1
|
Zanta NC, Assad N, Suchecki D. Neurobiological mechanisms involved in maternal deprivation-induced behaviours relevant to psychiatric disorders. Front Mol Neurosci 2023; 16:1099284. [PMID: 37122626 PMCID: PMC10133561 DOI: 10.3389/fnmol.2023.1099284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 03/23/2023] [Indexed: 05/02/2023] Open
Abstract
Parental care is essential for proper development of stress response and emotion-related behaviours. Epidemiological studies show that parental loss in childhood represents a major risk factor for the development of mental disorders throughout the lifespan, including schizophrenia, depression, and anxiety. In most mammalian species, the mother is the main source of care and maternal behaviours regulate several physiological systems. Maternal deprivation (DEP) for 24 h is a paradigm widely used to disinhibit the hypothalamic-pituitary-adrenal axis response to stress during the stress hyporesponsive period. In this mini-review we will highlight the main DEP-induced neurobiological and behavioural outcomes, including alterations on stress-related hormones, neurogenesis, neurotransmitter/neuromodulatory systems and neuroinflammation. These neurobiological changes may be reflected by aberrant behaviours, which are relevant to the study of mental disorders. The evidence indicates that DEP consequences depend on the sex, the age when the DEP takes place and the age when the animals are evaluated, reflecting dynamic plasticity and individual variability. Individual variability and sex differences have a great relevance for the study of biological factors of stress resilience and vulnerability and the DEP paradigm is a suitable model for evaluation of phenotypes of stress- and emotion-related psychopathologies.
Collapse
|
2
|
Repeated and single maternal separation specifically alter microglial morphology in the prefrontal cortex and neurogenesis in the hippocampus of 15-day-old male mice. Neuroreport 2020; 31:1256-1264. [DOI: 10.1097/wnr.0000000000001544] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
3
|
Závodská M, Fabianová K, Martončíková M, Raček A, Račeková E. Low Fos expression in newly generated neurons of the main and accessory olfactory bulb following single maternal separation. Stress 2020; 23:678-687. [PMID: 33375878 DOI: 10.1080/10253890.2020.1828337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
The main and accessory olfactory bulbs (MOB and AOB) are unique in that they produce new neurons throughout adulthood. Despite the recent knowledge about the involvement of postnatally generated cells in several aspects of olfaction, the functional role of these neurons is still not sufficiently understood. The function of newly generated olfactory bulb neurons is primarily investigated in relation to activities related to smell. Stress-induced activation of new olfactory neurons has not yet been studied. Thus, our work was aimed to investigate whether a stressful event, such as maternal separation (MS) can induce Fos expression in postnatally-born neurons in the MOB and AOB. Rat pups were exposed to single maternal separation (SMS) for 2 h at the postnatal days: P7, P14, and P21. Quantification of immunohistochemically labeled Fos + cells revealed that exposure to SMS in different age stages during the first postnatal month stimulates activity in cells of individual MOB/AOB layers in an age-dependent manner. In order to find out whether newly generated cells in the MOB/AOB could express Fos protein as a response to SMS, newborn rats were administrated with the marker of proliferation, bromodeoxyuridine (BrdU) at P0, and three weeks later (at P21) colocalization of Fos and BrdU in the neurons of the MOB and AOB was assessed. Quantitative analysis of BrdU/Fos double-labeled cells showed that Fos is expressed only in a small number of postnatally generated cells within the MOB/AOB. Our results indicate that postnatally generated MOB/AOB neurons are less sensitive to stress caused by MS than preexisting ones. LAY SUMMARY Our results showed that single maternal separation (SMS) is a stressful event that in age-dependent manner stimulates cellular activity in the main and accessory olfactory bulb (AOB) - the structures dedicated to odor information processing. The low level of Fos expression in newborn neurons of the main and accessory bulb indicates that postnatally generated cells are less sensitive to neonatal stress than preexisting neurons.
Collapse
Affiliation(s)
- Monika Závodská
- Department of Regenerative Medicine and Cell Therapy, Institute of Neurobiology, Biomedical Research Center, Slovak Academy of Sciences, Košice, Slovak Republic
| | - Kamila Fabianová
- Department of Regenerative Medicine and Cell Therapy, Institute of Neurobiology, Biomedical Research Center, Slovak Academy of Sciences, Košice, Slovak Republic
| | - Marcela Martončíková
- Department of Regenerative Medicine and Cell Therapy, Institute of Neurobiology, Biomedical Research Center, Slovak Academy of Sciences, Košice, Slovak Republic
| | - Adam Raček
- Department of Anatomy, Histology and Physiology, University of Veterinary Medicine and Pharmacy in Kosice, Košice, Slovak Republic
| | - Enikő Račeková
- Department of Regenerative Medicine and Cell Therapy, Institute of Neurobiology, Biomedical Research Center, Slovak Academy of Sciences, Košice, Slovak Republic
| |
Collapse
|
4
|
Italia M, Forastieri C, Longaretti A, Battaglioli E, Rusconi F. Rationale, Relevance, and Limits of Stress-Induced Psychopathology in Rodents as Models for Psychiatry Research: An Introductory Overview. Int J Mol Sci 2020; 21:E7455. [PMID: 33050350 PMCID: PMC7589795 DOI: 10.3390/ijms21207455] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/07/2020] [Accepted: 10/08/2020] [Indexed: 12/18/2022] Open
Abstract
Emotional and cognitive information processing represent higher-order brain functions. They require coordinated interaction of specialized brain areas via a complex spatial and temporal equilibrium among neuronal cell-autonomous, circuitry, and network mechanisms. The delicate balance can be corrupted by stressful experiences, increasing the risk of developing psychopathologies in vulnerable individuals. Neuropsychiatric disorders affect twenty percent of the western world population, but therapies are still not effective for some patients. Elusive knowledge of molecular pathomechanisms and scarcity of objective biomarkers in humans present complex challenges, while the adoption of rodent models helps to improve our understanding of disease correlate and aids the search for novel pharmacological targets. Stress administration represents a strategy to induce, trace, and modify molecular and behavioral endophenotypes of mood disorders in animals. However, a mouse or rat model will only display one or a few endophenotypes of a specific human psychopathology, which cannot be in any case recapitulated as a whole. To override this issue, shared criteria have been adopted to deconstruct neuropsychiatric disorders, i.e., depression, into specific behavioral aspects, and inherent neurobiological substrates, also recognizable in lower mammals. In this work, we provide a rationale for rodent models of stress administration. In particular, comparing each rodent model with a real-life human traumatic experience, we intend to suggest an introductive guide to better comprehend and interpret these paradigms.
Collapse
|
5
|
van der Veen R, Bonapersona V, Joëls M. The relevance of a rodent cohort in the Consortium on Individual Development. Dev Cogn Neurosci 2020; 45:100846. [PMID: 32957026 PMCID: PMC7509002 DOI: 10.1016/j.dcn.2020.100846] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 07/29/2020] [Accepted: 08/23/2020] [Indexed: 12/31/2022] Open
Abstract
One of the features of the Consortium on Individual Development is the existence of a rodent cohort, in parallel with the human cohorts. Here we give an overview of the current status. We first elaborate on the choice of rat and mouse models mimicking early life adverse or beneficial conditions during development. We performed a systematic literature search on early life adversity and adult social behavior to address the status quo. Next, we describe the behavioral tasks we used and designed to examine behavioral control and social competence in rodents. The results so far indicate that manipulation of the environment in the first postnatal week only subtly affects social behavior. Stronger effects were seen in the model that targeted early adolescence; once adult, these rats are characterized by increased attention, a higher degree of impulsiveness and reduced social interest in peers. Many experiments in our rodent models with tightly controlled conditions were inspired by findings in human cohorts, and now allow in-depth mechanistic investigations. Vice versa, some of the findings in rodents are currently followed up by dedicated investigations in the human cohorts. This exemplifies the added value of animal investigations in a consortium encompassing primarily human developmental cohorts.
Collapse
Affiliation(s)
- Rixt van der Veen
- Dept. Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; Faculty of Social and Behavioral Sciences, Leiden University, Leiden, the Netherlands.
| | - Valeria Bonapersona
- Dept. Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Marian Joëls
- Dept. Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
6
|
Nyman C, Hebert FO, Bessert‐Nettelbeck M, Aubin‐Horth N, Taborsky B. Transcriptomic signatures of social experience during early development in a highly social cichlid fish. Mol Ecol 2019; 29:610-623. [DOI: 10.1111/mec.15335] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 11/23/2019] [Accepted: 12/10/2019] [Indexed: 12/01/2022]
Affiliation(s)
- Cecilia Nyman
- Division of Behavioural Ecology Institute of Ecology and Evolution University of Bern Bern Switzerland
| | - Francois Olivier Hebert
- Département de Biologie and Institut de Biologie Intégrative et des Systèmes Université Laval Laval QC Canada
| | | | - Nadia Aubin‐Horth
- Département de Biologie and Institut de Biologie Intégrative et des Systèmes Université Laval Laval QC Canada
| | - Barbara Taborsky
- Division of Behavioural Ecology Institute of Ecology and Evolution University of Bern Bern Switzerland
| |
Collapse
|
7
|
Abstract
The developmental period constitutes a critical window of sensitivity to stress. Indeed, early-life adversity increases the risk to develop psychiatric diseases, but also gastrointestinal disorders such as the irritable bowel syndrome at adulthood. In the past decade, there has been huge interest in the gut-brain axis, especially as regards stress-related emotional behaviours. Animal models of early-life adversity, in particular, maternal separation (MS) in rodents, demonstrate lasting deleterious effects on both the gut and the brain. Here, we review the effects of MS on both systems with a focus on stress-related behaviours. In addition, we discuss more recent findings showing the impact of gut-directed interventions, including nutrition with pre- and probiotics, illustrating the role played by gut microbiota in mediating the long-term effects of MS. Overall, preclinical studies suggest that nutritional approaches with pro- and prebiotics may constitute safe and efficient strategies to attenuate the effects of early-life stress on the gut-brain axis. Further research is required to understand the complex mechanisms underlying gut-brain interaction dysfunctions after early-life stress as well as to determine the beneficial impact of gut-directed strategies in a context of early-life adversity in human subjects.
Collapse
|
8
|
Kentner AC, Cryan JF, Brummelte S. Resilience priming: Translational models for understanding resiliency and adaptation to early life adversity. Dev Psychobiol 2019; 61:350-375. [PMID: 30311210 PMCID: PMC6447439 DOI: 10.1002/dev.21775] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 06/22/2018] [Accepted: 07/10/2018] [Indexed: 12/20/2022]
Abstract
Despite the increasing attention to early life adversity and its long-term consequences on health, behavior, and the etiology of neurodevelopmental disorders, our understanding of the adaptations and interventions that promote resiliency and rescue against such insults are underexplored. Specifically, investigations of the perinatal period often focus on negative events/outcomes. In contrast, positive experiences (i.e. enrichment/parental care//healthy nutrition) favorably influence development of the nervous and endocrine systems. Moreover, some stressors result in adaptations and demonstrations of later-life resiliency. This review explores the underlying mechanisms of neuroplasticity that follow some of these early life experiences and translates them into ideas for interventions in pediatric settings. The emerging role of the gut microbiome in mediating stress susceptibility is also discussed. Since many negative outcomes of early experiences are known, it is time to identify mechanisms and mediators that promote resiliency against them. These range from enrichment, quality parental care, dietary interventions and those that target the gut microbiota.
Collapse
Affiliation(s)
- Amanda C. Kentner
- School of Arts & Sciences, Massachusetts College of Pharmacy and Health Sciences, 179 Longwood Ave, Boston, MA 02115,
| | - John F. Cryan
- Dept. Anatomy & Neuroscience & APC Microbiome Institute, University College Cork, College Rd., Cork, Ireland,
| | - Susanne Brummelte
- Department of Psychology, Wayne State University, 5057 Woodward Ave, Detroit, MI 48202,
| |
Collapse
|
9
|
Brain Mineralocorticoid Receptors and Resilience to Stress. VITAMINS AND HORMONES 2019; 109:341-359. [DOI: 10.1016/bs.vh.2018.11.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/26/2023]
|
10
|
Liu PZ, Nusslock R. How Stress Gets Under the Skin: Early Life Adversity and Glucocorticoid Receptor Epigenetic Regulation. Curr Genomics 2018; 19:653-664. [PMID: 30532645 PMCID: PMC6225447 DOI: 10.2174/1389202919666171228164350] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 05/31/2017] [Accepted: 12/17/2017] [Indexed: 11/22/2022] Open
Abstract
Early life adversity is associated with both persistent disruptions in the hypothalamic-pituitary-adrenal (HPA) axis and psychiatric symptoms. Glucocorticoid receptors (GRs), which are encoded by the NR3C1 gene, bind to cortisol and other glucocorticoids to create a negative feedback loop within the HPA axis to regulate the body's neuroendocrine response to stress. Excess methylation of a promoter sequence within NR3C1 that attenuates GR expression, however, has been associated with both early life adversity and psychopathology. As critical regulators within the HPA axis, GRs and their epigenetic regulation may mediate the link between early life adversity and the onset of psychopathology. The present review discusses this work as one mechanism by which stress may get under the skin to disrupt HPA functioning at an epigenetic level and create long-lasting vulnerabilities in the stress regulatory system that subsequently predispose individuals to psychopathology. Spanning prenatal influences to critical periods of early life and adolescence, we detail the impact that early adversity has on GR expression, physiological responses to stress, and their implications for long-term stress management. We next propose a dual transmission hypothesis regarding both genomic and non-genomic mechanisms by which chronic and acute stress propagate through numerous generations. Lastly, we outline several directions for future research, including potential reversibility of methylation patterns and its functional implications, variation in behavior determined solely by NR3C1, and consensus on which specific promoter regions should be studied.
Collapse
Affiliation(s)
- Patrick Z. Liu
- Department of Psychology, Northwestern University, Evanston, IL60208, USA
| | - Robin Nusslock
- Department of Psychology, Northwestern University, Evanston, IL60208, USA
| |
Collapse
|
11
|
Yoshida S, Ohnishi R, Tsuneoka Y, Yamamoto-Mimura Y, Muramatsu R, Kato T, Funato H, Kuroda KO. Corticotropin-Releasing Factor Receptor 1 in the Anterior Cingulate Cortex Mediates Maternal Absence-Induced Attenuation of Transport Response in Mouse Pups. Front Cell Neurosci 2018; 12:204. [PMID: 30057526 PMCID: PMC6053499 DOI: 10.3389/fncel.2018.00204] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 06/22/2018] [Indexed: 01/24/2023] Open
Abstract
A human infant initially shows non-selective sociality, and gradually develops selective attachment toward its caregiver, manifested as "separation anxiety." It was unclear whether such sophistication of attachment system occurs in non-human mammals. To seek a mouse model of separation anxiety, we utilized a primitive attachment behavior, the Transport Response, in that both human and mouse newborns immediately stop crying and stay immobile to cooperate with maternal carrying. We examined the mouse Transport Response in three social contexts: 30-min isolation in a novel environment, 30-min maternal absence experienced with littermates in the home cage, and the control home-cage condition with the mother and littermates. The pups after postnatal day (PND) 13 attenuated their Transport Response not only in complete isolation but also by maternal absence, and activated several brain areas including the periventricular nucleus of the hypothalamus, suggesting that 30-min maternal absence was perceived as a social stress by mouse pups after PND13. This attenuation of Transport Response by maternal absence was independent with plasma corticosterone, but was diminished by prior administration of a corticotropin-releasing factor receptor 1 (CRFR1) antagonist. Among 18 brain areas examined, only neurons in the anterior cingulate cortex (ACC) co-express c-fos mRNA and CRFR1 after maternal absence. Consistently, excitotoxic ACC lesions inhibited the maternal absence-induced attenuation of Transport Response. These data indicate that the expression of mouse Transport Response is influenced not only by social isolation but also by maternal absence even in their home cage with littermates after PND13, at least partly via CRF-CRFR1 signaling in the ACC.
Collapse
Affiliation(s)
- Sachine Yoshida
- Department of Anatomy, Faculty of Medicine, Toho University, Tokyo, Japan
- Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency (JST), Saitama, Japan
- Laboratory for Affiliative Social Behavior, Center for Brain Science, RIKEN, Saitama, Japan
| | - Ryuko Ohnishi
- Laboratory for Affiliative Social Behavior, Center for Brain Science, RIKEN, Saitama, Japan
- Department of Bioscience and Biotechnology, Faculty of Agriculture, University of the Ryukyus, Nishihara, Japan
| | - Yousuke Tsuneoka
- Department of Anatomy, Faculty of Medicine, Toho University, Tokyo, Japan
- Laboratory for Affiliative Social Behavior, Center for Brain Science, RIKEN, Saitama, Japan
| | | | - Reiko Muramatsu
- Department of Anatomy, Faculty of Medicine, Toho University, Tokyo, Japan
| | - Tadafumi Kato
- Laboratory for Molecular Dynamics of Mental Disorders, Center for Brain Science, RIKEN, Saitama, Japan
| | - Hiromasa Funato
- Department of Anatomy, Faculty of Medicine, Toho University, Tokyo, Japan
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba, Japan
| | - Kumi O. Kuroda
- Laboratory for Affiliative Social Behavior, Center for Brain Science, RIKEN, Saitama, Japan
| |
Collapse
|
12
|
Suchecki D. Maternal regulation of the infant's hypothalamic-pituitary-adrenal axis stress response: Seymour 'Gig' Levine's legacy to neuroendocrinology. J Neuroendocrinol 2018; 30:e12610. [PMID: 29774962 DOI: 10.1111/jne.12610] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 05/13/2018] [Accepted: 05/13/2018] [Indexed: 12/23/2022]
Abstract
Thirty years ago, Seymour 'Gig' Levine published a serendipitous, yet, seminal finding with respect to the regulatory role of maternal presence on the corticosterone stress response of neonatal rats during the developmental period known as the stress hyporesponsive period. At the same time, his group of students also investigated the stress response of infant monkeys with respect to maternal separation, as a means of understanding the stress to the primary caregiver resulting from disruptions of attachment. Gig and his group of students and collaborators, mainly in the USA and the Netherlands, investigated how initial social relationships buffer the stress response of nonhuman primates and rodent infants. His work in rodents involved determining how prolonged deprivation of maternal care disinhibits the stress response of neonates and how maternal behaviours regulate specific aspects of the hypothalamic-pituitary-adrenal axis. Maternal deprivation for 24 hours was useful for determining the importance of nutrition in suppressing the corticosterone stress response, whereas anogenital licking and grooming inhibited stress-induced adrenocortoctrophic hormone release, with the combination of both behaviours preventing the effects of maternal deprivation on the central hypothalamic stress response. Levine's group also studied the consequences of maternal deprivation on basal and stress-induced activity of the hypothalamic-pituitary-adrenal axis in juveniles and the persistent effects of the replacement of maternal behaviours on these parameters. Gig's legacy allowed many groups around the world to use the 24-hour maternal deprivation paradigm as an animal model of vulnerability and resilience to stress-related psychiatric disorders, as well as in studies of the neurobiological underpinnings of disruption of the mother-infant relationship and loss of parental care, a highly prevalent condition in humans. This review pays homage to a great scientist and mentor, whose discoveries paved the way for the understanding of how early social relationsships build resilience or lead to susceptibility to emotional disorders later in life.
Collapse
Affiliation(s)
- D Suchecki
- Departamento de Psicobiologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
13
|
Neonatal- maternal separation primes zymogenic cells in the rat gastric mucosa through glucocorticoid receptor activity. Sci Rep 2018; 8:9823. [PMID: 29959361 PMCID: PMC6026145 DOI: 10.1038/s41598-018-28223-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 06/19/2018] [Indexed: 12/21/2022] Open
Abstract
Neonatal- Maternal Separation (NMS) deprives mammals from breastfeeding and maternal care, influencing growth during suckling- weaning transition. In the gastric mucosa, Mist1 (encoded by Bhlha15 gene) and moesin organize the secretory apparatus for pepsinogen C in zymogenic cells. Our current hypothesis was that NMS would change corticosterone activity through receptors (GR), which would modify molecules involved in zymogenic cell differentiation in rats. We found that NMS increased corticosterone levels from 18 days onwards, as GR decreased in the gastric mucosa. However, as nuclear GR was detected, we investigated receptor binding to responsive elements (GRE) and observed an augment in NMS groups. Next, we demonstrated that NMS increased zymogenic population (18 and and 30 days), and targeted Mist1 and moesin. Finally, we searched for evolutionarily conserved sequences that contained GRE in genes involved in pepsinogen C secretion, and found that the genomic regions of Bhlha15 and PgC contained sites highly likely to be responsive to glucocorticoids. We suggest that NMS triggers GR- GRE to enhance the expression and to prime genes that organize cellular architecture in zymogenic population for PgC function. As pepsinogen C- pepsin is essential for digestion, disturbance of parenting through NMS might alter functions of gastric mucosa in a permanent manner.
Collapse
|
14
|
Katahira T, Miyazaki N, Motoyama J. Immediate effects of maternal separation on the development of interneurons derived from medial ganglionic eminence in the neonatal mouse hippocampus. Dev Growth Differ 2018; 60:278-290. [DOI: 10.1111/dgd.12540] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 04/25/2018] [Accepted: 04/25/2018] [Indexed: 01/21/2023]
Affiliation(s)
- Tatsuya Katahira
- Organization of Advanced Research and Education; Doshisha University; Kyoto Japan
| | | | - Jun Motoyama
- Laboratory of Developmental Neurobiology; Graduate School of Brain Science; Doshisha University; Kyoto Japan
| |
Collapse
|
15
|
Sominsky L, Jasoni CL, Twigg HR, Spencer SJ. Hormonal and nutritional regulation of postnatal hypothalamic development. J Endocrinol 2018; 237:R47-R64. [PMID: 29545398 DOI: 10.1530/joe-17-0722] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 03/15/2018] [Indexed: 12/24/2022]
Abstract
The hypothalamus is a key centre for regulation of vital physiological functions, such as appetite, stress responsiveness and reproduction. Development of the different hypothalamic nuclei and its major neuronal populations begins prenatally in both altricial and precocial species, with the fine tuning of neuronal connectivity and attainment of adult function established postnatally and maintained throughout adult life. The perinatal period is highly susceptible to environmental insults that, by disrupting critical developmental processes, can set the tone for the establishment of adult functionality. Here, we review the most recent knowledge regarding the major postnatal milestones in the development of metabolic, stress and reproductive hypothalamic circuitries, in the rodent, with a particular focus on perinatal programming of these circuitries by hormonal and nutritional influences. We also review the evidence for the continuous development of the hypothalamus in the adult brain, through changes in neurogenesis, synaptogenesis and epigenetic modifications. This degree of plasticity has encouraging implications for the ability of the hypothalamus to at least partially reverse the effects of perinatal mal-programming.
Collapse
Affiliation(s)
- Luba Sominsky
- School of Health and Biomedical SciencesRMIT University, Melbourne, Victoria, Australia
| | - Christine L Jasoni
- School of Biomedical SciencesCentre for Neuroendocrinology, Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - Hannah R Twigg
- School of Biomedical SciencesCentre for Neuroendocrinology, Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - Sarah J Spencer
- School of Health and Biomedical SciencesRMIT University, Melbourne, Victoria, Australia
| |
Collapse
|
16
|
Jacobskind JS, Rosinger ZJ, Zuloaga DG. Hypothalamic-pituitary-adrenal axis responsiveness to methamphetamine is modulated by gonadectomy in males. Brain Res 2017; 1677:74-85. [PMID: 28941573 DOI: 10.1016/j.brainres.2017.09.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 08/18/2017] [Accepted: 09/17/2017] [Indexed: 12/14/2022]
Abstract
Sex differences in patterns of methamphetamine (MA) abuse have been reported with females (humans and rodents) showing an elevated addiction phenotype. Previous findings indicate MA-induced hypothalamic-pituitary-adrenal (HPA) axis activation is also sexually dimorphic with females exhibiting an elevated glucocorticoid release and differential neural activation patterns within HPA axis-associated brain regions. These effects may contribute to sex differences in abuse. To determine the role of gonadal hormones in mediating sex differences in MA-induced glucocorticoids, male and female C57BL/6J mice were gonadectomized or sham-operated, and following recovery, injected with MA (5mg/kg) and sacrificed 60min or 120min later. Blood was collected for corticosterone radioimmunoassay, and brains were used to assess c-Fos, and c-Fos co-localization with glucocorticoid receptor (GR). At 120min after MA injection, corticosterone levels were elevated in females compared to males and gonadectomy in males increased corticosterone to female levels. C-Fos was greater in females than males in the medial preoptic area, bed nucleus of the stria terminalis, basolateral amygdala, and central amygdala. Female gonadectomy had little effect on either corticosterone or c-Fos, while male gonadectomy elevated c-Fos in the central amygdala. Relative to sham males, gonadectomized males also showed decreased c-Fos/GR cell number in the CA3 hippocampal area compared to sham males, indicating a central site for attenuated negative feedback. Together, these findings indicate that androgens regulate MA-induced activation of the HPA axis, potentially by enhancing negative feedback. These sex and gonadal hormone effects on the HPA axis may contribute to sex differences in MA abuse patterns.
Collapse
Affiliation(s)
- Jason S Jacobskind
- University at Albany, Department of Psychology, Albany, NY 12222, United States
| | - Zachary J Rosinger
- University at Albany, Department of Psychology, Albany, NY 12222, United States
| | - Damian G Zuloaga
- University at Albany, Department of Psychology, Albany, NY 12222, United States.
| |
Collapse
|
17
|
van Bodegom M, Homberg JR, Henckens MJAG. Modulation of the Hypothalamic-Pituitary-Adrenal Axis by Early Life Stress Exposure. Front Cell Neurosci 2017; 11:87. [PMID: 28469557 PMCID: PMC5395581 DOI: 10.3389/fncel.2017.00087] [Citation(s) in RCA: 325] [Impact Index Per Article: 46.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 03/13/2017] [Indexed: 12/20/2022] Open
Abstract
Exposure to stress during critical periods in development can have severe long-term consequences, increasing overall risk on psychopathology. One of the key stress response systems mediating these long-term effects of stress is the hypothalamic-pituitary-adrenal (HPA) axis; a cascade of central and peripheral events resulting in the release of corticosteroids from the adrenal glands. Activation of the HPA-axis affects brain functioning to ensure a proper behavioral response to the stressor, but stress-induced (mal)adaptation of the HPA-axis' functional maturation may provide a mechanistic basis for the altered stress susceptibility later in life. Development of the HPA-axis and the brain regions involved in its regulation starts prenatally and continues after birth, and is protected by several mechanisms preventing corticosteroid over-exposure to the maturing brain. Nevertheless, early life stress (ELS) exposure has been reported to have numerous consequences on HPA-axis function in adulthood, affecting both its basal and stress-induced activity. According to the match/mismatch theory, encountering ELS prepares an organism for similar ("matching") adversities during adulthood, while a mismatching environment results in an increased susceptibility to psychopathology, indicating that ELS can exert either beneficial or disadvantageous effects depending on the environmental context. Here, we review studies investigating the mechanistic underpinnings of the ELS-induced alterations in the structural and functional development of the HPA-axis and its key external regulators (amygdala, hippocampus, and prefrontal cortex). The effects of ELS appear highly dependent on the developmental time window affected, the sex of the offspring, and the developmental stage at which effects are assessed. Albeit by distinct mechanisms, ELS induced by prenatal stressors, maternal separation, or the limited nesting model inducing fragmented maternal care, typically results in HPA-axis hyper-reactivity in adulthood, as also found in major depression. This hyper-activity is related to increased corticotrophin-releasing hormone signaling and impaired glucocorticoid receptor-mediated negative feedback. In contrast, initial evidence for HPA-axis hypo-reactivity is observed for early social deprivation, potentially reflecting the abnormal HPA-axis function as observed in post-traumatic stress disorder, and future studies should investigate its neural/neuroendocrine foundation in further detail. Interestingly, experiencing additional (chronic) stress in adulthood seems to normalize these alterations in HPA-axis function, supporting the match/mismatch theory.
Collapse
Affiliation(s)
| | | | - Marloes J. A. G. Henckens
- Department of Cognitive Neuroscience, Centre for Neuroscience, Donders Institute for Brain, Cognition and BehaviourRadboudumc, Nijmegen, Netherlands
| |
Collapse
|
18
|
Moussaoui N, Jacobs JP, Larauche M, Biraud M, Million M, Mayer E, Taché Y. Chronic Early-life Stress in Rat Pups Alters Basal Corticosterone, Intestinal Permeability, and Fecal Microbiota at Weaning: Influence of Sex. J Neurogastroenterol Motil 2017; 23:135-143. [PMID: 27829577 PMCID: PMC5216644 DOI: 10.5056/jnm16105] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Revised: 08/09/2016] [Accepted: 10/11/2016] [Indexed: 12/16/2022] Open
Abstract
Background/Aims Wistar rat dams exposed to limited nesting stress (LNS) from post-natal days (PND) 2 to 10 display erratic maternal behavior, and their pups show delayed maturation of the hypothalamic-pituitary-adrenal axis and impaired epithelial barrier at PND10 and a visceral hypersensitivity at adulthood. Little is known about the impact of early life stress on the offspring before adulthood and the influence of sex. We investigated whether male and female rats previously exposed to LNS displays at weaning altered corticosterone, intestinal permeability, and microbiota. Methods Wistar rat dams and litters were maintained from PND2 to 10 with limited nesting/bedding materials and thereafter reverted to normal housing up to weaning (PND21). Control litters had normal housing. At weaning, we monitored body weight, corticosterone plasma levels (enzyme immunoassay), in vivo intestinal to colon permeability (fluorescein isothiocyanate-dextran 4 kDa) and fecal microbiota (DNA extraction and amplification of the V4 region of the 16S ribosomal RNA gene). Results At weaning, LNS pups had hypercorticosteronemia and enhanced intestinal permeability with females > males while body weights were similar. LNS decreased fecal microbial diversity and induced a distinct composition characterized by increased abundance of Gram positive cocci and reduction of fiber-degrading, butyrate-producing, and mucus-resident microbes. Conclusions These data indicate that chronic exposure to LNS during the first week post-natally has sustained effects monitored at weaning including hypercorticosteronemia, a leaky gut, and dysbiosis. These alterations may impact on the susceptibility to develop visceral hypersensitivity in adult rats and have relevance to the development of irritable bowel syndrome in childhood.
Collapse
Affiliation(s)
- Nabila Moussaoui
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, CURE: Digestive Diseases Research Center, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine and Brain Research Institute, University of California, and VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Jonathan P Jacobs
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, CURE: Digestive Diseases Research Center, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine and Brain Research Institute, University of California, and VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Muriel Larauche
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, CURE: Digestive Diseases Research Center, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine and Brain Research Institute, University of California, and VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Mandy Biraud
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, CURE: Digestive Diseases Research Center, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine and Brain Research Institute, University of California, and VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Mulugeta Million
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, CURE: Digestive Diseases Research Center, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine and Brain Research Institute, University of California, and VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Emeran Mayer
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, CURE: Digestive Diseases Research Center, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine and Brain Research Institute, University of California, and VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Yvette Taché
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, CURE: Digestive Diseases Research Center, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine and Brain Research Institute, University of California, and VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| |
Collapse
|
19
|
Varga J, Ferenczi S, Kovács KJ, Csáno Á, Prokopova B, Jezova D, Zelena D. Dissociation of adrenocorticotropin and corticosterone as well as aldosterone secretion during stress of hypoglycemia in vasopressin-deficient rats. Life Sci 2016; 166:66-74. [PMID: 27744052 DOI: 10.1016/j.lfs.2016.10.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 09/28/2016] [Accepted: 10/11/2016] [Indexed: 11/29/2022]
Affiliation(s)
- János Varga
- Department of Behavioral Neurobiology, Institute of Experimental Medicine, 1083 Budapest, Szigony 43, Hungary
| | - Szilamér Ferenczi
- Laboratory of Molecular Neuroendocrinology, Institute of Experimental Medicine, 1083 Budapest, Szigony 43, Hungary
| | - Krisztina J Kovács
- Laboratory of Molecular Neuroendocrinology, Institute of Experimental Medicine, 1083 Budapest, Szigony 43, Hungary
| | - Ágnes Csáno
- Laboratory of Pharmacological Neuroendocrinology, Institute of Experimental Endocrinology, BMC, Slovak Academy of Sciences, Dubravska cesta 9, 84505 Bratislava, Slovakia
| | - Barbora Prokopova
- Laboratory of Pharmacological Neuroendocrinology, Institute of Experimental Endocrinology, BMC, Slovak Academy of Sciences, Dubravska cesta 9, 84505 Bratislava, Slovakia
| | - Daniela Jezova
- Laboratory of Pharmacological Neuroendocrinology, Institute of Experimental Endocrinology, BMC, Slovak Academy of Sciences, Dubravska cesta 9, 84505 Bratislava, Slovakia
| | - Dóra Zelena
- Department of Behavioral Neurobiology, Institute of Experimental Medicine, 1083 Budapest, Szigony 43, Hungary.
| |
Collapse
|
20
|
How age, sex and genotype shape the stress response. Neurobiol Stress 2016; 6:44-56. [PMID: 28229108 PMCID: PMC5314441 DOI: 10.1016/j.ynstr.2016.11.004] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 11/19/2016] [Accepted: 11/21/2016] [Indexed: 12/21/2022] Open
Abstract
Exposure to chronic stress is a leading pre-disposing factor for several neuropsychiatric disorders as it often leads to maladaptive responses. The response to stressful events is heterogeneous, underpinning a wide spectrum of distinct changes amongst stress-exposed individuals'. Several factors can underlie a different perception to stressors and the setting of distinct coping strategies that will lead to individual differences on the susceptibility/resistance to stress. Beyond the factors related to the stressor itself, such as intensity, duration or predictability, there are factors intrinsic to the individuals that are relevant to shape the stress response, such as age, sex and genetics. In this review, we examine the contribution of such intrinsic factors to the modulation of the stress response based on experimental rodent models of response to stress and discuss to what extent that knowledge can be potentially translated to humans. Effect of age in the stress response. Effect of sex in the stress response. Effect of genotype in the stress response.
Collapse
|
21
|
Moussaoui N, Larauche M, Biraud M, Molet J, Million M, Mayer E, Taché Y. Limited Nesting Stress Alters Maternal Behavior and In Vivo Intestinal Permeability in Male Wistar Pup Rats. PLoS One 2016; 11:e0155037. [PMID: 27149676 PMCID: PMC4858303 DOI: 10.1371/journal.pone.0155037] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 04/22/2016] [Indexed: 12/17/2022] Open
Abstract
A few studies indicate that limited nesting stress (LNS) alters maternal behavior and the hypothalamic pituitary adrenal (HPA) axis of dams and offspring in male Sprague Dawley rats. In the present study, we evaluated the impact of LNS on maternal behavior in Wistar rats, and on the HPA axis, glycemia and in vivo intestinal permeability of male and female offspring. Intestinal permeability is known to be elevated during the first week postnatally and influenced by glucocorticoids. Dams and neonatal litters were subjected to LNS or normal nesting conditions (control) from days 2 to 10 postnatally. At day 10, blood was collected from pups for determination of glucose and plasma corticosterone by enzyme immunoassay and in vivo intestinal permeability by oral gavage of fluorescein isothiocyanate-dextran 4kDa. Dams exposed to LNS compared to control showed an increase in the percentage of time spent building a nest (118%), self-grooming (69%), and putting the pups back to the nest (167%). LNS male and female pups exhibited a reduction of body weight by 5% and 4%, adrenal weights/100g body weight by 17% and 18%, corticosterone plasma levels by 64% and 62% and blood glucose by 11% and 12% respectively compared to same sex control pups. In male LNS pups, intestinal permeability was increased by 2.7-fold while no change was observed in females compared to same sex control. There was no sex difference in any of the parameters in control pups except the body weight. These data indicate that Wistar dams subjected to LNS during the first postnatal week have an altered repertoire of maternal behaviors which affects the development of the HPA axis in both sexes and intestinal barrier function in male offspring.
Collapse
Affiliation(s)
- Nabila Moussaoui
- Center for Neurobiology of Stress, CURE: Digestive Diseases Research Center, Digestive Diseases Division, Department of Medicine and Brain Research Institute, University of California Los Angeles, and VA Greater Los Angeles Healthcare System, Los Angeles, California, 90073, United States of America
- * E-mail:
| | - Muriel Larauche
- Center for Neurobiology of Stress, CURE: Digestive Diseases Research Center, Digestive Diseases Division, Department of Medicine and Brain Research Institute, University of California Los Angeles, and VA Greater Los Angeles Healthcare System, Los Angeles, California, 90073, United States of America
| | - Mandy Biraud
- Center for Neurobiology of Stress, CURE: Digestive Diseases Research Center, Digestive Diseases Division, Department of Medicine and Brain Research Institute, University of California Los Angeles, and VA Greater Los Angeles Healthcare System, Los Angeles, California, 90073, United States of America
| | - Jenny Molet
- Department of Anatomy/Neurobiology, University of California Irvine, Irvine, CA, 92697–4475, United States of America
| | - Mulugeta Million
- Center for Neurobiology of Stress, CURE: Digestive Diseases Research Center, Digestive Diseases Division, Department of Medicine and Brain Research Institute, University of California Los Angeles, and VA Greater Los Angeles Healthcare System, Los Angeles, California, 90073, United States of America
| | - Emeran Mayer
- Center for Neurobiology of Stress, CURE: Digestive Diseases Research Center, Digestive Diseases Division, Department of Medicine and Brain Research Institute, University of California Los Angeles, and VA Greater Los Angeles Healthcare System, Los Angeles, California, 90073, United States of America
| | - Yvette Taché
- Center for Neurobiology of Stress, CURE: Digestive Diseases Research Center, Digestive Diseases Division, Department of Medicine and Brain Research Institute, University of California Los Angeles, and VA Greater Los Angeles Healthcare System, Los Angeles, California, 90073, United States of America
| |
Collapse
|
22
|
Kawakami SE, Quadros IMH, Suchecki D. Naltrexone Prevents in Males and Attenuates in Females the Expression of Behavioral Sensitization to Ethanol Regardless of Maternal Separation. Front Endocrinol (Lausanne) 2016; 7:135. [PMID: 27803689 PMCID: PMC5067536 DOI: 10.3389/fendo.2016.00135] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 09/20/2016] [Indexed: 02/04/2023] Open
Abstract
Maternal separation alters the activity of the opioid system, which modulates ethanol-induced stimulation and behavioral sensitization. This study examined the effects of an opioid antagonist, naltrexone (NTX), on the expression of behavioral sensitization to ethanol in adult male and female mice submitted to maternal separation from postnatal days (PNDs) 2 to 14. Whole litters of Swiss mice were either not separated [animal facility rearing (AFR)] or separated from their mothers for 3 h [long maternal separation (LMS)]. Starting on PND 90, male and female AFR and LMS mice received daily i.p. injections of saline (SAL) or ethanol (EtOH, 2.2 g/kg) for 21 days. Locomotor activity was assessed in cages containing photoelectric beams, once a week, to examine the development of behavioral sensitization. Five days after the end of the chronic treatment, animals were submitted to four locomotor activity tests spaced by 48 h, to assess the expression of behavioral sensitization. In all tests, animals received two i.p. injections with a 30-min interval and were then assessed for locomotor response to different treatment challenges, which were: SAL/SAL, SAL/EtOH (2.2 g/kg), NTX 2.0 mg/kg (NTX2)/EtOH, and NTX 4.0 mg/kg (NTX4)/EtOH. Regardless of maternal separation, EtOH-treated male and female mice displayed increased locomotor responses to EtOH during the 21-day treatment, indicating the development of behavioral sensitization. In the SAL/EtOH challenge, EtOH-treated LMS and AFR male and female mice exhibited higher locomotor activity than their SAL-treated counterparts, indicating the expression of sensitization. The coadministration of either dose of NTX blocked the expression of locomotor sensitization in both AFR and LMS male mice with a history of EtOH sensitization. In females, a significant attenuation of EtOH sensitization was promoted by both NTX doses, while still maintaining an augmented stimulant response to EtOH. Importantly, maternal separation did not interfere in this phenomenon. These results indicate that expression of behavioral sensitization was importantly modulated by opioidergic mechanisms both in male and female mice and that maternal separation did not play a major role in either development or expression of this EtOH sensitization.
Collapse
Affiliation(s)
- Suzi E. Kawakami
- Department of Psychobiology, Escola Paulista de Medicina – Universidade Federal de São Paulo (UNIFESP), Sao Paulo, Sao Paulo, Brazil
| | - Isabel M. H. Quadros
- Department of Psychobiology, Escola Paulista de Medicina – Universidade Federal de São Paulo (UNIFESP), Sao Paulo, Sao Paulo, Brazil
- *Correspondence: Isabel M. H. Quadros,
| | - Deborah Suchecki
- Department of Psychobiology, Escola Paulista de Medicina – Universidade Federal de São Paulo (UNIFESP), Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
23
|
Hoeijmakers L, Lucassen PJ, Korosi A. The interplay of early-life stress, nutrition, and immune activation programs adult hippocampal structure and function. Front Mol Neurosci 2015; 7:103. [PMID: 25620909 PMCID: PMC4288131 DOI: 10.3389/fnmol.2014.00103] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 12/16/2014] [Indexed: 01/08/2023] Open
Abstract
Early-life adversity increases the vulnerability to develop psychopathologies and cognitive decline later in life. This association is supported by clinical and preclinical studies. Remarkably, experiences of stress during this sensitive period, in the form of abuse or neglect but also early malnutrition or an early immune challenge elicit very similar long-term effects on brain structure and function. During early-life, both exogenous factors like nutrition and maternal care, as well as endogenous modulators, including stress hormones and mediator of immunological activity affect brain development. The interplay of these key elements and their underlying molecular mechanisms are not fully understood. We discuss here the hypothesis that exposure to early-life adversity (specifically stress, under/malnutrition and infection) leads to life-long alterations in hippocampal-related cognitive functions, at least partly via changes in hippocampal neurogenesis. We further discuss how these different key elements of the early-life environment interact and affect one another and suggest that it is a synergistic action of these elements that shapes cognition throughout life. Finally, we consider different intervention studies aiming to prevent these early-life adversity induced consequences. The emerging evidence for the intriguing interplay of stress, nutrition, and immune activity in the early-life programming calls for a more in depth understanding of the interaction of these elements and the underlying mechanisms. This knowledge will help to develop intervention strategies that will converge on a more complete set of changes induced by early-life adversity.
Collapse
Affiliation(s)
- Lianne Hoeijmakers
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam Amsterdam, Netherlands
| | - Paul J Lucassen
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam Amsterdam, Netherlands
| | - Aniko Korosi
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam Amsterdam, Netherlands
| |
Collapse
|
24
|
GABAA receptor-acting neurosteroids: a role in the development and regulation of the stress response. Front Neuroendocrinol 2015; 36:28-48. [PMID: 24929099 PMCID: PMC4349499 DOI: 10.1016/j.yfrne.2014.06.001] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 05/26/2014] [Accepted: 06/01/2014] [Indexed: 12/22/2022]
Abstract
Regulation of hypothalamic-pituitary-adrenocortical (HPA) axis activity by stress is a fundamental survival mechanism and HPA-dysfunction is implicated in psychiatric disorders. Adverse early life experiences, e.g. poor maternal care, negatively influence brain development and programs an abnormal stress response by encoding long-lasting molecular changes, which may extend to the next generation. How HPA-dysfunction leads to the development of affective disorders is complex, but may involve GABAA receptors (GABAARs), as they curtail stress-induced HPA axis activation. Of particular interest are endogenous neurosteroids that potently modulate the function of GABAARs and exhibit stress-protective properties. Importantly, neurosteroid levels rise rapidly during acute stress, are perturbed in chronic stress and are implicated in the behavioural changes associated with early-life adversity. We will appraise how GABAAR-active neurosteroids may impact on HPA axis development and the orchestration of the stress-evoked response. The significance of these actions will be discussed in the context of stress-associated mood disorders.
Collapse
|
25
|
Yam KY, Naninck EFG, Schmidt MV, Lucassen PJ, Korosi A. Early-life adversity programs emotional functions and the neuroendocrine stress system: the contribution of nutrition, metabolic hormones and epigenetic mechanisms. Stress 2015; 18:328-42. [PMID: 26260665 DOI: 10.3109/10253890.2015.1064890] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Clinical and pre-clinical studies have shown that early-life adversities, such as abuse or neglect, can increase the vulnerability to develop psychopathologies and cognitive decline later in life. Remarkably, the lasting consequences of stress during this sensitive period on the hypothalamic-pituitary-adrenal axis and emotional function closely resemble the long-term effects of early malnutrition and suggest a possible common pathway mediating these effects. During early-life, brain development is affected by both exogenous factors, like nutrition and maternal care as well as by endogenous modulators including stress hormones. These elements, while mostly considered for their independent actions, clearly do not act alone but rather in a synergistic manner. In order to better understand how the programming by early-life stress takes place, it is important to gain further insight into the exact interplay of these key elements, the possible common pathways as well as the underlying molecular mechanisms that mediate their effects. We here review evidence that exposure to both early-life stress and early-life under-/malnutrition similarly lead to life-long alterations on the neuroendocrine stress system and modify emotional functions. We further discuss how the different key elements of the early-life environment interact and affect one another and next suggest a possible role for the early-life adversity induced alterations in metabolic hormones and nutrient availability in shaping later stress responses and emotional function throughout life, possibly via epigenetic mechanisms. Such knowledge will help to develop intervention strategies, which gives the advantage of viewing the synergistic action of a more complete set of changes induced by early-life adversity.
Collapse
Affiliation(s)
- Kit-Yi Yam
- a Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam , XH Amsterdam , The Netherlands and
| | - Eva F G Naninck
- a Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam , XH Amsterdam , The Netherlands and
| | - Mathias V Schmidt
- b Department Stress Neurobiology and Neurogenetics , Max Planck Institute of Psychiatry , Munich , Germany
| | - Paul J Lucassen
- a Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam , XH Amsterdam , The Netherlands and
| | - Aniko Korosi
- a Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam , XH Amsterdam , The Netherlands and
| |
Collapse
|
26
|
Abstract
Objectives:To examine the ultrastructural effects of maternal deprivation during developmental periods of limbi-chypothalamo-pituitary-adrenal system on hippocampal dendritic structures in adult rats.Methods:The experiments were carried out with male and female wistar rats in our department. The rats were mated and, after birth, the pups were divided into four groups. The first group (control group) pups remained undisturbed with their dam until postweaning day 22. Maternal deprived groups were separated from their dams for 24 hours at postnatal day 4, 9 and 18. The subjects were provided with food and water ad libitum until 3-months-of-age. At the third month, the rats were transcardially perfused, samples were taken from CA1 and CA3 regions of the hippocampus. Tissues were prepared for electron microscopy.Results:When the data were analyzed, there were no differences between male and female rats in both ultrastructure and semiquantitative analysis of axodendritic synapses. The ultrastructure of Group 1 was seen as normal while in the second Group some neurons nuclear envelope made deep invagination into the nucleus. Additionally, axodendritic synapses were found normal. In Group 3, micrographs and axodendritic synapses were showed normal structure. However, in Group 4 in some neurons invaginations were seen similar to Group 2. Axodendritic synapses were found to be normal.Conclusion:These experiments establish that MD in rats produces slight ultrastructural changes and decreases the number of synapses in CA1 and CA3 subregions of the hippocampus.
Collapse
|
27
|
Bali A, Jaggi AS. Preclinical experimental stress studies: protocols, assessment and comparison. Eur J Pharmacol 2014; 746:282-92. [PMID: 25446911 DOI: 10.1016/j.ejphar.2014.10.017] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 10/08/2014] [Accepted: 10/09/2014] [Indexed: 01/05/2023]
Abstract
Stress is a state of threatened homeostasis during which a variety of adaptive processes are activated to produce physiological and behavioral changes. Preclinical models are pivotal for understanding these physiological or pathophysiological changes in the body in response to stress. Furthermore, these models are also important for the development of novel pharmacological agents for stress management. The well described preclinical stress models include immobilization, restraint, electric foot shock and social isolation stress. Stress assessment in animals is done at the behavioral level using open field, social interaction, hole board test; at the biochemical level by measuring plasma corticosterone and ACTH; at the physiological level by measuring food intake, body weight, adrenal gland weight and gastric ulceration. Furthermore the comparison between different stressors including electric foot shock, immobilization and cold stressor is described in terms of intensity, hormonal release, protein changes in brain, adaptation and sleep pattern. This present review describes these preclinical stress protocols, and stress assessment at different levels.
Collapse
Affiliation(s)
- Anjana Bali
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala 147002, India.
| | - Amteshwar Singh Jaggi
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala 147002, India.
| |
Collapse
|
28
|
Melo AI. Role of sensory, social, and hormonal signals from the mother on the development of offspring. ADVANCES IN NEUROBIOLOGY 2014; 10:219-48. [PMID: 25287543 DOI: 10.1007/978-1-4939-1372-5_11] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
For mammals, sensory, social, and hormonal experience early in life is essential for the continuity of the infant's development. These experiences come from the mother through maternal care, and have enduring effects on the physiology and behavior of the adult organism. Disturbing the mother-offspring interaction by maternal deprivation (neglect) or exposure to adverse events as chronic stress, maltreatment, or sexual abuse has negative effects on the mental, psychological, physiological, and behavioral health. Indeed, these kinds of negative experiences can be the source of some neuropsychiatric diseases as depression, anxiety, impulsive aggression, and antisocial behavior. The purpose of this chapter is to review the most relevant evidence that supports the participation of cues from the mother and/or littermates during the postnatal preweaning period for the development of nervous system of the offspring. These findings come from the most frequently utilized experimental paradigms used in animal models, such as natural variations in maternal behavior, handling, partial maternal deprivation, and total maternal deprivation and artificial rearing. Through the use of these experimental procedures, it is possible to positively (handling paradigm), or negatively (maternal deprivation paradigms), affect the offspring's development. Finally, this chapter reviews the importance of the hormones that pups ingest through the maternal milk during early lactation on the development of several physiological systems, including the immune, endocrine systems, as well as on the adult behavior of the offspring.
Collapse
Affiliation(s)
- Angel I Melo
- Centro de Investigación en Reproducción Animal, CINVESTAV-Laboratorio Tlaxcala, Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico,
| |
Collapse
|
29
|
Haller J, Harold G, Sandi C, Neumann ID. Effects of adverse early-life events on aggression and anti-social behaviours in animals and humans. J Neuroendocrinol 2014; 26:724-38. [PMID: 25059307 DOI: 10.1111/jne.12182] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 07/21/2014] [Accepted: 07/21/2014] [Indexed: 12/12/2022]
Abstract
We review the impact of early adversities on the development of violence and antisocial behaviour in humans, and present three aetiological animal models of escalated rodent aggression, each disentangling the consequences of one particular adverse early-life factor. A review of the human data, as well as those obtained with the animal models of repeated maternal separation, post-weaning social isolation and peripubertal stress, clearly shows that adverse developmental conditions strongly affect aggressive behaviour displayed in adulthood, the emotional responses to social challenges and the neuronal mechanisms activated by conflict. Although similarities between models are evident, important differences were also noted, demonstrating that the behavioural, emotional and neuronal consequences of early adversities are to a large extent dependent on aetiological factors. These findings support recent theories on human aggression, which suggest that particular developmental trajectories lead to specific forms of aggressive behaviour and brain dysfunctions. However, dissecting the roles of particular aetiological factors in humans is difficult because these occur in various combinations; in addition, the neuroscientific tools employed in humans still lack the depth of analysis of those used in animal research. We suggest that the analytical approach of the rodent models presented here may be successfully used to complement human findings and to develop integrative models of the complex relationship between early adversity, brain development and aggressive behaviour.
Collapse
Affiliation(s)
- J Haller
- Institute of Experimental Medicine, Budapest, Hungary
| | | | | | | |
Collapse
|
30
|
Bravo JA, Dinan TG, Cryan JF. Early-life stress induces persistent alterations in 5-HT1A receptor and serotonin transporter mRNA expression in the adult rat brain. Front Mol Neurosci 2014; 7:24. [PMID: 24782706 PMCID: PMC3989758 DOI: 10.3389/fnmol.2014.00024] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 03/19/2014] [Indexed: 11/13/2022] Open
Abstract
Early-life experience plays a major role in the stress response throughout life. Neonatal maternal separation (MS) is an animal model of depression with an altered serotonergic response. We hypothesize that this alteration may be caused by differences in 5-HT1A receptor and serotonin transporter (SERT) mRNA expression in brain areas involved in the control of emotions, memory, and fear as well as in regions controlling the central serotonergic tone. To test this, Sprague-Dawley rats were subjected to MS for 3 h daily during postnatal days 2-12. As control, age matched rats were non-separated (NS) from their dams. When animals reached adulthood (11-13 weeks) brain was extracted and mRNA expression of 5-HT1A receptor in amygdala, hippocampus and dorsal raphé nucleus (DRN) and SERT in the DRN was analyzed through in situ hybridisation. Densitometric analysis revealed that MS increased 5-HT1A receptor mRNA expression in the amygdala, and reduced its expression in the DRN, but no changes were observed in the hippocampus in comparison to NS controls. Also, MS reduced SERT mRNA expression in the DRN when compared to NS rats. These results suggest that early-life stress induces persistent changes in 5-HT1A receptor and SERT mRNA expression in key brain regions involved in the development of stress-related psychiatric disorders. The reduction in SERT mRNA indicates an alteration that is in line with clinical findings such as polymorphic variants in individuals with higher risk of depression. These data may help to understand how early-life stress contributes to the development of mood disorders in adulthood.
Collapse
Affiliation(s)
- Javier A Bravo
- Grupo de NeuroGastroBioquímica, Laboratorio de Química Biológica, Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso Valparaíso, Chile
| | - Timothy G Dinan
- Department of Psychiatry, University College Cork Cork, Ireland ; Laboratory of Neurogastroenterology, Alimentary Pharmabiotic Centre, University College Cork Cork, Ireland
| | - John F Cryan
- Laboratory of Neurogastroenterology, Alimentary Pharmabiotic Centre, University College Cork Cork, Ireland ; Department of Anatomy, University College Cork Cork, Ireland
| |
Collapse
|
31
|
Moussaoui N, Braniste V, Ait-Belgnaoui A, Gabanou M, Sekkal S, Olier M, Théodorou V, Martin PGP, Houdeau E. Changes in intestinal glucocorticoid sensitivity in early life shape the risk of epithelial barrier defect in maternal-deprived rats. PLoS One 2014; 9:e88382. [PMID: 24586321 PMCID: PMC3930527 DOI: 10.1371/journal.pone.0088382] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 01/06/2014] [Indexed: 12/17/2022] Open
Abstract
Glucocorticoids (GC) contribute to human intestine ontogeny and accelerate gut barrier development in preparation to birth. Rat gut is immature at birth, and high intestinal GC sensitivity during the first two weeks of life resembles that of premature infants. This makes suckling rats a model to investigate postpartum impact of maternal separation (MS)-associated GC release in preterm babies, and whether GC sensitivity may shape MS effects in immature gut. A 4 hours-MS applied once at postnatal day (PND)10 enhanced plasma corticosterone in male and female pups, increased by two times the total in vivo intestinal permeability (IP) to oral FITC-Dextran 4 kDa (FD4) immediately after the end of MS, and induced bacterial translocation (BT) to liver and spleen. Ussing chamber experiments demonstrated a 2-fold increase of permeability to FD4 in the colon immediately after the end of MS, but not in the ileum. Colonic permeability was not only increased for FD4 but also to intact horseradish peroxidase 44 kDa in MS pups. In vivo, the glucocorticoid receptor (GR) antagonist RU486 or ML7 blockade of myosin light chain kinase controlling epithelial cytoskeleton contraction prevented MS-induced IP increase to oral FD4 and BT. In addition, the GR agonist dexamethasone dose-dependently mimicked MS-increase of IP to oral FD4. In contrast, MS effects on IP to oral FD4 and BT were absent at PND20, a model for full-term infant, characterized by a marked drop of IP to FD4 in response to dexamethasone, and decreased GR expression in the colon only compared to PND10 pups. These results show that high intestinal GC responsiveness in a rat model of prematurity defines a vulnerable window for a post-delivery MS, evoking immediate disruption of epithelial integrity in the large intestine, and increasing susceptibility to macromolecule passage and bacteremia.
Collapse
Affiliation(s)
- Nabila Moussaoui
- Intestinal Development, Xenobiotics & Immunotoxicology, Institut National de la Recherche Agronomique (INRA), Research Centre in Food Toxicology (Toxalim), Toulouse, France
- Integrative Toxicology & Metabolism, INRA, Toxalim, Toulouse, France
| | - Viorica Braniste
- Intestinal Development, Xenobiotics & Immunotoxicology, Institut National de la Recherche Agronomique (INRA), Research Centre in Food Toxicology (Toxalim), Toulouse, France
| | | | - Mélissa Gabanou
- Intestinal Development, Xenobiotics & Immunotoxicology, Institut National de la Recherche Agronomique (INRA), Research Centre in Food Toxicology (Toxalim), Toulouse, France
| | - Soraya Sekkal
- Neurogastroenterology & Nutrition, INRA, Toxalim, Toulouse, France
| | - Maiwenn Olier
- Neurogastroenterology & Nutrition, INRA, Toxalim, Toulouse, France
| | | | - Pascal G. P. Martin
- Integrative Toxicology & Metabolism, INRA, Toxalim, Toulouse, France
- GeT-TRiX facility, INRA, Toxalim, Toulouse, France
| | - Eric Houdeau
- Intestinal Development, Xenobiotics & Immunotoxicology, Institut National de la Recherche Agronomique (INRA), Research Centre in Food Toxicology (Toxalim), Toulouse, France
- * E-mail:
| |
Collapse
|
32
|
Zuluaga MJ, Agrati D, Uriarte N, Ferreira A. Social aversive stimuli presented to the mother produce the precocious expression of fear in rat pups. Dev Psychobiol 2014; 56:1187-98. [DOI: 10.1002/dev.21199] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 12/18/2013] [Indexed: 11/05/2022]
Affiliation(s)
- María J. Zuluaga
- Sección Fisiología y Nutrición, Facultad de Ciencias; Universidad de la República (UdelaR); Igua 4225 piso 10 ala sur, CP.11400 Montevideo Uruguay
| | - Daniella Agrati
- Sección Fisiología y Nutrición, Facultad de Ciencias; Universidad de la República (UdelaR); Igua 4225 piso 10 ala sur, CP.11400 Montevideo Uruguay
| | - Natalia Uriarte
- Laboratorio de Neurociencias; Sección Biomatemática; Facultad de Ciencias; UdelaR; Montevideo Uruguay
| | - Annabel Ferreira
- Sección Fisiología y Nutrición, Facultad de Ciencias; Universidad de la República (UdelaR); Igua 4225 piso 10 ala sur, CP.11400 Montevideo Uruguay
| |
Collapse
|
33
|
Daskalakis NP, Diamantopoulou A, Claessens SEF, Remmers E, Tjälve M, Oitzl MS, Champagne DL, de Kloet ER. Early experience of a novel-environment in isolation primes a fearful phenotype characterized by persistent amygdala activation. Psychoneuroendocrinology 2014; 39:39-57. [PMID: 24275003 DOI: 10.1016/j.psyneuen.2013.09.021] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Accepted: 09/19/2013] [Indexed: 02/06/2023]
Abstract
Prolonged maternal separation (MS) activates the neonate's hypothalamus-pituitary-adrenal axis causing elevated basal and stress-induced corticosterone levels that may initiate amygdala-dependent fear learning. Here we test the hypothesis that the adult fearful phenotype is programmed by the pup's stressful experience during prolonged MS rather than by prolonged maternal absence per se. For this purpose, Wistar rat pups were exposed, on postnatal-day (pnd) 3, to: (i) repeated-MS in home-environment (HOME-SEP), 8h-MS daily for three days with the pups remaining together in the home-cage; (ii) repeated-MS in a novel-environment (NOVEL-SEP), with the same separation procedure, but now the pups were individually housed in a novel-environment during the 8h dam's absence; (iii) repeated handling, which consisted of daily brief (15 min instead of 8h) MS in the home-altogether or in a novel-environment individually (HOME-HAN and NOVEL-HAN, respectively); (iv) no-separation/no-handling (NON-SEP/NON-HAN) control condition, in which pups were left undisturbed in their home-cage. Compared to HOME-SEP rats, the NOVEL-SEP rats showed one day after the last MS enhanced stress-induced amygdala c-Fos expression and ACTH-release, despite of reduced adrenal corticosterone secretion. The higher amygdala c-Fos expression, ACTH-release and reduced corticosterone output observed postnatally, persisted into adulthood of the NOVEL-SEP animals. Behaviorally, NOVEL-SEP juvenile rats displayed deficits in social play, had intact spatial memory in the peri-pubertal period and showed more contextual fear memory compared to HOME-SEP in adulthood. Finally, NOVEL-HAN, compared to HOME-HAN, displayed increased stress-induced corticosterone output, no deficits in social play and reduced contextual fear. In conclusion, programming of an adult fearful phenotype linked to amygdala priming develops if pups are repeatedly isolated from peers in a novel-environment, while away from the dam for a prolonged period of time.
Collapse
Affiliation(s)
- Nikolaos P Daskalakis
- Division of Medical Pharmacology, Leiden Academic Center for Drug Research, Leiden University Medical Center, Leiden University, Leiden, The Netherlands; Department of Endocrinology and Metabolism, Leiden University Medical Center, Leiden University, Leiden, The Netherlands; Traumatic Stress Studies Division & Laboratory of Molecular Neuropsychiatry, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, USA; Mental Health Care Center, PTSD Clinical Research Program and Laboratory of Clinical Neuroendocrinology and Neurochemistry, James J. Peters Veterans Affairs Medical Center, Bronx, USA.
| | - Anastasia Diamantopoulou
- Division of Medical Pharmacology, Leiden Academic Center for Drug Research, Leiden University Medical Center, Leiden University, Leiden, The Netherlands; Laboratory of Biology-Biochemistry, Faculty of Nursing, School of Health Sciences, University of Athens, Greece; Department of Psychiatry, Columbia University, New York, NY, USA
| | - Sanne E F Claessens
- Division of Medical Pharmacology, Leiden Academic Center for Drug Research, Leiden University Medical Center, Leiden University, Leiden, The Netherlands
| | - Elisa Remmers
- Division of Medical Pharmacology, Leiden Academic Center for Drug Research, Leiden University Medical Center, Leiden University, Leiden, The Netherlands
| | - Marika Tjälve
- Division of Medical Pharmacology, Leiden Academic Center for Drug Research, Leiden University Medical Center, Leiden University, Leiden, The Netherlands
| | - Melly S Oitzl
- Division of Medical Pharmacology, Leiden Academic Center for Drug Research, Leiden University Medical Center, Leiden University, Leiden, The Netherlands
| | - Danielle L Champagne
- Division of Medical Pharmacology, Leiden Academic Center for Drug Research, Leiden University Medical Center, Leiden University, Leiden, The Netherlands
| | - E Ronald de Kloet
- Division of Medical Pharmacology, Leiden Academic Center for Drug Research, Leiden University Medical Center, Leiden University, Leiden, The Netherlands; Department of Endocrinology and Metabolism, Leiden University Medical Center, Leiden University, Leiden, The Netherlands
| |
Collapse
|
34
|
Maniam J, Antoniadis C, Morris MJ. Early-Life Stress, HPA Axis Adaptation, and Mechanisms Contributing to Later Health Outcomes. Front Endocrinol (Lausanne) 2014; 5:73. [PMID: 24860550 PMCID: PMC4026717 DOI: 10.3389/fendo.2014.00073] [Citation(s) in RCA: 187] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Accepted: 04/28/2014] [Indexed: 12/17/2022] Open
Abstract
Stress activates the hypothalamic-pituitary-adrenal (HPA) axis, which then modulates the degree of adaptation and response to a later stressor. It is known that early-life stress can impact on later health but less is known about how early-life stress impairs HPA axis activity, contributing to maladaptation of the stress-response system. Early-life stress exposure (either prenatally or in the early postnatal period) can impact developmental pathways resulting in lasting structural and regulatory changes that predispose to adulthood disease. Epidemiological, clinical, and experimental studies have demonstrated that early-life stress produces long term hyper-responsiveness to stress with exaggerated circulating glucocorticoids, and enhanced anxiety and depression-like behaviors. Recently, evidence has emerged on early-life stress-induced metabolic derangements, for example hyperinsulinemia and altered insulin sensitivity on exposure to a high energy diet later in life. This draws our attention to the contribution of later environment to disease vulnerability. Early-life stress can alter the expression of genes in peripheral tissues, such as the glucocorticoid receptor and 11-beta hydroxysteroid dehydrogenase (11β-HSD1). We propose that interactions between altered HPA axis activity and liver 11β-HSD1 modulates both tissue and circulating glucocorticoid availability, with adverse metabolic consequences. This review discusses the potential mechanisms underlying early-life stress-induced maladaptation of the HPA axis, and its subsequent effects on energy utilization and expenditure. The effects of positive later environments as a means of ameliorating early-life stress-induced health deficits, and proposed mechanisms underpinning the interaction between early-life stress and subsequent detrimental environmental exposures on metabolic risk will be outlined. Limitations in current methodology linking early-life stress and later health outcomes will also be addressed.
Collapse
Affiliation(s)
- Jayanthi Maniam
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Sydney, NSW, Australia
| | - Christopher Antoniadis
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Sydney, NSW, Australia
| | - Margaret J. Morris
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Sydney, NSW, Australia
- *Correspondence: Margaret J. Morris, Department of Pharmacology, School of Medical Sciences, UNSW Australia, Sydney, NSW 2052, Australia e-mail:
| |
Collapse
|
35
|
Daskalakis NP, Enthoven L, Schoonheere E, de Kloet ER, Oitzl MS. Immediate Effects of Maternal Deprivation on the (Re)Activity of the HPA-Axis Differ in CD1 and C57Bl/6J Mouse Pups. Front Endocrinol (Lausanne) 2014; 5:190. [PMID: 25414695 PMCID: PMC4220727 DOI: 10.3389/fendo.2014.00190] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 10/18/2014] [Indexed: 11/13/2022] Open
Abstract
The postnatal development of the mouse is characterized by a period of hypo-responsiveness of the hypothalamic-pituitary-adrenal (HPA) axis to mild stressors. Maternal deprivation (MD) during this period can disrupt the quiescence of the HPA-axis. The present study examined the influence of strain (outbred CD1 vs. inbred C57BL/6J mice) on some central and peripheral components of the HPA-axis in neonatal mice (5-day-old) in the presence of their mother or after 24 h MD (on postnatal day 4) under basal or mild stressful conditions. In the presence of the dam, adrenal corticosterone (CORT) secretion was low in both mouse strains. Compared to CD1 mice, C57BL/6J had lower CORT levels associated with higher ACTH levels and ACTH/CORT ratio (i.e., lower adrenal sensitivity to ACTH), and higher glucocorticoid receptor (GR) mRNA expression in the paraventricular nucleus. Although MD disinhibited the HPA-axis in both strains as reflected by increased basal CORT and ACTH, we found a strain-dependent pattern. MD increased CORT more in C57BL/6J compared to CD1 mice together with a lower ACTH/CORT ratio (i.e., higher adrenal sensitivity to ACTH), while GR mRNA was no longer different in the two strains. However, this increased adrenal sensitivity in maternally deprived C57BL/6J mice was not reflected in their CORT response to a subsequent novelty stressor, possibly due to an MD-induced ceiling effect in their steroidogenic capacity. In conclusion, the immediate outcome of MD depends on the genetic background of the mother-infant dyad, suggesting that maybe also the outcome in later-life cannot be generalized.
Collapse
Affiliation(s)
- Nikolaos P. Daskalakis
- Division of Medical Pharmacology, Leiden Academic Center for Drug Research, Leiden University, Leiden, Netherlands
- Department of Endocrinology and Metabolism, Leiden University Medical Center, Leiden University, Leiden, Netherlands
- Traumatic Stress Studies Division, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Laboratory of Molecular Neuropsychiatry, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- PTSD Research Program, Mental Health Patient Care Center, James J. Peters Veterans Affairs Medical Center, Bronx, NY, USA
- *Correspondence: Nikolaos P. Daskalakis, Laboratory of Molecular Neuropsychiatry and Traumatic Stress Studies Division, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1668, New York, NY 10029-6574, USA e-mail:
| | - Leo Enthoven
- Division of Medical Pharmacology, Leiden Academic Center for Drug Research, Leiden University, Leiden, Netherlands
| | - Edwige Schoonheere
- Division of Medical Pharmacology, Leiden Academic Center for Drug Research, Leiden University, Leiden, Netherlands
| | - Edo Ronald de Kloet
- Division of Medical Pharmacology, Leiden Academic Center for Drug Research, Leiden University, Leiden, Netherlands
- Department of Endocrinology and Metabolism, Leiden University Medical Center, Leiden University, Leiden, Netherlands
| | - Melly S. Oitzl
- Division of Medical Pharmacology, Leiden Academic Center for Drug Research, Leiden University, Leiden, Netherlands
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
36
|
Strüber N, Strüber D, Roth G. Impact of early adversity on glucocorticoid regulation and later mental disorders. Neurosci Biobehav Rev 2014; 38:17-37. [DOI: 10.1016/j.neubiorev.2013.10.015] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2013] [Revised: 10/04/2013] [Accepted: 10/30/2013] [Indexed: 12/19/2022]
|
37
|
Oliveira S, Fontanelli BAF, Stefanini M, Chuffa L, Teixeira G, Lizarte F, Tirapelli L, Quitete V, Matheus S, Padovani C, Martinez M, Martinez F. Interaction of maternal separation on the UCh rat Cerebellum. Microsc Res Tech 2013; 77:44-51. [DOI: 10.1002/jemt.22311] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 10/23/2013] [Accepted: 10/23/2013] [Indexed: 12/17/2022]
Affiliation(s)
- S.A. Oliveira
- Department of Anatomy, Biosciences Institute; UNESP-Univ. Estadual Paulista; Botucatu SP Brazil
| | - B. A. F. Fontanelli
- Department of Anatomy, Biosciences Institute; UNESP-Univ. Estadual Paulista; Botucatu SP Brazil
| | - M.A. Stefanini
- Department of Morphology and Pathology; UFSCar-Federal University of São Carlos; São Carlos SP Brazil
| | - L.G.A. Chuffa
- Department of Anatomy, Biosciences Institute; UNESP-Univ. Estadual Paulista; Botucatu SP Brazil
| | - G.R. Teixeira
- Department of Anatomy, Biosciences Institute; UNESP-Univ. Estadual Paulista; Botucatu SP Brazil
| | - F.S.N. Lizarte
- Department of Surgery and Anatomy; USP-University of São Paulo; Ribeirão Preto SP Brazil
| | - L.F. Tirapelli
- Department of Surgery and Anatomy; USP-University of São Paulo; Ribeirão Preto SP Brazil
| | - V.H.A. Quitete
- Department of Structural and Functional Biology, Institute of Biology; State University of Campinas (UNICAMP); Campinas, São Paulo Brazil
| | - S.M.M. Matheus
- Department of Anatomy, Biosciences Institute; UNESP-Univ. Estadual Paulista; Botucatu SP Brazil
| | - C.R. Padovani
- Department of Biostatistics, Biosciences Institute; UNESP-Univ. Estadual Paulista; Botucatu SP Brazil
| | - M. Martinez
- Department of Morphology and Pathology; UFSCar-Federal University of São Carlos; São Carlos SP Brazil
| | - F.E. Martinez
- Department of Anatomy, Biosciences Institute; UNESP-Univ. Estadual Paulista; Botucatu SP Brazil
| |
Collapse
|
38
|
Daskalakis NP, Bagot RC, Parker KJ, Vinkers CH, de Kloet ER. The three-hit concept of vulnerability and resilience: toward understanding adaptation to early-life adversity outcome. Psychoneuroendocrinology 2013; 38:1858-73. [PMID: 23838101 PMCID: PMC3773020 DOI: 10.1016/j.psyneuen.2013.06.008] [Citation(s) in RCA: 373] [Impact Index Per Article: 33.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 06/06/2013] [Accepted: 06/07/2013] [Indexed: 01/08/2023]
Abstract
Stressful experiences during early-life can modulate the genetic programming of specific brain circuits underlying emotional and cognitive aspects of behavioral adaptation to stressful experiences later in life. Although this programming effect exerted by experience-related factors is an important determinant of mental health, its outcome depends on cognitive inputs and hence the valence an individual assigns to a given environmental context. From this perspective we will highlight, with studies in rodents, non-human primates and humans, the three-hit concept of vulnerability and resilience to stress-related mental disorders, which is based on gene-environment interactions during critical phases of perinatal and juvenile brain development. The three-hit (i.e., hit-1: genetic predisposition, hit-2: early-life environment, and hit-3: later-life environment) concept accommodates the cumulative stress hypothesis stating that in a given context vulnerability is enhanced when failure to cope with adversity accumulates. Alternatively, the concept also points to the individual's predictive adaptive capacity, which underlies the stress inoculation and match/mismatch hypotheses. The latter hypotheses propose that the experience of relatively mild early-life adversity prepares for the future and promotes resilience to similar challenges in later-life; when a mismatch occurs between early and later-life experience, coping is compromised and vulnerability is enhanced. The three-hit concept is fundamental for understanding how individuals can either be prepared for coping with life to come and remain resilient or are unable to do so and succumb to a stress-related mental disorder, under seemingly identical circumstances.
Collapse
Affiliation(s)
- Nikolaos P. Daskalakis
- Traumatic Stress Studies Division & Laboratory of Molecular Neuropsychiatry, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA,PTSD Clinical Research Program & Laboratory of Clinical Neuroendocrinology and Neurochemistry, James J. Peters Veterans Affairs Medical Center, Bronx, USA,Division of Medical Pharmacology, Leiden/ Amsterdam Center for Drug Research & Leiden University Medical Center, Leiden University, Leiden, The Netherlands,Correspondence: Dr. Nikolaos Daskalakis, Laboratory of Molecular Neuropsychiatry, Mount Sinai School of Medicine, One Gustave L. Levy Place, Box 1668, Annenberg building - Room 22-38, New York, NY 10029-6574, USA,
| | - Rosemary C. Bagot
- Neuroscience Division, Douglas Mental Health University Institute, Montreal, Quebec, Canada,Laboratory of Molecular Psychiatry, Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Karen J. Parker
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, USA
| | - Christiaan H. Vinkers
- Department of Psychiatry, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, Utrecht, the Netherlands,Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences and Rudolf Magnus Institute of Neuroscience, Utrecht University, Utrecht, the Netherlands
| | - E. R. de Kloet
- Division of Medical Pharmacology, Leiden/ Amsterdam Center for Drug Research & Leiden University Medical Center, Leiden University, Leiden, The Netherlands
| |
Collapse
|
39
|
Horii-Hayashi N, Sasagawa T, Matsunaga W, Matsusue Y, Azuma C, Nishi M. Developmental changes in desensitisation of c-Fos expression induced by repeated maternal separation in pre-weaned mice. J Neuroendocrinol 2013; 25:158-67. [PMID: 22913644 PMCID: PMC4038260 DOI: 10.1111/j.1365-2826.2012.02377.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Revised: 08/01/2012] [Accepted: 08/19/2012] [Indexed: 01/17/2023]
Abstract
Early-life stress has long-lasting effects on neuroendocrine and behaviour in adulthood. Maternal separation (MS) is used as a model of early-life stress and daily repeated MS (RMS) for 3 h during the first two postnatal weeks is widely used in rodent studies. However, it is not fully understood whether early-life animals desensitise/habituate to repeated stress. In the present study, we investigated the effects of daily RMS for 3 h and acute/single time MS (SMS) for 3 h on the plasma corticosterone level and c-Fos expression in the brain in mice at different postnatal ages. Mice were subjected to: (i) RMS from postnatal day (PND) 1 to 14 (RMS14); (ii) RMS from PND14 to 21 (RMS21); (iii) SMS on PND14 (SMS14); and (iv) SMS on PND21 (SMS21). Plasma corticosterone and c-Fos expression were examined on the final day in each experiment. The basal corticosterone levels in RMS14 and RMS21 were equal to those in respective age-matched controls. After the final separation, the levels were significantly increased and were comparable with those after SMS14 and SMS21, respectively. Histological analysis indicated that c-Fos expression significantly increased in many brain regions, including the paraventricular nucleus, prefrontal cortex, hippocampus, and basolateral and medial amygdale in both SMS14 and SMS21 mice. However, c-Fos expression in RMS14 mice significantly increased in many regions, whereas such increases were hardly seen in RMS21 mice. These results indicate that repeated early-life stress neither increases basal corticosterone, nor decreases the magnitude of the corticosterone response during the first three postnatal weeks, although desensitisation of c-Fos expression induced by repeated stress is changed during postnatal development.
Collapse
Affiliation(s)
- N Horii-Hayashi
- Department of Anatomy and Cell Biology, Nara
Medical UniversityKashihara, Nara, Japan
| | - T Sasagawa
- Department of Anatomy and Cell Biology, Nara
Medical UniversityKashihara, Nara, Japan
| | - W Matsunaga
- Department of Anatomy and Cell Biology, Nara
Medical UniversityKashihara, Nara, Japan
| | - Y Matsusue
- Department of Anatomy and Cell Biology, Nara
Medical UniversityKashihara, Nara, Japan
- Department of Oral and Maxillofacial Surgery, Nara
Medical UniversityKashihara, Nara, Japan
| | - C Azuma
- Department of Anatomy and Cell Biology, Nara
Medical UniversityKashihara, Nara, Japan
| | - M Nishi
- Department of Anatomy and Cell Biology, Nara
Medical UniversityKashihara, Nara, Japan
- Correspondence to: M. Nishi, Department of Anatomy and Cell Biology, Faculty of
Medicine, Nara Medical University, Kashihara, Nara 634-8521, Japan (e-mail:
)
| |
Collapse
|
40
|
Macrì S, Lanuzza L, Merola G, Ceci C, Gentili S, Valli A, Macchia T, Laviola G. Behavioral responses to acute and sub-chronic administration of the synthetic cannabinoid JWH-018 in adult mice prenatally exposed to corticosterone. Neurotox Res 2013; 24:15-28. [PMID: 23296549 DOI: 10.1007/s12640-012-9371-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Revised: 12/06/2012] [Accepted: 12/21/2012] [Indexed: 12/27/2022]
Abstract
Recent data indicate that both availability and consumption of synthetic and natural psychoactive substances, marketed under the name of "legal highs", has increased. Among them, the aminoalkylindole-derivative JWH-018 is widely distributed due to its capability of binding the cannabinoid receptors CB1 and CB2 thereby mimicking the effects of classical drug agonists. To address whether the behavioral effects of the synthetic compound JWH-018 are similar to those induced by classical cannabinoid agonists, we investigated, in outbred CD1 mice, the consequences of its acute and sub-chronic administration (0, 0.03, 0.1, or 0.3 mg/kg, IP) at the level of body temperature, pain perception, general locomotion, and anxiety. In order to address whether the exposure to precocious stressors-modified individual reactivity to this psychoactive substance, we also investigated its effects in adult mice previously exposed to prenatal stress in the form of corticosterone supplementation in the maternal drinking water (33 or 100 mg/L). In the absence of major effects on motor coordination, JWH-018-reduced body temperature, locomotion and pain reactivity, and increased indices of anxiety. Prenatal corticosterone administration-reduced individual sensitivity to the effects of JWH-018 administration in all the aforementioned parameters. This altered response is not due to variations in JWH-018 metabolism. Present data support the hypothesis that precocious stress may affect, in the long-term, the functional status, and reactivity of the endocannabinoid system.
Collapse
Affiliation(s)
- Simone Macrì
- Section of Behavioural Neuroscience, Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
Epidemiological and clinical studies have shown that children exposed to adverse experiences are at increased risk for the development of depression, anxiety disorders, and posttraumatic stress disorder (PTSD). A history of child abuse and maltreatment increases the likelihood of being subsequently exposed to traumatic events or of developing PTSD as an adult. The brain is highly plastic during early life and encodes acquired information into lasting memories that normally subserve adaptation. Translational studies in rodents showed that enduring sensitization of neuronal and neuroendocrine circuits in response to early life adversity are likely risk factors of life time vulnerability to stress. Hereby, the hypothalamic-pituitary-adrenal (HPA) axis integrates cognitive, behavioral, and emotional responses to early-life stress and can be epigenetically programed during sensitive windows of development. Epigenetic mechanisms, comprising reciprocal regulation of chromatin structure and DNA methylation, are important to establish and maintain sustained, yet potentially reversible, changes in gene transcription. The relevance of these findings for the development of PTSD requires further studies in humans where experience-dependent epigenetic programing can additionally depend on genetic variation in the underlying substrates which may protect from or advance disease development. Overall, identification of early-life stress-associated epigenetic risk markers informing on previous stress history can help to advance early diagnosis, personalized prevention, and timely therapeutic interventions, thus reducing long-term social and health costs.
Collapse
|
42
|
Hendricks S, Ojuka E, Kellaway LA, Mabandla MV, Russell VA. Effect of maternal separation on mitochondrial function and role of exercise in a rat model of Parkinson's disease. Metab Brain Dis 2012; 27:387-92. [PMID: 22527997 PMCID: PMC3402666 DOI: 10.1007/s11011-012-9305-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2012] [Accepted: 03/26/2012] [Indexed: 11/29/2022]
Abstract
Early life stress, such as maternal separation, causes adaptive changes in neural mechanisms that have adverse effects on the neuroplasticity of the brain in adulthood. As a consequence, children who are exposed to stress during development may be predisposed to neurodegenerative disorders in adulthood. A possible mechanism for increased vulnerability to neurodegeneration may be dysfunctional mitochondria. Protection from neurotoxins, such as 6-hydroxydopamine (6-OHDA), has been observed following voluntary exercise. The mechanism of this neuroprotection is not understood and mitochondria may play a role. The purpose of this study was to determine the effects of maternal separation and exercise on mitochondrial function in a rat model of Parkinson's disease. Maternally separated (pups separated from the dam for 3 h per day from postnatal day (P) 2-14) and non-separated rats were placed in individual cages with or without attached running wheels for 1 week prior to unilateral infusion of 6-OHDA (5 μg/4 μl, 0.5 μl/min) into the left medial forebrain bundle at P60. After 2 h recovery, rats were returned to their cages and wheel revolutions recorded for a further 2 weeks. On P72, the rats' motor function was assessed using the forelimb akinesia test. On P74, rats were sacrificed for measurement of mitochondrial function. Exercise increased the respiratory control index (RCI) in the non-lesioned hemisphere of 6-OHDA-lesioned rats. This effect was evident in the striatum of non-separated rats and the prefrontal cortex of maternally separated rats. These results suggest that early life stress may reduce the adaptive response to exercise in the striatum, a major target of dopamine neurons, but not the prefrontal cortex in this model of Parkinson's disease.
Collapse
Affiliation(s)
- Sharief Hendricks
- MRC/UCT Exercise Science and Sports Medicine Research Unit, Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.
| | | | | | | | | |
Collapse
|
43
|
Macrì S, Ceci C, Canese R, Laviola G. Prenatal stress and peripubertal stimulation of the endocannabinoid system differentially regulate emotional responses and brain metabolism in mice. PLoS One 2012; 7:e41821. [PMID: 22848620 PMCID: PMC3405010 DOI: 10.1371/journal.pone.0041821] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Accepted: 06/26/2012] [Indexed: 12/22/2022] Open
Abstract
The central endocannabinoid system (ECS) and the hypothalamic-pituitary-adrenal-axis mediate individual responses to emotionally salient stimuli. Their altered developmental adjustment may relate to the emergence of emotional disturbances. Although environmental influences regulate the individual phenotype throughout the entire lifespan, their effects may result particularly persistent during plastic developmental stages (e.g. prenatal life and adolescence). Here, we investigated whether prenatal stress – in the form of gestational exposure to corticosterone supplemented in the maternal drinking water (100 mg/l) during the last week of pregnancy – combined with a pharmacological stimulation of the ECS during adolescence (daily fatty acid amide hydrolase URB597 i.p. administration - 0.4 mg/kg - between postnatal days 29–38), influenced adult mouse emotional behaviour and brain metabolism measured through in vivo quantitative magnetic resonance spectroscopy. Compared to control mice, URB597-treated subjects showed, in the short-term, reduced locomotion and, in the long term, reduced motivation to execute operant responses to obtain palatable rewards paralleled by reduced levels of inositol and taurine in the prefrontal cortex. Adult mice exposed to prenatal corticosterone showed increased behavioural anxiety and reduced locomotion in the elevated zero maze, and altered brain metabolism (increased glutamate and reduced taurine in the hippocampus; reduced inositol and N-Acetyl-Aspartate in the hypothalamus). Present data further corroborate the view that prenatal stress and pharmacological ECS stimulation during adolescence persistently regulate emotional responses in adulthood. Yet, whilst we hypothesized these factors to be interactive in nature, we observed that the consequences of prenatal corticosterone administration were independent from those of ECS drug-induced stimulation during adolescence.
Collapse
Affiliation(s)
- Simone Macrì
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Roma, Italy.
| | | | | | | |
Collapse
|
44
|
Kember RL, Dempster EL, Lee THA, Schalkwyk LC, Mill J, Fernandes C. Maternal separation is associated with strain-specific responses to stress and epigenetic alterations to Nr3c1, Avp, and Nr4a1 in mouse. Brain Behav 2012; 2:455-67. [PMID: 22950049 PMCID: PMC3432968 DOI: 10.1002/brb3.69] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Revised: 05/03/2012] [Accepted: 05/14/2012] [Indexed: 12/13/2022] Open
Abstract
Stressful events early in life have been widely linked to behavioral phenotypes and have been implicated in the development of psychiatric disorders. Using a maternal separation paradigm, we investigated phenotypic and epigenetic changes following early life stress in two inbred strains of mice, C57BL/6J and DBA/2J. We found an increase in the corticosterone response to stress in male, C57BL/6J mice that had undergone maternal separation compared to controls. In addition, early life stress induced a number of mild but significant behavioral changes, many of which were sex and strain dependent. Following maternal separation anxiety was decreased in males but increased in DBA/2J females, DBA/2J males displayed reduced exploration of a novel object, and baseline activity was altered in males of both strains. Finally, we examined DNA methylation levels in the hippocampus across promoter regions of Nr3c1, Avp, and Nr4a1, and found altered levels at several CpG sites in maternally separated male mice compared to controls. This study contributes to a growing body of recent literature suggesting that epigenetic changes may mediate the impact of early life stress on behavior. In particular, we establish that the phenotypic and epigenetic responses to an adverse environment differ as a function of genetic background.
Collapse
Affiliation(s)
- R. L. Kember
- Social, Genetic and Developmental Psychiatry Centre; Institute of Psychiatry; King's College London; De Crespigny Park; London; UK
| | - E. L. Dempster
- Social, Genetic and Developmental Psychiatry Centre; Institute of Psychiatry; King's College London; De Crespigny Park; London; UK
| | - T. H. A. Lee
- Department of Neuroscience; Institute of Psychiatry; King's College London; De Crespigny Park; London; UK
| | - L. C. Schalkwyk
- Social, Genetic and Developmental Psychiatry Centre; Institute of Psychiatry; King's College London; De Crespigny Park; London; UK
| | - J. Mill
- Social, Genetic and Developmental Psychiatry Centre; Institute of Psychiatry; King's College London; De Crespigny Park; London; UK
| | - C. Fernandes
- Social, Genetic and Developmental Psychiatry Centre; Institute of Psychiatry; King's College London; De Crespigny Park; London; UK
| |
Collapse
|
45
|
Kalinina TS, Shishkina GT, Dygalo NN. Induction of Tyrosine Hydroxylase Gene Expression by Glucocorticoids in the Perinatal Rat Brain is Age-Dependent. Neurochem Res 2012; 37:811-8. [DOI: 10.1007/s11064-011-0676-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Revised: 12/09/2011] [Accepted: 12/15/2011] [Indexed: 11/30/2022]
|
46
|
Krugers HJ, Oomen CA, Gumbs M, Li M, Velzing EH, Joels M, Lucassen PJ. Maternal deprivation and dendritic complexity in the basolateral amygdala. Neuropharmacology 2012; 62:534-7. [DOI: 10.1016/j.neuropharm.2011.09.022] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Revised: 09/16/2011] [Accepted: 09/17/2011] [Indexed: 12/19/2022]
|
47
|
A common and functional mineralocorticoid receptor haplotype enhances optimism and protects against depression in females. Transl Psychiatry 2011; 1:e62. [PMID: 22832354 PMCID: PMC3309494 DOI: 10.1038/tp.2011.59] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Mineralocorticoid (MR) and glucocorticoid receptors (GR) are abundantly expressed in the limbic brain and mediate cortisol effects on the stress-response and behavioral adaptation. Dysregulation of the stress response impairs adaptation and is a risk factor for depression, which is twice as abundant in women than in men. Because of the importance of MR for appraisal processes underlying the initial phase of the stress response we investigated whether specific MR haplotypes were associated with personality traits that predict the risk of depression. We discovered a common gene variant (haplotype 2, frequency ∼0.38) resulting in enhanced MR activity. Haplotype 2 was associated with heightened dispositional optimism in study 1 and with less hopelessness and rumination in study 2. Using data from a large genome-wide association study we then established that haplotype 2 was associated with a lower risk of depression. Interestingly, all effects were restricted to women. We propose that common functional MR haplotypes are important determinants of inter-individual variability in resilience to depression in women by differentially mediating cortisol effects on the stress system.
Collapse
|
48
|
Barbosa Neto JB, Tiba PA, Faturi CB, de Castro-Neto EF, da Graça Naffah-Mazacoratti M, de Jesus Mari J, de Mello MF, Suchecki D. Stress during development alters anxiety-like behavior and hippocampal neurotransmission in male and female rats. Neuropharmacology 2011; 62:518-26. [PMID: 21945413 DOI: 10.1016/j.neuropharm.2011.09.011] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Revised: 08/18/2011] [Accepted: 09/09/2011] [Indexed: 12/18/2022]
Abstract
Epidemiological data indicate that early stress increases vulnerability to psychiatric disorders, including anxiety and depression. In the present study we sought to investigate the long-term behavioral and neurochemical consequences of increased and sustained corticosterone levels induced by a 24 h bout of maternal deprivation (DEP) imposed on postnatal day 11 (DEP11). As adults, animals were exposed to the elevated plus maze for assessment of anxiety-like behavior and corticosterone response to this challenge, or decapitated for determination of monoamines and amino acid neurotransmitters content in the hippocampus by HPLC method. The results showed that DEP11 male and female rats displayed increased time in the central hub of the maze and more risk assessment behavior, reflecting increased anxiety-like behavior; in addition, these animals continuously secreted corticosterone in response to the behavioral test until the latest time-point, e.g., 60 min post-stress. In males, maternal deprivation increased aspartate and glutamate levels and reduced taurine levels compared to non-deprived (NDEP) rats. DEP11 females displayed reduced noradrenaline, aspartate and GABA levels compared to NDEP counterparts. These results indicate that maternal deprivation at 11 days of age produced changes in hippocampal neurotransmission that may mediate the increased anxiety-like behavior observed in male and female deprived rats. This article is part of a Special Issue entitled 'Anxiety and Depression'.
Collapse
|
49
|
Daskalakis NP, Claessens SEF, Laboyrie JJL, Enthoven L, Oitzl MS, Champagne DL, de Kloet ER. The newborn rat's stress system readily habituates to repeated and prolonged maternal separation, while continuing to respond to stressors in context dependent fashion. Horm Behav 2011; 60:165-76. [PMID: 21570400 DOI: 10.1016/j.yhbeh.2011.04.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2011] [Revised: 04/09/2011] [Accepted: 04/17/2011] [Indexed: 10/18/2022]
Abstract
Adrenal corticosterone secretion of newborn mice rapidly desensitizes to repeated maternal absence. The present study investigated the effects of novelty exposure, maternal care and genotype on this phenomenon. Maternal separation (MS) took place on postnatal days (pnd) 3-5. In Wistar rats, the degree of novelty in the MS-environment was varied by exposing pups to: (i) "home separation": pups remained in the home cage; (ii) "novel separation": pups were placed individually in a novel cage. Maternal care was recorded on pnd 1 to 4. To investigate the effect of genotype, we also examined Long Evans in the "home separation" condition. Basal and stress-induced ACTH and corticosterone levels were measured. Adrenal tyrosine hydroxylase (TH) and melanocortin receptor-2 (MCR-2) proteins served as markers for adrenal function. We show, in both rat strains, that the rise in plasma corticosterone induced by a single 8h-MS on pnd 5 was abolished, when this separation procedure had also been performed on pnd 3 and 4. Habituation to maternal absence occurred irrespective of housing conditions. However, pups in the "home separation" condition received less maternal care upon reunion than those placed in the "novel separation". These "home separation" pups appeared more responsive to a subsequent acute novelty-stressor, and their adrenal TH and MCR-2 were higher. Long Evans rats appeared more stress responsive than the Wistars, in the home separation condition. In conclusion, separation environment, maternal care and genotype do not affect adrenal desensitization to repeated 8 h-MS itself, but may modulate the adrenal stress-responsiveness of separated pups.
Collapse
Affiliation(s)
- Nikolaos P Daskalakis
- Division of Medical Pharmacology, Leiden/Amsterdam Center for Drug Research, Leiden University Medical Center, Leiden University, Gorlaeus Laboratories, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
50
|
Zelena D, Barna I, Pintér O, Klausz B, Varga J, Makara GB. Congenital absence of vasopressin and age-dependent changes in ACTH and corticosterone stress responses in rats. Stress 2011; 14:420-30. [PMID: 21438769 DOI: 10.3109/10253890.2011.552991] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The hypothalamic components of the hypothalamo-pituitary-adrenal axis (HPA) are corticotropin-releasing hormone (CRH) and vasopressin. To test the hypothesis that HPA regulation changes with age, we compared ether and bacterial lipopolysaccharide (LPS) injection induced stress reactions in adult and 10-day-old Brattleboro rats, which naturally lack vasopressin owing to mutation of the gene (di/di). The LPS stimulus was used also with V(1b) receptor antagonist pretreatment (SSR149415). In adult di/di or V(1b) pretreated rats, we observed normal pituitary and adrenocortical secretory responses, while in all 10-day-old rats stress-induced serum corticosterone increases were marked, but adrenocorticotropin (ACTH) increases were significantly smaller. Compared to control pups the adenohypophysis of the 10-day-old di/di rats responded normally to CRH, but their adrenal glands were hyper-responsive to ACTH, while in adults there was greater secretion at both levels with no difference between the genotypes. The serum transcortin level was higher in adults than pups, with the di/di pups having higher transcortin levels than controls. Hence, using the same stressors in adults and pups with both a genetic model and pharmacological pretreatment, we have shown that the role of vasopressin in ACTH regulation is more important during the neonatal period than in adulthood. Blunted hypophysial sensitivity to CRH and similar adrenal gland sensitivity to ACTH in the pups compared to adults suggest that hypothalamic factors could be responsible for the neonatal stress hyporesponsive period.
Collapse
Affiliation(s)
- D Zelena
- Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary.
| | | | | | | | | | | |
Collapse
|