in sex differences. Nonetheless, the source and implications of this epidemiologic sex difference remain unclear, as does the practical significance of the multitude of sex differences that have been reported in brain structure and function. In this article, we attempt to provide a framework for thinking about how sex and reproductive hormones (particularly estradiol as an example) might contribute to affective illness. After briefly reviewing some observed sex differences in depression, we discuss how sex might alter brain function through hormonal effects (both organizational (programmed) and activational (acute)), sex chromosome effects, and the interaction of sex with the environment. We next review sex differences in the brain at the structural, cellular, and network levels. We then focus on how sex and reproductive hormones regulate systems implicated in the pathophysiology of depression, including neuroplasticity, genetic and neural networks, the stress axis, and immune function. Finally, we suggest several models that might explain a sex-dependent differential regulation of affect and susceptibility to affective illness. As a disclaimer, the studies cited in this review are not intended to be comprehensive but rather serve as examples of the multitude of levels at which sex and reproductive hormones regulate brain structure and function. As such and despite our current ignorance regarding both the ontogeny of affective illness and the impact of sex on that ontogeny, sex differences may provide a lens through which we may better view the mechanisms underlying affective regulation and dysfunction.
Collapse
|
730
|
Sep MSC, Gorter R, van Ast VA, Joëls M, Geuze E. No Time-Dependent Effects of Psychosocial Stress on Fear Contextualization and Generalization: A Randomized-Controlled Study With Healthy Participants. CHRONIC STRESS (THOUSAND OAKS, CALIF.) 2019; 3:2470547019896547. [PMID: 32440603 PMCID: PMC7219903 DOI: 10.1177/2470547019896547] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 12/03/2019] [Indexed: 01/10/2023]
Abstract
The formation of context-dependent fear memories (fear contextualization) can aid the recognition of danger in new, similar, situations. Overgeneralization of fear is often seen as hallmark of anxiety and trauma-related disorders. In this randomized-controlled study, we investigated whether exposure to a psychosocial stressor influences retention of fear contextualization and generalization in a time-dependent manner. The Trier Social Stress Test was used to induce psychosocial stress. Healthy male participants (n = 117) were randomly divided into three experimental groups that were subjected to the acquisition phase of the Fear Generalization Task: (1) without stress, (2) immediately after acute stress, or (3) 2 h after acute stress. In this task, a male with neutral facial expression (conditioned stimuli) was depicted in two different contexts that modulated the conditioned stimuli-unconditioned stimuli (=shock) association (threat, safe). Salivary alpha-amylase and cortisol levels were measured throughout the experiment. After a 24-h delay, context-dependency of fear memory was investigated with an unannounced memory test consisting of the threat and safe contexts alternated with a novel context (the generalization context). Multilevel analyses revealed that participants showed increased fear-potentiated startle responses to the conditioned stimuli in the threat compared to the safe context, at the end of the acquisition phase, indicating adequate fear contextualization. Directly after acquisition, there were no time-dependent effects of psychosocial stress on fear contextualization. Context-dependency of fear memories was retained 24 h later, as fear-potentiated startle responding was modulated by context (threat > safe or novel). At that time, the context-dependency of fear memories was also not influenced by the early or late effects of the endogenous stress response during acquisition. These results with experimental stress deviate in some aspects from those earlier obtained with exogenous hydrocortisone administration, suggesting a distinct role for stress mediators other than cortisol.
Collapse
Affiliation(s)
- Milou S. C. Sep
- Brain Research and Innovation
Centre, Ministry of Defence, Utrecht, the Netherlands
- Department of Translational
Neuroscience, UMC Utrecht Brain Center, Utrecht University, Utrecht, the
Netherlands
| | - Rosalie Gorter
- Brain Research and Innovation
Centre, Ministry of Defence, Utrecht, the Netherlands
| | - Vanessa A. van Ast
- Department of Clinical Psychology,
University
of Amsterdam, Amsterdam, the
Netherlands
| | - Marian Joëls
- Department of Translational
Neuroscience, UMC Utrecht Brain Center, Utrecht University, Utrecht, the
Netherlands
- University of Groningen, University
Medical Center Groningen, Groningen, the Netherlands
| | - Elbert Geuze
- Brain Research and Innovation
Centre, Ministry of Defence, Utrecht, the Netherlands
- Department of Psychiatry, UMC
Utrecht Brain Center, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
731
|
Mitjans M, Seidel J, Begemann M, Bockhop F, Moya-Higueras J, Bansal V, Wesolowski J, Seelbach A, Ibáñez MI, Kovacevic F, Duvar O, Fañanás L, Wolf HU, Ortet G, Zwanzger P, Klein V, Lange I, Tänzer A, Dudeck M, Penke L, van Elst LT, Bittner RA, Schmidmeier R, Freese R, Müller-Isberner R, Wiltfang J, Bliesener T, Bonn S, Poustka L, Müller JL, Arias B, Ehrenreich H. Violent aggression predicted by multiple pre-adult environmental hits. Mol Psychiatry 2019; 24:1549-1564. [PMID: 29795411 PMCID: PMC6756097 DOI: 10.1038/s41380-018-0043-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 02/19/2018] [Accepted: 02/21/2018] [Indexed: 01/26/2023]
Abstract
Early exposure to negative environmental impact shapes individual behavior and potentially contributes to any mental disease. We reported previously that accumulated environmental risk markedly decreases age at schizophrenia onset. Follow-up of matched extreme group individuals (≤1 vs. ≥3 risks) unexpectedly revealed that high-risk subjects had >5 times greater probability of forensic hospitalization. In line with longstanding sociological theories, we hypothesized that risk accumulation before adulthood induces violent aggression and criminal conduct, independent of mental illness. We determined in 6 independent cohorts (4 schizophrenia and 2 general population samples) pre-adult risk exposure, comprising urbanicity, migration, physical and sexual abuse as primary, and cannabis or alcohol as secondary hits. All single hits by themselves were marginally associated with higher violent aggression. Most strikingly, however, their accumulation strongly predicted violent aggression (odds ratio 10.5). An epigenome-wide association scan to detect differential methylation of blood-derived DNA of selected extreme group individuals yielded overall negative results. Conversely, determination in peripheral blood mononuclear cells of histone-deacetylase1 mRNA as 'umbrella mediator' of epigenetic processes revealed an increase in the high-risk group, suggesting lasting epigenetic alterations. Together, we provide sound evidence of a disease-independent unfortunate relationship between well-defined pre-adult environmental hits and violent aggression, calling for more efficient prevention.
Collapse
Affiliation(s)
- Marina Mitjans
- 0000 0001 0668 6902grid.419522.9Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany ,DFG Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany ,grid.469673.9Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain
| | - Jan Seidel
- 0000 0001 0668 6902grid.419522.9Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Martin Begemann
- 0000 0001 0668 6902grid.419522.9Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany ,0000 0001 2364 4210grid.7450.6Department of Psychiatry & Psychotherapy, University of Göttingen, Göttingen, Germany
| | - Fabian Bockhop
- 0000 0001 0668 6902grid.419522.9Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Jorge Moya-Higueras
- grid.469673.9Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain ,0000 0001 2163 1432grid.15043.33Department of Psychology, Faculty of Education, Psychology and Social Work, University of Lleida, Lleida, Spain
| | - Vikas Bansal
- 0000 0001 0668 6902grid.419522.9Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany ,0000 0001 2180 3484grid.13648.38Center for Molecular Neurobiology, Institute of Medical Systems Biology, University Clinic Hamburg-Eppendorf, Hamburg, Germany
| | - Janina Wesolowski
- 0000 0001 0668 6902grid.419522.9Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Anna Seelbach
- 0000 0001 0668 6902grid.419522.9Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Manuel Ignacio Ibáñez
- grid.469673.9Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain ,0000 0001 1957 9153grid.9612.cDepartment of Basic and Clinical Psychology and Psychobiology, Universitat Jaume I, Castelló, Spain
| | - Fatka Kovacevic
- 0000 0001 0668 6902grid.419522.9Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Oguzhan Duvar
- 0000 0001 0668 6902grid.419522.9Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Lourdes Fañanás
- grid.469673.9Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain ,0000 0004 1937 0247grid.5841.8Departament Biologia Evolutiva, Ecologia i Ciències Ambientals, Facultat de Biologia and Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Hannah-Ulrike Wolf
- 0000 0001 0668 6902grid.419522.9Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Generós Ortet
- grid.469673.9Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain ,0000 0001 1957 9153grid.9612.cDepartment of Basic and Clinical Psychology and Psychobiology, Universitat Jaume I, Castelló, Spain
| | - Peter Zwanzger
- KBO-Inn-Salzach-Klinikum, Gabersee, Wasserburg am Inn Germany
| | - Verena Klein
- KBO-Isar-Amper-Klinikum, Taufkirchen (Vils), Germany
| | - Ina Lange
- Competence Center for Forensic Psychiatry, Lower Saxony, MRV Moringen Germany
| | - Andreas Tänzer
- 0000 0000 9597 1037grid.412811.fDepartment of Forensic Psychiatry & Psychotherapy, KRH, Wunstorf, Germany
| | - Manuela Dudeck
- 0000 0004 1936 9748grid.6582.9Forensic Psychiatry and Psychotherapy, University of Ulm, Ulm, Germany
| | - Lars Penke
- 0000 0001 2364 4210grid.7450.6Institute of Psychology, University of Göttingen, Göttingen, Germany
| | - Ludger Tebartz van Elst
- grid.5963.9Department of Psychiatry & Psychotherapy, University of Freiburg, Freiburg, Germany
| | - Robert A. Bittner
- 0000 0004 1936 9721grid.7839.5Department of Psychiatry & Psychotherapy, University of Frankfurt, Frankfurt, Germany
| | | | | | | | - Jens Wiltfang
- 0000 0001 2364 4210grid.7450.6Department of Psychiatry & Psychotherapy, University of Göttingen, Göttingen, Germany
| | - Thomas Bliesener
- 0000 0000 8700 8822grid.462495.8Criminological Research Institute of Lower Saxony, Hannover, Germany
| | - Stefan Bonn
- DFG Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany ,0000 0001 2180 3484grid.13648.38Center for Molecular Neurobiology, Institute of Medical Systems Biology, University Clinic Hamburg-Eppendorf, Hamburg, Germany
| | - Luise Poustka
- 0000 0001 2364 4210grid.7450.6Department of Child and Adolescent Psychiatry & Psychotherapy, University of Göttingen, Göttingen, Germany
| | - Jürgen L. Müller
- 0000 0001 2364 4210grid.7450.6Department of Psychiatry & Psychotherapy, University of Göttingen, Göttingen, Germany ,Asklepios Hospital for Forensic Psychiatry & Psychotherapy, Göttingen, Germany
| | - Bárbara Arias
- Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain. .,Departament Biologia Evolutiva, Ecologia i Ciències Ambientals, Facultat de Biologia and Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Spain.
| | - Hannelore Ehrenreich
- Clinical Neuroscience, Max Planck Institute of Experimental Medicine, Göttingen, Germany. .,DFG Research Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany.
| |
Collapse
|
732
|
Development of the MAM model of schizophrenia in mice: Sex similarities and differences of hippocampal and prefrontal cortical function. Neuropharmacology 2019; 144:193-207. [DOI: 10.1016/j.neuropharm.2018.10.026] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 10/06/2018] [Accepted: 10/19/2018] [Indexed: 12/31/2022]
|
733
|
Thomas N, Gurvich C, Kulkarni J. Borderline personality disorder, trauma, and the hypothalamus-pituitary-adrenal axis. Neuropsychiatr Dis Treat 2019; 15:2601-2612. [PMID: 31564884 PMCID: PMC6743631 DOI: 10.2147/ndt.s198804] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 08/26/2019] [Indexed: 12/14/2022] Open
Abstract
Borderline personality disorder (BPD) is a complex psychiatric illness for which treatment poses a significant challenge due to limited effective pharmacologic treatments, and under-resourced psychological interventions. BPD is one of the most stigmatized conditions in psychiatry today, but can be understood as a modifiable, neurodevelopmental disorder that arises from maladaptive responses to trauma and stress. Stress susceptibility and reactivity in BPD is thought to mediate both the development and maintenance of BPD symptomatology, with trauma exposure considered an early life risk factor of development, and acute stress moderating symptom trajectory. An altered stress response has been characterized in BPD at the structural, neural, and neurobiological level, and is believed to underlie the maladaptive behavioral and cognitive symptomatology presented in BPD. The endocrine hypothalamus-pituitary-adrenal (HPA) axis represents a key stress response system, and growing evidence suggests it is dysfunctional in the BPD patient population. This theoretical review examines BPD in the context of a neurodevelopmental stress-related disorder, providing an overview of measurements of stress with a focus on HPA-axis measurement. Potential confounding factors associated with measurement of the HPA system are discussed, including sex and sex hormones, genetic factors, and the influence of sample collection methods. HPA-axis dysfunction in BPD largely mirrors findings demonstrated in post-traumatic stress disorder and may represent a valuable neuroendocrine target for diagnostic or treatment response biomarkers, or for which novel treatments can be investigated.
Collapse
Affiliation(s)
- Natalie Thomas
- Central Clinical School , Monash Alfred Psychiatry Research Centre, Monash University, Melbourne, Victoria, Australia
| | - Caroline Gurvich
- Central Clinical School , Monash Alfred Psychiatry Research Centre, Monash University, Melbourne, Victoria, Australia
| | - Jayashri Kulkarni
- Central Clinical School , Monash Alfred Psychiatry Research Centre, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
734
|
Mulkey SB, du Plessis AJ. Autonomic nervous system development and its impact on neuropsychiatric outcome. Pediatr Res 2019; 85:120-126. [PMID: 30166644 PMCID: PMC6353676 DOI: 10.1038/s41390-018-0155-0] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 07/27/2018] [Accepted: 08/01/2018] [Indexed: 12/22/2022]
Abstract
The central autonomic nervous system (ANS) is essential for maintaining cardiovascular and respiratory homeostasis in the newborn and has a critical role in supporting higher cortical functions. At birth, the central ANS is maturing and is vulnerable to adverse environmental and physiologic influences. Critical connections are formed early in development between the ANS and limbic system to integrate psychological and body responses. The Polyvagal Theory, developed by Stephen Porges, describes how modulation of the autonomic vagal impulse controls social responses and that a broad range of neuropsychiatric disorders may be due to impaired vagal balance, with either deficient vagal tone or excessive vagal reactivity. Under additional circumstances of prematurity, growth restriction, and environmental stress in the fetus and newborn, the immature ANS may undergo "dysmaturation". Maternal stress and health as well as the intrauterine environment are also quite important and have been implicated in causing ANS changes in the infant and neuropsychiatric diseases in children. This review will cover the aspects of ANS development and maturation that have been associated with neuropsychiatric disorders in children.
Collapse
Affiliation(s)
- Sarah B. Mulkey
- Assistant Professor, Department of Pediatrics and Neurology, George Washington University School of Medicine and Health Sciences, Fetal-Neonatal Neurologist, Division of Fetal and Transitional Medicine, Children’s National Health System, Washington, District of Columbia
| | - Adre J. du Plessis
- Professor, Department of Pediatrics and Neurology, George Washington University School of Medicine and Health Sciences, Chief, Division of Fetal and Transitional Medicine, Children’s National Health System, Washington, District of Columbia
| |
Collapse
|
735
|
Suwabe K, Byun K, Hyodo K, Reagh ZM, Roberts JM, Matsushita A, Saotome K, Ochi G, Fukuie T, Suzuki K, Sankai Y, Yassa MA, Soya H. Reply to Gronwald et al.: Exercise intensity does indeed matter; maximal oxygen uptake is the gold-standard indicator. Proc Natl Acad Sci U S A 2018; 115:E11892-E11893. [PMID: 30559331 PMCID: PMC6304988 DOI: 10.1073/pnas.1818247115] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Kazuya Suwabe
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Faculty of Health and Sport Sciences, University of Tsukuba, 305-8574 Ibaraki, Japan
- Sports Neuroscience Division, Advanced Research Initiative for Human High Performance (ARIHHP), Faculty of Health and Sport Sciences, University of Tsukuba, 305-8574 Ibaraki, Japan
| | - Kyeongho Byun
- Sports Neuroscience Division, Advanced Research Initiative for Human High Performance (ARIHHP), Faculty of Health and Sport Sciences, University of Tsukuba, 305-8574 Ibaraki, Japan
| | - Kazuki Hyodo
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Faculty of Health and Sport Sciences, University of Tsukuba, 305-8574 Ibaraki, Japan
| | - Zachariah M Reagh
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, CA 92697
| | - Jared M Roberts
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, CA 92697
| | - Akira Matsushita
- Center for Cybernics Research, University of Tsukuba, 305-8574 Ibaraki, Japan
- Department of Neurology, Ibaraki Prefectural University of Health Sciences, 300-0394 Ibaraki, Japan
| | - Kousaku Saotome
- Center for Cybernics Research, University of Tsukuba, 305-8574 Ibaraki, Japan
| | - Genta Ochi
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Faculty of Health and Sport Sciences, University of Tsukuba, 305-8574 Ibaraki, Japan
| | - Takemune Fukuie
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Faculty of Health and Sport Sciences, University of Tsukuba, 305-8574 Ibaraki, Japan
| | - Kenji Suzuki
- Center for Cybernics Research, University of Tsukuba, 305-8574 Ibaraki, Japan
| | - Yoshiyuki Sankai
- Center for Cybernics Research, University of Tsukuba, 305-8574 Ibaraki, Japan
| | - Michael A Yassa
- Sports Neuroscience Division, Advanced Research Initiative for Human High Performance (ARIHHP), Faculty of Health and Sport Sciences, University of Tsukuba, 305-8574 Ibaraki, Japan;
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697
- Center for the Neurobiology of Learning and Memory, University of California, Irvine, CA 92697
| | - Hideaki Soya
- Laboratory of Exercise Biochemistry and Neuroendocrinology, Faculty of Health and Sport Sciences, University of Tsukuba, 305-8574 Ibaraki, Japan;
- Sports Neuroscience Division, Advanced Research Initiative for Human High Performance (ARIHHP), Faculty of Health and Sport Sciences, University of Tsukuba, 305-8574 Ibaraki, Japan
| |
Collapse
|
736
|
Rogers CE, Lean RE, Wheelock MD, Smyser CD. Aberrant structural and functional connectivity and neurodevelopmental impairment in preterm children. J Neurodev Disord 2018; 10:38. [PMID: 30541449 PMCID: PMC6291944 DOI: 10.1186/s11689-018-9253-x] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 11/14/2018] [Indexed: 12/15/2022] Open
Abstract
Background Despite advances in antenatal and neonatal care, preterm birth remains a leading cause of neurological disabilities in children. Infants born prematurely, particularly those delivered at the earliest gestational ages, commonly demonstrate increased rates of impairment across multiple neurodevelopmental domains. Indeed, the current literature establishes that preterm birth is a leading risk factor for cerebral palsy, is associated with executive function deficits, increases risk for impaired receptive and expressive language skills, and is linked with higher rates of co-occurring attention deficit hyperactivity disorder, anxiety, and autism spectrum disorders. These same infants also demonstrate elevated rates of aberrant cerebral structural and functional connectivity, with persistent changes evident across advanced magnetic resonance imaging modalities as early as the neonatal period. Emerging findings from cross-sectional and longitudinal investigations increasingly suggest that aberrant connectivity within key functional networks and white matter tracts may underlie the neurodevelopmental impairments common in this population. Main body This review begins by highlighting the elevated rates of neurodevelopmental disorders across domains in this clinical population, describes the patterns of aberrant structural and functional connectivity common in prematurely-born infants and children, and then reviews the increasingly established body of literature delineating the relationship between these brain abnormalities and adverse neurodevelopmental outcomes. We also detail important, typically understudied, clinical, and social variables that may influence these relationships among preterm children, including heritability and psychosocial risks. Conclusion Future work in this domain should continue to leverage longitudinal evaluations of preterm infants which include both neuroimaging and detailed serial neurodevelopmental assessments to further characterize relationships between imaging measures and impairment, information necessary for advancing our understanding of modifiable risk factors underlying these disorders and best practices for improving neurodevelopmental trajectories in this high-risk clinical population.
Collapse
Affiliation(s)
- Cynthia E Rogers
- Departments of Psychiatry and Pediatrics, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8504, St. Louis, MO, 63110, USA.
| | - Rachel E Lean
- Departments of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8504, St. Louis, MO, 63110, USA
| | - Muriah D Wheelock
- Departments of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8504, St. Louis, MO, 63110, USA
| | - Christopher D Smyser
- Departments of Neurology, Pediatrics and Mallinckrodt Institute of Radiology, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8111, St. Louis, MO, 63110, USA
| |
Collapse
|
737
|
Seitz J, Kubicki M, Jacobs EG, Cherkerzian S, Weiss BK, Papadimitriou G, Mouradian P, Buka S, Goldstein JM, Makris N. Impact of sex and reproductive status on memory circuitry structure and function in early midlife using structural covariance analysis. Hum Brain Mapp 2018; 40:1221-1233. [PMID: 30548738 DOI: 10.1002/hbm.24441] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 10/11/2018] [Accepted: 10/13/2018] [Indexed: 01/13/2023] Open
Abstract
Research on age-related memory alterations traditionally targets individuals aged ≥65 years. However, recent studies emphasize the importance of early aging processes. We therefore aimed to characterize variation in brain gray matter structure in early midlife as a function of sex and menopausal status. Subjects included 94 women (33 premenopausal, 29 perimenopausal, and 32 postmenopausal) and 99 demographically comparable men from the New England Family Study. Subjects were scanned with a high-resolution T1 sequence on a 3 T whole body scanner. Sex and reproductive-dependent structural differences were evaluated using Box's M test and analysis of covariances (ANCOVAs) for gray matter volumes. Brain regions of interest included dorsolateral prefrontal cortex (DLPFC), inferior parietal lobule (iPAR), anterior cingulate cortex (ACC), hippocampus (HIPP), and parahippocampus. While we observed expected significant sex differences in volume of hippocampus with women of all groups having higher volumes than men relative to cerebrum size, we also found significant differences in the covariance matrices of perimenopausal women compared with postmenopausal women. Associations between ACC and HIPP/iPAR/DLPFC were higher in postmenopausal women and correlated with better memory performance. Findings in this study underscore the importance of sex and reproductive status in early midlife for understanding memory function with aging.
Collapse
Affiliation(s)
- Johanna Seitz
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Marek Kubicki
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.,Departments of Psychiatry, Neurology and Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Center for Morphometric Analysis, Center for Neural Systems Investigations, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts.,Department of Psychiatry, Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts
| | - Emily G Jacobs
- Department of Medicine, Harvard Medical School, Boston, Massachusetts.,Division of Women's Health, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Sara Cherkerzian
- Department of Psychiatry, Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts.,Division of Women's Health, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Blair K Weiss
- Department of Medicine, Harvard Medical School, Boston, Massachusetts.,Division of Women's Health, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - George Papadimitriou
- Departments of Psychiatry, Neurology and Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Center for Morphometric Analysis, Center for Neural Systems Investigations, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts
| | - Palig Mouradian
- Departments of Psychiatry, Neurology and Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Center for Morphometric Analysis, Center for Neural Systems Investigations, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts
| | - Stephen Buka
- Department of Community Health, Brown University, Providence, Rhode Island
| | - Jill M Goldstein
- Departments of Psychiatry, Neurology and Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Center for Morphometric Analysis, Center for Neural Systems Investigations, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts.,Department of Psychiatry, Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts.,Division of Women's Health, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Nikos Makris
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.,Departments of Psychiatry, Neurology and Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Center for Morphometric Analysis, Center for Neural Systems Investigations, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts
| |
Collapse
|
738
|
Low brain-derived neurotrophic factor levels in post-mortem brains of older adults with depression and dementia in a large clinicopathological sample. J Affect Disord 2018; 241:176-181. [PMID: 30125821 DOI: 10.1016/j.jad.2018.08.025] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 07/02/2018] [Accepted: 08/07/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND Disturbances in peripheral brain-derived neurotrophic factor (BDNF) have been reported in major depressive disorder (MDD). However, there are no studies measuring BDNF levels directly in post-mortem brains of older subjects with MDD and dementia. We aimed to verify if brain BDNF levels were lower in older adults with lifetime history of MDD with and without dementia. METHODS BDNF levels of post-mortem brains from 80 community-dwelling older individuals with lifetime MDD with and without dementia were compared with levels from 80 controls without lifetime MDD. Participants with no reliable close informant, or with prolonged agonal state were excluded. Lifetime MDD was defined as at least one previous episode according to the Structured Clinical Interview for DSM (SCID). RESULTS BDNF levels were lower in the MDD group with dementia than in participants with dementia and without MDD as confirmed by multivariate analysis adjusted for clinical and cardiovascular risk factors (ß = -0.106, 95%CI = -0.204; -0.009, p = 0.034). No difference was found in the group with MDD without dementia compared with their controls. LIMITATIONS The retrospective assessment of a lifetime history of depression may be subject to information bias and this study only establishes a cross-sectional association between lifetime history of MDD and lower levels of BDNF in patients with dementia. CONCLUSIONS In this community sample of older individuals, lower brain BDNF levels were found in cases with both lifetime MDD and dementia. Low BDNF levels could be a moderator to accelerated brain aging observed in MDD with dementia.
Collapse
|
739
|
Woodward NC, Haghani A, Johnson RG, Hsu TM, Saffari A, Sioutas C, Kanoski SE, Finch CE, Morgan TE. Prenatal and early life exposure to air pollution induced hippocampal vascular leakage and impaired neurogenesis in association with behavioral deficits. Transl Psychiatry 2018; 8:261. [PMID: 30498214 PMCID: PMC6265287 DOI: 10.1038/s41398-018-0317-1] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 11/08/2018] [Accepted: 11/13/2018] [Indexed: 01/12/2023] Open
Abstract
Exposure to traffic-related air pollution (TRAP) is associated with a range of neurodevelopmental disorders in human populations. In rodent models, prenatal TRAP exposure increased depressive behaviors and increased brain microglial activity. To identify cellular mechanisms, we examined adult neurogenesis and the blood-brain barrier (BBB) in relation to cognition and motivated behaviors in rats that were exposed to a nano-sized TRAP subfraction from gestation into adulthood. At age 5 months, exposed male rats had 70% fewer newly generated neurons in the dentate gyrus (DG) of the hippocampus. Microglia were activated in DG and CA1 subfields (35% more Iba1). The BBB was altered, with a 75% decrease of the tight junction protein ZO-1 in the CA1 layer, and twofold more iron deposits, a marker of microhemorrhages. The exposed rats had impaired contextual memory (novel object in context), reduced food-seeking behavior, and increased depressive behaviors (forced swim). Deficits of de novo neurogenesis were inversely correlated with depressive behavior, whereas increased microbleeds were inversely correlated with deficits in contextual memory. These findings give the first evidence that prenatal and early life exposure to TRAP impairs adult hippocampal neurogenesis and increases microbleeds in association with behavioral deficits.
Collapse
Affiliation(s)
- N. C. Woodward
- 0000 0001 2156 6853grid.42505.36Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA USA
| | - A. Haghani
- 0000 0001 2156 6853grid.42505.36Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA USA
| | - R. G. Johnson
- 0000 0001 2156 6853grid.42505.36Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA USA
| | - T. M. Hsu
- 0000 0001 2156 6853grid.42505.36Human and Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA USA ,0000 0001 2156 6853grid.42505.36Neuroscience Program, University of Southern California, Los Angeles, CA USA
| | - A. Saffari
- 0000 0001 2156 6853grid.42505.36Viterbi School of Engineering, University of Southern California, Los Angeles, CA USA
| | - C. Sioutas
- 0000 0001 2156 6853grid.42505.36Viterbi School of Engineering, University of Southern California, Los Angeles, CA USA
| | - S. E. Kanoski
- 0000 0001 2156 6853grid.42505.36Human and Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA USA ,0000 0001 2156 6853grid.42505.36Neuroscience Program, University of Southern California, Los Angeles, CA USA
| | - C. E. Finch
- 0000 0001 2156 6853grid.42505.36Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA USA ,0000 0001 2156 6853grid.42505.36Dornsife College, University of Southern California, Los Angeles, CA USA
| | - T. E. Morgan
- 0000 0001 2156 6853grid.42505.36Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA USA
| |
Collapse
|
740
|
Hodges TE, Louth EL, Bailey CDC, McCormick CM. Adolescent social instability stress alters markers of synaptic plasticity and dendritic structure in the medial amygdala and lateral septum in male rats. Brain Struct Funct 2018; 224:643-659. [DOI: 10.1007/s00429-018-1789-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 11/03/2018] [Indexed: 10/27/2022]
|
741
|
Marcolin ML, Hodges TE, Baumbach JL, McCormick CM. Adolescent social stress and social context influence the intake of ethanol and sucrose in male rats soon and long after the stress exposures. Dev Psychobiol 2018; 61:81-95. [PMID: 30402884 DOI: 10.1002/dev.21800] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 09/01/2018] [Accepted: 10/04/2018] [Indexed: 12/31/2022]
Abstract
Social instability stress in adolescent rats (SS; postnatal day 30-45, daily 1 hr isolation +new cage partner) alters behavioural responses to psychostimulants, but differences in voluntary consumption of natural and drug rewards are unknown. SS also is associated with an atypical behavioural repertoire, for example reduced social interactions. Here, we investigated whether SS rats differ from control (CTL) rats in ethanol (EtOH) or sucrose intake in experiments involving different social contexts: alone, in the presence of an unfamiliar peer, in the presence of its cage partner, or in competition against its cage partner. SS rats drank more EtOH than CTL rats irrespective of social context, although the effects were driven primarily by those tested soon after the test procedure rather than weeks later in adulthood. SS and CTL rats did not differ in sucrose intake, except in adulthood under conditions of competition for limited access (SS>CTL). Adolescent rats drank more sucrose than adults, in keeping with evidence that adolescents are more sensitive to natural rewards than adult animals. Overall, adolescent SS modified the reward value of EtOH and sucrose, perhaps through stress/glucocorticoids modifying the development of the mesocorticolimbic system.
Collapse
Affiliation(s)
- Marina L Marcolin
- Department of Biological Sciences, Brock University, St. Catharines, Ontario
| | - Travis E Hodges
- Department of Psychology, Brock University, St. Catharines, Ontario
| | | | - Cheryl M McCormick
- Department of Psychology, Brock University, St. Catharines, Ontario.,Centre for Neuroscience, Brock University, St. Catharines, Ontario
| |
Collapse
|
742
|
Goldfarb EV, Sinha R. Drug-Induced Glucocorticoids and Memory for Substance Use. Trends Neurosci 2018; 41:853-868. [PMID: 30170822 PMCID: PMC6204074 DOI: 10.1016/j.tins.2018.08.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 07/13/2018] [Accepted: 08/08/2018] [Indexed: 11/29/2022]
Abstract
The biological stress response of the body forms one of the foundations of adaptive behavior, including promoting (and impairing) different forms of memory. This response transcends stressful experiences and underlies reactions to challenges and even reinforcers such as addictive substances. Nevertheless, drug-induced stress responses are rarely incorporated into models of addiction. We propose here that drug-induced stress responses (particularly glucocorticoids) play a crucial role in addictive behavior by modulating the formation of memories for substance-use experiences. We review the contributions of amygdala-, striatum-, and hippocampus-based memory systems to addiction, and reveal common effects of addictive drugs and acute stress on these different memories. We suggest that the contributions of drug-induced stress responses to memory may provide insights into the mechanisms driving addictive behavior.
Collapse
Affiliation(s)
- Elizabeth V Goldfarb
- Department of Diagnostic Radiology; Yale Stress Center; Yale University School of Medicine, New Haven, CT, USA.
| | - Rajita Sinha
- Departments of Psychiatry and Neuroscience; Yale Stress Center; Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
743
|
Cappucci U, Torromino G, Casale AM, Camon J, Capitano F, Berloco M, Mele A, Pimpinelli S, Rinaldi A, Piacentini L. Stress-induced strain and brain region-specific activation of LINE-1 transposons in adult mice. Stress 2018; 21:575-579. [PMID: 29996702 DOI: 10.1080/10253890.2018.1485647] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Transposable elements (TEs) are conserved mobile genetic elements that are highly abundant in most eukaryotic genomes. Although the exact function of TEs is still largely unknown, it is increasingly clear that they are significantly modulated in response to stress in a wide range of organisms, either directly or indirectly through regulation of epigenetic silencing. We investigated the effect of repeated restraint stress (2 h a day, for 5 d) on transcription levels of LINE-1 (L1) retrotransposon in the brain of inbred BALB/c, DBA/2, C57BL/6N, and outbred CD1 mice. Repeated restraint stress induced strain and brain region-specific modulation of L1 activity. We observed a significant derepression of L1 transcription in the hippocampus (HIPP) of BALB/c mice and a significant downregulation in the hippocampus of C57BL/6N mice. No significant change in L1 expression was found in the other strains and brain regions. These findings indicate in mice the control of transposons expression as an additional mechanism in stress-induced pathophysiological responses, demonstrating that their regulation is highly dependent on the strain genetic background and the brain region. Lay summary Hippocampal expression of the transposon L1 is significantly altered by repeated restraint stress in mice. L1 modulation is not only region specific, but also strain dependent, suggesting that the genetic background is an important determinant of L1 response to environmental stimuli.
Collapse
Affiliation(s)
- Ugo Cappucci
- a Department of Biology and Biotechnology "C. Darwin" , Sapienza University of Rome , Rome , Italy
- b Istituto Pasteur Italia , Fondazione Cenci-Bolognetti , Rome , Italy
| | - Giulia Torromino
- a Department of Biology and Biotechnology "C. Darwin" , Sapienza University of Rome , Rome , Italy
| | - Assunta Maria Casale
- a Department of Biology and Biotechnology "C. Darwin" , Sapienza University of Rome , Rome , Italy
- b Istituto Pasteur Italia , Fondazione Cenci-Bolognetti , Rome , Italy
| | - Jeremy Camon
- a Department of Biology and Biotechnology "C. Darwin" , Sapienza University of Rome , Rome , Italy
| | - Fabrizio Capitano
- a Department of Biology and Biotechnology "C. Darwin" , Sapienza University of Rome , Rome , Italy
| | - Maria Berloco
- c Department of Biology , University of Bari "Aldo Moro" , Bari , Italy
| | - Andrea Mele
- a Department of Biology and Biotechnology "C. Darwin" , Sapienza University of Rome , Rome , Italy
- d Center for Research in Neurobiology "D. Bovet" , Sapienza University of Rome , Rome , Italy
| | - Sergio Pimpinelli
- a Department of Biology and Biotechnology "C. Darwin" , Sapienza University of Rome , Rome , Italy
- b Istituto Pasteur Italia , Fondazione Cenci-Bolognetti , Rome , Italy
| | - Arianna Rinaldi
- a Department of Biology and Biotechnology "C. Darwin" , Sapienza University of Rome , Rome , Italy
- d Center for Research in Neurobiology "D. Bovet" , Sapienza University of Rome , Rome , Italy
| | - Lucia Piacentini
- a Department of Biology and Biotechnology "C. Darwin" , Sapienza University of Rome , Rome , Italy
- b Istituto Pasteur Italia , Fondazione Cenci-Bolognetti , Rome , Italy
| |
Collapse
|
744
|
Dopfel D, Zhang N. Mapping stress networks using functional magnetic resonance imaging in awake animals. Neurobiol Stress 2018; 9:251-263. [PMID: 30450389 PMCID: PMC6234259 DOI: 10.1016/j.ynstr.2018.06.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Revised: 05/27/2018] [Accepted: 06/26/2018] [Indexed: 12/15/2022] Open
Abstract
The neurobiology of stress is studied through behavioral neuroscience, endocrinology, neuronal morphology and neurophysiology. There is a shift in focus toward progressive changes throughout stress paradigms and individual susceptibility to stress that requires methods that allow for longitudinal study design and study of individual differences in stress response. Functional magnetic resonance imaging (fMRI), with the advantages of noninvasiveness and a large field of view, can be used for functionally mapping brain-wide regions and circuits critical to the stress response, making it suitable for longitudinal studies and understanding individual variability of short-term and long-term consequences of stress exposure. In addition, fMRI can be applied to both animals and humans, which is highly valuable in translating findings across species and examining whether the physiology and neural circuits involved in the stress response are conserved in mammals. However, compared to human fMRI studies, there are a number of factors that are essential for the success of fMRI studies in animals. This review discussed the use of fMRI in animal studies of stress. It reviewed advantages, challenges and technical considerations of the animal fMRI methodology as well as recent literature of stress studies using fMRI in animals. It also highlighted the development of combining fMRI with other methods and the future potential of fMRI in animal studies of stress. We conclude that animal fMRI studies, with their flexibility, low cost and short time frame compared to human studies, are crucial to advancing our understanding of the neurobiology of stress.
Collapse
Affiliation(s)
- David Dopfel
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, 16802, USA
| | - Nanyin Zhang
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, 16802, USA
- The Huck Institutes of Life Sciences, Pennsylvania State University, University Park, PA, 16802, USA
| |
Collapse
|
745
|
Saxbe D, Lyden H, Del Piero L, Stoycos SA, Gimbel SI, Margolin G, Kaplan JT. Community violence exposure in early adolescence: Longitudinal associations with hippocampal and amygdala volume and resting state connectivity. Dev Sci 2018; 21:e12686. [PMID: 29890029 PMCID: PMC11694245 DOI: 10.1111/desc.12686] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 02/01/2018] [Indexed: 11/29/2022]
Abstract
Community violence exposure is a common stressor, known to compromise youth cognitive and emotional development. In a diverse, urban sample of 22 adolescents, participants reported on community violence exposure (witnessing a beating or illegal drug use, hearing gun shots, or other forms of community violence) in early adolescence (average age 12.99), and underwent a neuroimaging scan 3-5 years later (average age 16.92). Community violence exposure in early adolescence predicted smaller manually traced left and right hippocampal and amygdala volumes in a model controlling for age, gender, and concurrent community violence exposure, measured in late adolescence. Community violence continued to predict hippocampus (but not amygdala) volumes after we also controlled for family aggression exposure in early adolescence. Community violence exposure was also associated with stronger resting state connectivity between the right hippocampus (using the manually traced structure as a seed region) and bilateral frontotemporal regions including the superior temporal gyrus and insula. These resting state connectivity results held after controlling for concurrent community violence exposure, SES, and family aggression. Although this is the first study focusing on community violence in conjunction with brain structure and function, these results dovetail with other research linking childhood adversity with smaller subcortical volumes in adolescence and adulthood, and with altered frontolimbic resting state connectivity. Our findings suggest that even community-level exposure to neighborhood violence can have detectable neural correlates in adolescents.
Collapse
Affiliation(s)
- Darby Saxbe
- Department of Psychology, University of Southern California, Los Angeles CA
| | - Hannah Lyden
- Department of Psychology, University of Southern California, Los Angeles CA
| | | | - Sarah A. Stoycos
- Department of Psychology, University of Southern California, Los Angeles CA
| | - Sarah I. Gimbel
- Brain and Creativity Institute, University of Southern California, Los Angeles, CA
| | - Gayla Margolin
- Department of Psychology, University of Southern California, Los Angeles CA
| | - Jonas T. Kaplan
- Department of Psychology, University of Southern California, Los Angeles CA
| |
Collapse
|
746
|
Ottino-González J, Jurado MA, García-García I, Segura B, Marqués-Iturria I, Sender-Palacios MJ, Tor E, Prats-Soteras X, Caldú X, Junqué C, Pasternak O, Garolera M. Allostatic load and disordered white matter microstructure in overweight adults. Sci Rep 2018; 8:15898. [PMID: 30367110 PMCID: PMC6203765 DOI: 10.1038/s41598-018-34219-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 10/12/2018] [Indexed: 12/29/2022] Open
Abstract
Overweight and stress are both related to brain structural abnormalities. The allostatic load model states that frequent disruption of homeostasis is inherently linked to oxidative stress and inflammatory responses that in turn can damage the brain. However, the effects of the allostatic load on the central nervous system remain largely unknown. The current study aimed to assess the relationship between the allostatic load and the composition of whole-brain white matter tracts in overweight subjects. Additionally, we have also tested for grey matter changes regarding allostatic load increase. Thirty-one overweight-to-obese adults and 21 lean controls participated in the study. Our results showed that overweight participants presented higher allostatic load indexes. Such increases correlated with lower fractional anisotropy in the inferior fronto-occipital fasciculi and the right anterior corona radiata, as well as with grey matter reductions in the left precentral gyrus, the left lateral occipital gyrus, and the right pars opercularis. These results suggest that an otherwise healthy overweight status is linked to long-term biological changes potentially harmful to the brain.
Collapse
Affiliation(s)
- J Ottino-González
- Departament de Psicologia Clínica i Psicobiologia, Universitat de Barcelona, Barcelona, Spain
- Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu (IRSJD), Barcelona, Spain
| | - M A Jurado
- Departament de Psicologia Clínica i Psicobiologia, Universitat de Barcelona, Barcelona, Spain.
- Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.
- Institut de Recerca Sant Joan de Déu (IRSJD), Barcelona, Spain.
| | - I García-García
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - B Segura
- Departament de Medicina, Universitat de Barcelona, Barcelona, Spain
- Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - I Marqués-Iturria
- Departament de Psicologia Clínica i Psicobiologia, Universitat de Barcelona, Barcelona, Spain
| | - M J Sender-Palacios
- CAP Terrassa Nord, Consorci Sanitari de Terrassa, Barcelona, Spain
- Brain, Cognition and Behavior Clinical Research Group, Consorci Sanitari de Terrassa, Barcelona, Spain
| | - E Tor
- CAP Terrassa Nord, Consorci Sanitari de Terrassa, Barcelona, Spain
- Brain, Cognition and Behavior Clinical Research Group, Consorci Sanitari de Terrassa, Barcelona, Spain
| | - X Prats-Soteras
- Departament de Psicologia Clínica i Psicobiologia, Universitat de Barcelona, Barcelona, Spain
- Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu (IRSJD), Barcelona, Spain
| | - X Caldú
- Departament de Psicologia Clínica i Psicobiologia, Universitat de Barcelona, Barcelona, Spain
- Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
- Institut de Recerca Sant Joan de Déu (IRSJD), Barcelona, Spain
| | - C Junqué
- Departament de Medicina, Universitat de Barcelona, Barcelona, Spain
- Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - O Pasternak
- Departments of Psychiatry and Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - M Garolera
- Unitat de Neuropsicologia, Hospital de Terrassa, Consorci Sanitari de Terrassa, Barcelona, Spain
- Brain, Cognition and Behavior Clinical Research Group, Consorci Sanitari de Terrassa, Barcelona, Spain
| |
Collapse
|
747
|
Functional Neurochemistry of the Ventral and Dorsal Hippocampus: Stress, Depression, Dementia and Remote Hippocampal Damage. Neurochem Res 2018; 44:1306-1322. [PMID: 30357653 DOI: 10.1007/s11064-018-2662-0] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 09/15/2018] [Accepted: 10/15/2018] [Indexed: 12/15/2022]
Abstract
The hippocampus is not a homogeneous brain area, and the complex organization of this structure underlies its relevance and functional pleiotropism. The new data related to the involvement of the ventral hippocampus in the cognitive function, behavior, stress response and its association with brain pathology, in particular, depression, are analyzed with a focus on neuroplasticity, specializations of the intrinsic neuronal network, corticosteroid signaling through mineralocorticoid and glucocorticoid receptors and neuroinflammation in the hippocampus. The data on the septo-temporal hippicampal gradient are analyzed with particular emphasis on the ventral hippocampus, a region where most important alteration underlying depressive disorders occur. According to the recent data, the existing simple paradigm "learning (dorsal hippocampus) versus emotions (ventral hippocampus)" should be substantially revised and specified. A new hypothesis is suggested on the principal involvement of stress response mechanisms (including interaction of released glucocorticoids with hippocampal receptors and subsequent inflammatory events) in the remote hippocampal damage underlying delayed dementia and depression induced by focal brain damage (e.g. post-stroke and post-traumatic). The translational validity of this hypothesis comprising new approaches in preventing post-stroke and post-trauma depression and dementia can be confirmed in experimental and clinical studies.
Collapse
|
748
|
Rombold-Bruehl F, Otte C, Renneberg B, Hellmann-Regen J, Bruch L, Wingenfeld K, Roepke S. Impact of stress response systems on forced choice recognition in an experimental trauma film paradigm. Neurobiol Learn Mem 2018; 156:45-52. [PMID: 30352264 DOI: 10.1016/j.nlm.2018.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 08/31/2018] [Accepted: 10/20/2018] [Indexed: 10/28/2022]
Abstract
INTRODUCTION Traumatic events are often followed by memory impairments of key features of the trauma. Stress hormones are involved in emotional memory formation. However, little is known about their influence during trauma on subsequent recognition memory. MATERIAL AND METHODS A pooled analysis of two double-blind, placebo-controlled studies (N = 175) was performed to assess the influence of the noradrenergic system and the hypothalamus-pituitaryadrenal (HPA) axis on intrusion formation. Participants received either 10 mg yohimbine (stimulating noradrenergic activity), 0.15 mg clonidine (inhibiting noradrenergic activity), or placebo (noradrenergic manipulation study) or 20 mg hydrocortisone or placebo (hydrocortisone manipulation study), each 60 min before watching a distressing film depicting severe sexual and physical violence. After seven days, the participants performed a 24-item forced choice recognition test. Memory was assessed for pre-, peri-, and post-trauma film scenes. RESULTS A significant film scene by intervention interaction indicated a differential influence of drug intervention on the number of correct pre-, peri-, and post-trauma film scene memories one week after the distressing film. Post hoc tests revealed that clonidine led to significantly fewer correct peri-trauma film scene memories compared to placebo and, on a trend level, to yohimbine. DISCUSSION Pharmacological inhibition of noradrenaline during a distressing film leads to impaired emotional recognition memory for the peri-trauma film scene.
Collapse
Affiliation(s)
- Felicitas Rombold-Bruehl
- Department of Psychiatry and Psychotherapy, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Benjamin Franklin, Germany; Department of Psychology, Freie Universität Berlin, Germany.
| | - Christian Otte
- Department of Psychiatry and Psychotherapy, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Benjamin Franklin, Germany
| | | | - Julian Hellmann-Regen
- Department of Psychiatry and Psychotherapy, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Benjamin Franklin, Germany
| | - Linda Bruch
- Department of Psychiatry and Psychotherapy, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Benjamin Franklin, Germany
| | - Katja Wingenfeld
- Department of Psychiatry and Psychotherapy, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Benjamin Franklin, Germany
| | - Stefan Roepke
- Department of Psychiatry and Psychotherapy, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Benjamin Franklin, Germany
| |
Collapse
|
749
|
Akter S, Sasaki H, Ikeda Y, Miyakawa H, Shibata S. γ-oryzanol ameliorates the acute stress induced by behavioral anxiety testing in mice. J Pharmacol Sci 2018; 138:155-159. [PMID: 30322802 DOI: 10.1016/j.jphs.2018.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 05/08/2018] [Accepted: 05/28/2018] [Indexed: 10/14/2022] Open
Abstract
We evaluated the anxiolytic effect of γ-oryzanol (GORZ) and elucidated the molecular mechanisms involved in its inhibition of behavioral test-induced anxiety. Behavioral tests were conducted on day 13, and mice were subjected to 30 min of acute restraint stress treatment (ARST) before sacrifice on day 16. In other group, behavioral tests were conducted on day 13 and 14 after ARST. 0.5% GORZ significantly weakened the effect of behavioral stress, but not the effect of strong ARST. GORZ downregulated ARST-induced cFos levels in the cerebral cortex. In conclusion, GORZ has potential ant-anxiety effect in the treatment of weak behavioral test-induced stress.
Collapse
Affiliation(s)
- Salina Akter
- Laboratory of Physiology and Pharmacology, School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Hiroyuki Sasaki
- Laboratory of Physiology and Pharmacology, School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Yuko Ikeda
- Laboratory of Physiology and Pharmacology, School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Hiroki Miyakawa
- Laboratory of Physiology and Pharmacology, School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Shigenobu Shibata
- Laboratory of Physiology and Pharmacology, School of Advanced Science and Engineering, Waseda University, Tokyo, Japan.
| |
Collapse
|
750
|
Maghami S, Zardooz H, Khodagholi F, Binayi F, Ranjbar Saber R, Hedayati M, Sahraei H, Ansari MA. Maternal separation blunted spatial memory formation independent of peripheral and hippocampal insulin content in young adult male rats. PLoS One 2018; 13:e0204731. [PMID: 30332425 PMCID: PMC6192583 DOI: 10.1371/journal.pone.0204731] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 09/13/2018] [Indexed: 01/22/2023] Open
Abstract
This study explores the effects of maternal separation as a chronic early life stress (ELS) on pancreatic islets insulin content and secretion, and their potential relationship with the hippocampus insulin content and spatial memory in young adulthood. Male rat offspring were divided into two groups: stress (STR) and non-stress (non-STR) groups. The animals of the STR group were separated from their mothers during postnatal days (PND) 1 to 21. During the weaning time, that is, PND-0 to PND-21, the body weight and length of the pups were measured. Blood samples were collected on PND-1, 21, 29 and 34 and during young adulthood (53±2 days) to determine plasma corticosterone and insulin levels. The young adult animals were also tested for spatial memory. One day after the memory test, the animals were decapitated and their pancreases were removed to measure the islets insulin content and secretion. Finally, the animals' hippocampi were isolated to determine their insulin content and insulin receptor protein amounts. During the period of weaning, the body weight and length of pups belonging to the STR group were significantly lower as compared to those in the non-STR group. Maternal separation did not change the plasma levels of insulin but increased plasma corticosterone levels from PND-21 to young adulthood and also reduced the islets insulin content but did not affect insulin secretion and the hippocampus insulin content and insulin receptor protein amount. Although, at the end of the memory tests, rats of the STR group reached the escape box at almost the same time and distance and with the same errors as rats of the non-STR group, the distance traveled to reach the escape box showed a steep reduction in the non-STR group as compared to the STR group after the first trial. Moreover, as compared to the STR group, the non-STR group showed an increasing trend for direct strategy to find the escape box. The islets insulin content and secretion, and the plasma insulin concentration were not significantly correlated with the hippocampus insulin content. From the results of the present study, it appears that the main behavioral effect of the maternal separation stress in the spatial memory task was to impair the strategy used by the animals to reach the escape box. This may indicate that maternal separation stress affects brain regions other than the hippocampus. Moreover, due to the reduction of the body weight and length of offspring belonging to the STR group, it should be further considered that both maternal separation and early life malnutrition are directly (and mechanistically) linked to cognitive alterations later in life in ways that are not dependent on peripheral and hippocampal insulin content.
Collapse
Affiliation(s)
- Soheila Maghami
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Homeira Zardooz
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- * E-mail: ,
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Binayi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Roya Ranjbar Saber
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Hedayati
- Cellular and Molecular Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hedayat Sahraei
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Ansari
- Laser and Plasma Research Institute, Shahid Beheshti University, Tehran, Iran
| |
Collapse
|