1
|
Surber-Cunningham LL, Jimenez LS, Mobo LW, Westrick SE, Fischer EK. Early development of the glucocorticoid stress response in dyeing poison frog tadpoles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.31.596457. [PMID: 38895357 PMCID: PMC11185533 DOI: 10.1101/2024.05.31.596457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
In vertebrates, the glucocorticoid "stress" response (corticosterone or cortisol) through the hypothalamic-pituitary-adrenal (HPA) axis influences many essential functions, including behavior, metabolism, immunity, and ontogenetic transitions. During development, stress responses can be adaptive if they facilitate antipredator behavior and modulate developmental speed to adjust to environmental conditions; however, these same responses can be maladaptive when energetic costs become too high and developmental speed trades-off with size and health at maturity. Thus, the timing of HPA-axis development may be aligned with specific developmental challenges and opportunities presented by a species' life history strategy. In anurans (frogs and toads), corticosterone plays critical roles in development and behavior, and concentrations can fluctuate in response to environmental stressors. Given the role of corticosterone in ontogenetic changes and behaviors, we studied the development of the HPA axis in tadpoles of the dyeing poison frog ( Dendrobates tinctorius ), a species with a unique life history that includes transport to water after hatching on land and aggressive and cannibalistic behavior. We measured the excretion rate and whole-body concentration of corticosterone and the corticosterone response to adrenocorticotropic hormone (ACTH) in free-swimming tadpoles after transport and throughout metamorphosis. We found no significant differences across development in excretion rates or whole-body concentration of corticosterone, nor corticosterone response to ACTH, indicating that that the glucocorticoid response develops early in ontogeny. This pattern differs from those in other species of tadpoles, suggesting the unique ecological pressures faced by D. tinctorius have shaped the development of its HPA axis. More broadly, this study illustrates how life history strategies and tradeoffs impact the timing of the HPA axis development.
Collapse
|
2
|
Li Y, Liu ZW, Santana GM, Capaz AM, Doumazane E, Gao XB, Renier N, Dietrich MO. Neurons for infant social behaviors in the mouse zona incerta. Science 2024; 385:409-416. [PMID: 39052814 DOI: 10.1126/science.adk7411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 06/07/2024] [Indexed: 07/27/2024]
Abstract
Understanding the neural basis of infant social behaviors is crucial for elucidating the mechanisms of early social and emotional development. In this work, we report a specific population of somatostatin-expressing neurons in the zona incerta (ZISST) of preweaning mice that responds dynamically to social interactions, particularly those with their mother. Bidirectional neural activity manipulations in pups revealed that widespread connectivity of preweaning ZISST neurons to sensory, emotional, and cognitive brain centers mediates two key adaptive functions associated with maternal presence: the reduction of behavior distress and the facilitation of learning. These findings reveal a population of neurons in the infant mouse brain that coordinate the positive effects of the relationship with the mother on an infant's behavior and physiology.
Collapse
Affiliation(s)
- Yuexuan Li
- Laboratory of Physiology of Behavior, Department of Comparative Medicine, School of Medicine, Yale University, New Haven, CT 06520, USA
- Department of Comparative Medicine, School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Zhong-Wu Liu
- Department of Comparative Medicine, School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Gustavo M Santana
- Laboratory of Physiology of Behavior, Department of Comparative Medicine, School of Medicine, Yale University, New Haven, CT 06520, USA
- Department of Neuroscience, School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Ana Marta Capaz
- Laboratoire de Plasticité Structurale, Sorbonne Université, ICM Paris Brain Institute, INSERM U1127, CNRS UMR7225, AP-HP, 75013 Paris, France
| | - Etienne Doumazane
- Laboratoire de Plasticité Structurale, Sorbonne Université, ICM Paris Brain Institute, INSERM U1127, CNRS UMR7225, AP-HP, 75013 Paris, France
| | - Xiao-Bing Gao
- Department of Comparative Medicine, School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Nicolas Renier
- Laboratoire de Plasticité Structurale, Sorbonne Université, ICM Paris Brain Institute, INSERM U1127, CNRS UMR7225, AP-HP, 75013 Paris, France
| | - Marcelo O Dietrich
- Laboratory of Physiology of Behavior, Department of Comparative Medicine, School of Medicine, Yale University, New Haven, CT 06520, USA
- Department of Comparative Medicine, School of Medicine, Yale University, New Haven, CT 06520, USA
- Department of Neuroscience, School of Medicine, Yale University, New Haven, CT 06520, USA
| |
Collapse
|
3
|
Paul B, Buchholz DR. Minireview: Glucocorticoid-Leptin Crosstalk: Role of Glucocorticoid-Leptin Counterregulation in Metabolic Homeostasis and Normal Development. Integr Comp Biol 2023; 63:1127-1139. [PMID: 37708034 DOI: 10.1093/icb/icad119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 08/16/2023] [Accepted: 08/30/2023] [Indexed: 09/16/2023] Open
Abstract
Glucocorticoids and leptin are two important hormones that regulate metabolic homeostasis by controlling appetite and energy expenditure in adult mammals. Also, glucocorticoids and leptin strongly counterregulate each other, such that chronic stress-induced glucocorticoids upregulate the production of leptin and leptin suppresses glucocorticoid production directly via action on endocrine organs and indirectly via action on food intake. Altered glucocorticoid or leptin levels during development can impair organ development and increase the risk of chronic diseases in adults, but there are limited studies depicting the significance of glucocorticoid-leptin interaction during development and its impact on developmental programming. In mammals, leptin-induced suppression of glucocorticoid production is critical during development, where leptin prevents stress-induced glucocorticoid production by inducing a period of short-hyporesponsiveness when the adrenal glands fail to respond to certain mild to moderate stressors. Conversely, reduced or absent leptin signaling increases glucocorticoid levels beyond what is appropriate for normal organogenesis. The counterregulatory interactions between leptin and glucocorticoids suggest the potential significant involvement of leptin in disorders that occur from stress during development.
Collapse
Affiliation(s)
- Bidisha Paul
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Daniel R Buchholz
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH 45221, USA
| |
Collapse
|
4
|
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
|
5
|
Parker KN, Donovan MH, Smith K, Noble-Haeusslein LJ. Traumatic Injury to the Developing Brain: Emerging Relationship to Early Life Stress. Front Neurol 2021; 12:708800. [PMID: 34484104 PMCID: PMC8416304 DOI: 10.3389/fneur.2021.708800] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/22/2021] [Indexed: 12/01/2022] Open
Abstract
Despite the high incidence of brain injuries in children, we have yet to fully understand the unique vulnerability of a young brain to an injury and key determinants of long-term recovery. Here we consider how early life stress may influence recovery after an early age brain injury. Studies of early life stress alone reveal persistent structural and functional impairments at adulthood. We consider the interacting pathologies imposed by early life stress and subsequent brain injuries during early brain development as well as at adulthood. This review outlines how early life stress primes the immune cells of the brain and periphery to elicit a heightened response to injury. While the focus of this review is on early age traumatic brain injuries, there is also a consideration of preclinical models of neonatal hypoxia and stroke, as each further speaks to the vulnerability of the brain and reinforces those characteristics that are common across each of these injuries. Lastly, we identify a common mechanistic trend; namely, early life stress worsens outcomes independent of its temporal proximity to a brain injury.
Collapse
Affiliation(s)
- Kaila N. Parker
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX, United States
- Department of Psychology, Behavioral Neuroscience, College of Liberal Arts, University of Texas at Austin, Austin, TX, United States
| | - Michael H. Donovan
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX, United States
- Department of Psychology, Behavioral Neuroscience, College of Liberal Arts, University of Texas at Austin, Austin, TX, United States
| | - Kylee Smith
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX, United States
- Department of Psychology, Behavioral Neuroscience, College of Liberal Arts, University of Texas at Austin, Austin, TX, United States
| | - Linda J. Noble-Haeusslein
- Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX, United States
- Department of Psychology, Behavioral Neuroscience, College of Liberal Arts, University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
6
|
Sprowles JLN, Vorhees CV, Williams MT. Impact of preweaning stress on long-term neurobehavioral outcomes in Sprague-Dawley rats: Differential effects of barren cage rearing, pup isolation, and the combination. Neurotoxicol Teratol 2021; 84:106956. [PMID: 33524508 DOI: 10.1016/j.ntt.2021.106956] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 01/06/2021] [Accepted: 01/17/2021] [Indexed: 12/23/2022]
Abstract
Two developmental stressors were compared in preweaning rats exposed to either one stressor or both. Stressors were barren cage rearing or maternal separation (pup isolation). 40 gravid Sprague-Dawley CD/IGS rats were randomly assigned to two cage conditions: standard (Std) cage or barren cage (Bar), 20 litters/condition throughout gestation and lactation. After delivery, litters were randomly culled to 4 males and 4 females. The second stressor was maternal separation: Two male/female pairs per litter were isolated from their dam 4 h/day (Iso) and two pairs were not (Norm). Hence, there were 4 conditions: Std-Norm, Std-Iso, Bar-Norm, and Bar-Iso. One pair/litter/stress condition received the following: elevated zero-maze (EZM), open-field, swim channel, Cincinnati water maze, conditioned fear, and open-field with methamphetamine challenge. The second pair/litter/condition received the light-dark test, swim channel, Morris water maze, forced swim, and EZM with diazepam challenge. Barren rearing reduced EZM time-in-open, whereas isolation rearing reduced open-field activity in males and increased it in females. Effects on straight channel swimming were minor. In the Cincinnati water maze test of egocentric learning, isolation rearing increased errors whereas barren cage housing reduced errors in combination with normal rearing. Barren cage with maternal separation (pup isolation) increased Cincinnati water maze escape latency but not errors. Barren cage housing reduced hyperactivity in response to methamphetamine. Isolation rearing increased time in open in the EZM after diazepam challenge. Trends were seen in the Morris water maze. These suggested that barren cage and isolation rearing in combination reduced latency on acquisition on days 1 and 2 in males, whereas females had increased latency on days 2 and 3. Combined exposure to two developmental stressors did not induce additive or synergistic effects, however the data show that these stressors had long-term effects with some evidence that the combination of both caused effects when either stressor alone did not, but synergism was not observed.
Collapse
Affiliation(s)
- Jenna L N Sprowles
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - Charles V Vorhees
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA; Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, USA.
| | - Michael T Williams
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA; Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, USA.
| |
Collapse
|
7
|
The HPA axis dysregulation in severe mental illness: Can we shift the blame to gut microbiota? Prog Neuropsychopharmacol Biol Psychiatry 2020; 102:109951. [PMID: 32335265 DOI: 10.1016/j.pnpbp.2020.109951] [Citation(s) in RCA: 167] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/15/2020] [Accepted: 04/21/2020] [Indexed: 12/12/2022]
Abstract
Accumulating evidence indicates that patients with severe mental disorders, including major depression, bipolar disorder and schizophrenia present with various alterations of the gut microbiota and increased intestinal permeability. In addition, the hypothalamic-pituitary-adrenal (HPA) axis dysregulation and subclinical inflammation have been reported in this group of patients. Although it has been found that the HPA axis dysregulation appears as a consequence of psychosocial stress, especially traumatic life events, the exact mechanisms of this observation remain unclear. Animal model studies have unraveled several mechanisms linking the gut microbiota with the HPA axis dysfunction. Indeed, the gut microbiota can activate the HPA axis through several mediators that cross the blood-brain barrier and include microbial antigens, cytokines and prostaglandins. There is also evidence that various microbial species can affect ileal corticosterone production that may impact the activity of the HPA axis. However, some metabolites released by various microbes, e.g., short-chain fatty acids, can attenuate the HPA axis response. Moreover, several bacteria release neurotransmitters that can directly interact with vagal afferents. It has been postulated that the HPA axis activation can impact the gut microbiota and intestinal permeability. In this article, we discuss various mechanisms linking the gut microbiota with the HPA axis activity and summarize current evidence for a cross-talk between the gut-brain axis and the HPA axis from studies of patients with mood and psychotic disorders. Finally, we show potential clinical implications that can arise from future studies investigating the HPA axis activity with respect to the gut microbiota in severe mental disorders.
Collapse
|
8
|
Orefice LL. Peripheral Somatosensory Neuron Dysfunction: Emerging Roles in Autism Spectrum Disorders. Neuroscience 2020; 445:120-129. [PMID: 32035119 DOI: 10.1016/j.neuroscience.2020.01.039] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 01/27/2020] [Accepted: 01/28/2020] [Indexed: 12/26/2022]
Abstract
Alterations in somatosensory (touch and pain) behaviors are highly prevalent among people with autism spectrum disorders (ASDs). However, the neural mechanisms underlying abnormal touch and pain-related behaviors in ASDs and how altered somatosensory reactivity might contribute to ASD pathogenesis has not been well studied. Here, we provide a brief review of somatosensory alterations observed in people with ASDs and recent evidence from animal models that implicates peripheral neurons as a locus of dysfunction for somatosensory abnormalities in ASDs. Lastly, we describe current efforts to understand how altered peripheral sensory neuron dysfunction may impact brain development and complex behaviors in ASD models, and whether targeting peripheral somatosensory neurons to improve their function might also improve related ASD phenotypes.
Collapse
Affiliation(s)
- Lauren L Orefice
- Department of Molecular Biology, Massachusetts General Hospital and Department of Genetics, Harvard Medical School, 185 Cambridge Street, Boston, MA 02114, USA.
| |
Collapse
|
9
|
Pucci M, Micioni Di Bonaventura MV, Wille-Bille A, Fernández MS, Maccarrone M, Pautassi RM, Cifani C, D’Addario C. Environmental stressors and alcoholism development: Focus on molecular targets and their epigenetic regulation. Neurosci Biobehav Rev 2019; 106:165-181. [DOI: 10.1016/j.neubiorev.2018.07.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 06/13/2018] [Accepted: 07/09/2018] [Indexed: 01/17/2023]
|
10
|
Candemir E, Post A, Dischinger US, Palme R, Slattery DA, O'Leary A, Reif A. Limited effects of early life manipulations on sex-specific gene expression and behavior in adulthood. Behav Brain Res 2019; 369:111927. [PMID: 31034851 DOI: 10.1016/j.bbr.2019.111927] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 04/23/2019] [Accepted: 04/25/2019] [Indexed: 01/19/2023]
Abstract
Exposure to childhood adversity is associated with increased vulnerability to stress-related disorders in adulthood which has been replicated in rodent stress models, whereas environmental enrichment has been suggested to have beneficial effects. However, the exact neurobiological mechanisms underlying these environment influences on adult brain and behavior are not well understood. Therefore, we investigated the long-term effects of maternal separation (MS) or environmental enrichment (EE) in male and female CD1 mice. We found clear sex-specific effects, but limited influence of environmental manipulations, on adult behavior, fecal corticosterone metabolite (FCM) levels and stress- and plasticity related gene expression in discrete brain regions. In detail, adult females displayed higher locomotor activity and FCM levels compared to males and EE resulted in attenuation in both measures, but only in females. There were no sex- or postnatal manipulation-dependent differences in anxiety-related behaviors in either sex. Gene expression analyses revealed that adult males showed higher Fkbp5 mRNA levels in hippocampus, hypothalamus and raphe nuclei, and higher hippocampal Nos1 levels. Interestingly, MS elevated Nos1 levels in hippocampus but reduced Fkbp5 expression in hypothalamus of males. Finally, we also found higher Maoa expression in the hypothalamus of adult females, however no differences were observed in the expression levels of Bdnf, Crhr1, Nr3c1 and Htr1a. Our findings further contribute to sex-dependent differences in behavior, corticosterone and gene expression and reveal that the effects of postnatal manipulations on these parameters in outbred CD1 mice are limited.
Collapse
Affiliation(s)
- Esin Candemir
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Antonia Post
- Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital of Würzburg, Germany
| | - Ulrich Severin Dischinger
- Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital of Würzburg, Germany
| | - Rupert Palme
- Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria
| | - David A Slattery
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Aet O'Leary
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt am Main, Germany; Department of Neuropsychopharmacology, Institute of Psychology, University of Tartu, Tartu, Estonia
| | - Andreas Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Frankfurt am Main, Germany.
| |
Collapse
|
11
|
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: 7.8] [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
|
12
|
Reshetnikov VV, Lepeshko AA, Ryabushkina YA, Studenikina AA, Merkulova TI, Bondar NP. The Long-Term Effects of Early Postnatal Stress on Cognitive Abilities and Expression of Genes of the Glutamatergic System in Mice. NEUROCHEM J+ 2018. [DOI: 10.1134/s1819712418020095] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
13
|
On the Developmental Timing of Stress: Delineating Sex-Specific Effects of Stress across Development on Adult Behavior. Brain Sci 2018; 8:brainsci8070121. [PMID: 29966252 PMCID: PMC6071226 DOI: 10.3390/brainsci8070121] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 06/26/2018] [Accepted: 06/27/2018] [Indexed: 12/11/2022] Open
Abstract
Stress, and the chronic overactivation of major stress hormones, is associated with several neuropsychiatric disorders. However, clinical literature on the exact role of stress either as a causative, triggering, or modulatory factor to mental illness remains unclear. We suggest that the impact of stress on the brain and behavior is heavily dependent on the developmental timing at which the stress has occurred, and as such, this may contribute to the overall variability reported on the association of stress and mental illness. Here, animal models provide a way to comprehensively assess the temporal impact of stress on behavior in a controlled manner. This review particularly focuses on the long-term impact of stress on behavior in various rodent stress models at three major developmental time points: early life, adolescence, and adulthood. We characterize the various stressor paradigms into physical, social, and pharmacological, and discuss commonalities and differences observed across these various stress-inducing methods. In addition, we discuss here how sex can influence the impact of stress at various developmental time points. We conclude here that early postnatal life and adolescence represent particular periods of vulnerability, but that stress exposure during early life can sometimes lead to resilience, particularly to fear-potentiated memories. In the adult brain, while shorter periods of stress tended to enhance spatial memory, longer periods caused impairments. Overall, males tended to be more vulnerable to the long-term effects of early life and adolescent stress, albeit very few studies incorporate both sexes, and further well-powered sex comparisons are needed.
Collapse
|
14
|
Reshetnikov V, Studenikina A, Ryabushkina J, Merkulova T, Bondar N. The impact of early-life stress on the expression of HPA-associated genes in the adult murine brain. BEHAVIOUR 2018. [DOI: 10.1163/1568539x-00003482] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Abstract
Early life is an important period for the development of the nervous system and for the programming of behavioural phenotypes in adulthood. In our study, two types of early-life stress were used: prolonged separation of pups from their mothers (for 3 h/day, maternal separation (MS)) and brief separation (for 15 min/day, handling (HD)). We analysed the effects of early-life stress on behaviour and the expression of HPA-associated genes in the hypothalamus, hippocampus, and frontal cortex of male mice. Adult mice in the MS group demonstrated reduced locomotor activity and deficiencies in spatial long-term memory, while the HD showed no significant changes. Additionally, early-life MS resulted in reduced hippocampal Crhr1 mRNA, increased MR/GR mRNA in the hippocampus and hypothalamus. Both groups, HD and MS, showed increased Avp mRNA in the hypothalamus. Thus, prolonged maternal separation but not brief leads to adverse behavioural changes and influences the expression of HPA-associated genes in a brain region-specific manner.
Collapse
Affiliation(s)
- V.V. Reshetnikov
- aLaboratory of Gene Expression Regulation, Institute of Cytology and Genetics, SB RAS, Novosibirsk, Russia
| | - A.A. Studenikina
- aLaboratory of Gene Expression Regulation, Institute of Cytology and Genetics, SB RAS, Novosibirsk, Russia
- bNovosibirsk State Medical University, Novosibirsk, Russia
| | - J.A. Ryabushkina
- aLaboratory of Gene Expression Regulation, Institute of Cytology and Genetics, SB RAS, Novosibirsk, Russia
- cNovosibirsk State University, Novosibirsk, Russia
| | - T.I. Merkulova
- aLaboratory of Gene Expression Regulation, Institute of Cytology and Genetics, SB RAS, Novosibirsk, Russia
- cNovosibirsk State University, Novosibirsk, Russia
| | - N.P. Bondar
- aLaboratory of Gene Expression Regulation, Institute of Cytology and Genetics, SB RAS, Novosibirsk, Russia
- cNovosibirsk State University, Novosibirsk, Russia
| |
Collapse
|
15
|
Toyoda A. Social defeat models in animal science: What we have learned from rodent models. Anim Sci J 2017; 88:944-952. [PMID: 28436163 PMCID: PMC5518448 DOI: 10.1111/asj.12809] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Accepted: 02/13/2017] [Indexed: 12/30/2022]
Abstract
Studies on stress and its impacts on animals are very important in many fields of science, including animal science, because various stresses influence animal production and animal welfare. In particular, the social stresses within animal groups have profound impact on animals, with the potential to induce abnormal behaviors and health problems. In humans, social stress induces several health problems, including psychiatric disorders. In animal stress models, social defeat models are well characterized and used in various research fields, particularly in studies concerning mental disorders. Recently, we have focused on behavior, nutrition and metabolism in rodent models of social defeat to elucidate how social stresses affect animals. In this review, recent significant progress in studies related to animal social defeat models are described. In the field of animal science, these stress models may contribute to advances in the development of functional foods and in the management of animal welfare.
Collapse
Affiliation(s)
- Atsushi Toyoda
- College of Agriculture, Ibaraki University, Ami, Ibaraki, Japan.,Ibaraki University Cooperation between Agriculture and Medical Science (IUCAM), Ami, Ibaraki, Japan.,United Graduate School of Agricultural Science, Tokyo University of Agriculture and Technology, Fuchu-city, Tokyo, Japan
| |
Collapse
|
16
|
Ochoa-Repáraz J, Kasper LH. The Second Brain: Is the Gut Microbiota a Link Between Obesity and Central Nervous System Disorders? Curr Obes Rep 2016; 5:51-64. [PMID: 26865085 PMCID: PMC4798912 DOI: 10.1007/s13679-016-0191-1] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The gut-brain axis is a bi-directional integrated system composed by immune, endocrine, and neuronal components by which the gap between the gut microbiota and the brain is significantly impacted. An increasing number of different gut microbial species are now postulated to regulate brain function in health and disease. The westernized diet is hypothesized to be the cause of the current obesity levels in many countries, a major socio-economical health problem. Experimental and epidemiological evidence suggest that the gut microbiota is responsible for significant immunologic, neuronal, and endocrine changes that lead to obesity. We hypothesize that the gut microbiota, and changes associated with diet, affect the gut-brain axis and may possibly contribute to the development of mental illness. In this review, we discuss the links between diet, gut dysbiosis, obesity, and immunologic and neurologic diseases that impact brain function and behavior.
Collapse
Affiliation(s)
| | - Lloyd H. Kasper
- Department of Microbiology and Immunology, Remsen Building, Room 132A, Geisel School of Medicine at Dartmouth College, Hanover NH 03755, United States, Phone: (603) 653-9909
| |
Collapse
|
17
|
Ghaisas S, Maher J, Kanthasamy A. Gut microbiome in health and disease: Linking the microbiome-gut-brain axis and environmental factors in the pathogenesis of systemic and neurodegenerative diseases. Pharmacol Ther 2015; 158:52-62. [PMID: 26627987 DOI: 10.1016/j.pharmthera.2015.11.012] [Citation(s) in RCA: 341] [Impact Index Per Article: 34.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The gut microbiome comprises the collective genome of the trillions of microorganisms residing in our gastrointestinal ecosystem. The interaction between the host and its gut microbiome is a complex relationship whose manipulation could prove critical to preventing or treating not only various gut disorders, like irritable bowel syndrome (IBS) and ulcerative colitis (UC), but also central nervous system (CNS) disorders, such as Alzheimer's and Parkinson's diseases. The purpose of this review is to summarize what is known about the gut microbiome, how it is connected to the development of disease and to identify the bacterial and biochemical targets that should be the focus of future research. Understanding the mechanisms behind the activity and proliferation of the gut microbiome will provide us new insights that may pave the way for novel therapeutic strategies.
Collapse
Affiliation(s)
- Shivani Ghaisas
- Parkinson's Disorder Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA
| | - Joshua Maher
- Parkinson's Disorder Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA
| | - Anumantha Kanthasamy
- Parkinson's Disorder Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA.
| |
Collapse
|
18
|
Ghosal S, Nunley A, Mahbod P, Lewis AG, Smith EP, Tong J, D'Alessio DA, Herman JP. Mouse handling limits the impact of stress on metabolic endpoints. Physiol Behav 2015; 150:31-7. [PMID: 26079207 DOI: 10.1016/j.physbeh.2015.06.021] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 05/28/2015] [Accepted: 06/11/2015] [Indexed: 01/08/2023]
Abstract
Studies focused on end-points that are confounded by stress are best performed under minimally stressful conditions. The objective of this study was to demonstrate the impact of handling designed to reduce animal stress on measurements of glucose tolerance. A cohort of mice (CD1.C57BL/6) naïve to any specific handling was subjected to either a previously described "cup" handling method, or a "tail-picked" method in which the animals were picked up by the tail (as is common for metabolic studies). Following training, an elevated plus maze (EPM) test was performed followed by measurement of blood glucose and plasma corticosterone. A second cohort (CD1.C57BL/6) was rendered obese by exposure to a high fat diet, handled with either the tail-picked or cup method and subjected to an intraperitoneal glucose tolerance test. A third cohort of C57BL/6 mice was exposed to a cup regimen that included a component of massage and was subjected to tests of anxiety-like behavior, glucose homeostasis, and corticosterone secretion. We found that the cup mice showed reduced anxiety-like behaviors in the EPM coupled with a reduction in blood glucose levels compared to mice handled by the tail-picked method. Additionally, cup mice on the high fat diet exhibited improved glucose tolerance compared to tail-picked controls. Finally, we found that the cup/massage group showed lower glucose levels following an overnight fast, and decreased anxiety-like behaviors associated with lower stress-induced plasma corticosterone concentration compared to tail-picked controls. These data demonstrate that application of handling methods that reduce anxiety-like behaviors in mice mitigates the confounding contribution of stress to interpretation of metabolic endpoints (such as glucose tolerance).
Collapse
Affiliation(s)
- Sriparna Ghosal
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH 45237, United States.
| | - Amanda Nunley
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45237, United States
| | - Parinaz Mahbod
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45237, United States
| | - Alfor G Lewis
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45237, United States
| | - Eric P Smith
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45237, United States
| | - Jenny Tong
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45237, United States
| | - David A D'Alessio
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH 45237, United States
| | - James P Herman
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH 45237, United States
| |
Collapse
|
19
|
Sudo N. Microbiome, HPA axis and production of endocrine hormones in the gut. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 817:177-94. [PMID: 24997034 DOI: 10.1007/978-1-4939-0897-4_8] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Recent accumulating evidence indicates that the gut microbiome can affect the development and regulation of the hypothalamic-pituitary-adrenal axis and behavior, with central integrative systems being crucial in the successful physiological adaptation of the organism to external stressor. In contrast, host-derived hormones increase the bacterial proliferative capacity and pathogenicity. In the gut lumen, this type of cross-talk between microorganisms and the host is presumed to be performed continually through various kinds of luminal molecules, as numerous types of bacteria and host cells are in close proximity in the gastrointestinal tract of mammals.We herein focus on bidirectional signaling between the gut microbiome and the host in terms of commensal microbiota affecting the hypothalamic-pituitary-adrenal HPA axis response and behaviors and further discuss the role of gut luminal catecholamines and γ-aminobutyric acid, both of which are presumed to be involved in this signaling.
Collapse
Affiliation(s)
- Nobuyuki Sudo
- Department of Psychosomatic Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan,
| |
Collapse
|
20
|
Maccari S, Krugers HJ, Morley-Fletcher S, Szyf M, Brunton PJ. The consequences of early-life adversity: neurobiological, behavioural and epigenetic adaptations. J Neuroendocrinol 2014; 26:707-23. [PMID: 25039443 DOI: 10.1111/jne.12175] [Citation(s) in RCA: 256] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 07/09/2014] [Accepted: 07/10/2014] [Indexed: 12/12/2022]
Abstract
During the perinatal period, the brain is particularly sensitive to remodelling by environmental factors. Adverse early-life experiences, such as stress exposure or suboptimal maternal care, can have long-lasting detrimental consequences for an individual. This phenomenon is often referred to as 'early-life programming' and is associated with an increased risk of disease. Typically, rodents exposed to prenatal stress or postnatal maternal deprivation display enhanced neuroendocrine responses to stress, increased levels of anxiety and depressive-like behaviours, and cognitive impairments. Some of the phenotypes observed in these models of early-life adversity are likely to share common neurobiological mechanisms. For example, there is evidence for impaired glucocorticoid negative-feedback control of the hypothalamic-pituitary-adrenal axis, altered glutamate neurotransmission and reduced hippocampal neurogenesis in both prenatally stressed rats and rats that experienced deficient maternal care. The possible mechanisms through which maternal stress during pregnancy may be transmitted to the offspring are reviewed, with special consideration given to altered maternal behaviour postpartum. We also discuss what is known about the neurobiological and epigenetic mechanisms that underpin early-life programming of the neonatal brain in the first generation and subsequent generations, with a view to abrogating programming effects and potentially identifying new therapeutic targets for the treatment of stress-related disorders and cognitive impairment.
Collapse
Affiliation(s)
- S Maccari
- LIA, International Laboratory Associated, UMR 8576 CNRS Neural plasticity Team, University of Lille 1, France and Sapienza University of Rome, IRCCS NEUROMED, Italy
| | | | | | | | | |
Collapse
|
21
|
Sotnikov S, Wittmann A, Bunck M, Bauer S, Deussing J, Schmidt M, Touma C, Landgraf R, Czibere L. Blunted HPA axis reactivity reveals glucocorticoid system dysbalance in a mouse model of high anxiety-related behavior. Psychoneuroendocrinology 2014; 48:41-51. [PMID: 24995583 DOI: 10.1016/j.psyneuen.2014.06.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 05/19/2014] [Accepted: 06/10/2014] [Indexed: 01/15/2023]
Abstract
Depression and anxiety disorders are often characterized by altered hypothalamic-pituitary-adrenal (HPA) axis re-/activity. However, the presence of a molecular link between dysbalanced neuroendocrine regulation and psychopathologies is not yet fully established. Earlier, we reported that high (HAB), normal (NAB) and low (LAB) anxiety-related behavior mice express divergent anxiety-related and passive/active coping phenotypes. Here, we studied mechanisms that might contribute to the different HPA axis reactivity observed in HAB, NAB and LAB mice and their involvement in the regulation of anxiety-related behavior and passive/active coping style. We found that HAB mice respond with significantly reduced corticosterone (CORT) secretion to an acute stressful stimulus and a blunted response in the Dex/CRH test compared to NAB and LAB mice. At the molecular level, higher expression of the glucocorticoid receptor (GR/Nr3c1) and decreased corticotropin-releasing hormone receptor 1 (CRHR1) expression were observed in the pituitary of HAB mice. We further analyzed whether these stress mediators differed between the HAB, NAB and LAB lines in limbic system-associated brain regions and whether their interplay contributes to the phenotype. Interestingly, not only in the pituitary but also in almost all brain regions investigated, GR expression was significantly higher in HAB mice. In contrast, the amount of CORT in the brain structures analyzed was significantly lower in these animals. The expression of CRHR1 varied in the prefrontal cortex only. Since glucocorticoids regulate both GR and CRHR1, we treated HAB and NAB mice chronically with CORT. After 6 weeks of administration, reduced anxiety- and depression-like behaviors were observed in HAB mice, whereas increased anxiety was found in NABs. In both groups, GR, but not CRHR1, were significantly reduced. Taken together, our study proposes HAB mice as an animal model of simultaneous features of increased anxiety-related and depression-like behaviors with blunted HPA axis reactivity suggesting a dysregulated GR/CORT system as one key mechanism behind their phenotype.
Collapse
Affiliation(s)
- Sergey Sotnikov
- Max Planck Institute of Psychiatry, 80804 Munich, Germany; Department of Normal Physiology, Sechenov First Moscow State Medical University, 119991 Moscow, Russia.
| | - Anke Wittmann
- Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Mirjam Bunck
- Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Sabrina Bauer
- Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Jan Deussing
- Max Planck Institute of Psychiatry, 80804 Munich, Germany; Clinical Cooperation Group Molecular Neurogenetics, Institute of Developmental Genetics, Helmholtz Center Munich, 85764 Neuherberg, Germany
| | | | - Chadi Touma
- Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | | | - Ludwig Czibere
- Max Planck Institute of Psychiatry, 80804 Munich, Germany
| |
Collapse
|
22
|
Noguchi KK. Glucocorticoid Induced Cerebellar Toxicity in the Developing Neonate: Implications for Glucocorticoid Therapy during Bronchopulmonary Dysplasia. Cells 2014; 3:36-52. [PMID: 24501683 PMCID: PMC3910303 DOI: 10.3390/cells3010036] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Prematurely born infants commonly suffer respiratory dysfunction due to the immature state of their lungs. As a result, clinicians often administer glucocorticoid (GC) therapy to accelerate lung maturation and reduce inflammation. Unfortunately, several studies have found GC therapy can also produce neuromotor/cognitive deficits and selectively stunt the cerebellum. However, despite its continued use, relatively little is known about how exposure to this hormone might produce neurodevelopmental deficits. In this review, we use rodent and human research to provide evidence that GC therapy may disrupt cerebellar development through the rapid induction of apoptosis in the cerebellar external granule layer (EGL). The EGL is a transient proliferative region responsible for the production of over 90% of the neurons in the cerebellum. During normal development, endogenous GC stimulation is thought to selectively signal the elimination of the EGL once production of new neurons is complete. As a result, GC therapy may precociously eliminate the EGL before it can produce enough neurons for normal cerebellar function. It is hoped that this review may provide information for future clinical research in addition to translational guidance for the safer use of GC therapy.
Collapse
Affiliation(s)
- Kevin K Noguchi
- Department of Psychiatry, School of Medicine, Washington University in St. Louis, 660 South Euclid, Box #8134, St. Louis, MO 63110, USA; Tel.: +1-314-362-7007
| |
Collapse
|
23
|
Huang LT. Early-life stress impacts the developing hippocampus and primes seizure occurrence: cellular, molecular, and epigenetic mechanisms. Front Mol Neurosci 2014; 7:8. [PMID: 24574961 PMCID: PMC3918912 DOI: 10.3389/fnmol.2014.00008] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Accepted: 01/21/2014] [Indexed: 01/03/2023] Open
Abstract
Early-life stress includes prenatal, postnatal, and adolescence stress. Early-life stress can affect the development of the hypothalamic-pituitary-adrenal (HPA) axis, and cause cellular and molecular changes in the developing hippocampus that can result in neurobehavioral changes later in life. Epidemiological data implicate stress as a cause of seizures in both children and adults. Emerging evidence indicates that both prenatal and postnatal stress can prime the developing brain for seizures and an increase in epileptogenesis. This article reviews the cellular and molecular changes encountered during prenatal and postnatal stress, and assesses the possible link between these changes and increases in seizure occurrence and epileptogenesis in the developing hippocampus. In addititon, the priming effect of prenatal and postnatal stress for seizures and epileptogenesis is discussed. Finally, the roles of epigenetic modifications in hippocampus and HPA axis programming, early-life stress, and epilepsy are discussed.
Collapse
Affiliation(s)
- Li-Tung Huang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine Kaohsiung, Taiwan ; Department of Traditional Chinese Medicine, Chang Gung University Linkou, Taiwan
| |
Collapse
|
24
|
Wang L, Zhang W, Wu R, Kong L, Feng W, Cao Y, Tai F, Zhang X. Neuroendocrine responses to social isolation and paternal deprivation at different postnatal ages in Mandarin voles. Dev Psychobiol 2014; 56:1214-28. [PMID: 24464494 DOI: 10.1002/dev.21202] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 01/02/2014] [Indexed: 02/05/2023]
Abstract
Neonatal isolation and paternal deprivation have long lasting effects on the behavior and neuroendocrine system at adulthood. Whether these effects at adulthood are induced by neonatal changes in relevant neuroendocrine parameters lead by these early-life social experiences is not well understood. Whether monogamous rodents exhibit a stress hypo-responsive period (SHRP) also remains unclear. Using the monogamous mandarin vole, we found that 30 min of isolation did not affect levels of corticosterone (CORT) and adrenocorticotropin (ACTH) at postnatal days 8, 10, and 12 displaying a SHRP, but increased these at postnatal days 4, 14, 16, and 18. Isolation increased vasopressin (AVP)-ir neurons in the paraventricular nucleus (PVN) and supraoptic nucleus (SON) from postnatal days 4 to 12, and up-regulated oxytocin (OT)-ir neurons in the PVN at postnatal days 4 and 8 and SON at postnatal day 4. Paternally deprived pups showed increase in ACTH and CORT after 30 min of social isolation from postnatal days 8 to 14, increase in AVP-ir neurons in the PVN from postnatal days 10 to 14, reduction in OT-ir neurons in the PVN from postnatal days 10 to 14 and in the SON at postnatal days 12 and 14. These results indicate that monogamous mandarin voles display a short SHRP which can be disrupted by paternal deprivation. Central AVP and OT levels may also be altered by paternal deprivation and social isolation. We propose that changes in these neuroendocrine parameters induced by early-life social experiences such as those tested here persist and result.
Collapse
Affiliation(s)
- Lu Wang
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, 710062, Shaanxi, China; Key laboratory of Modern Teaching Technology, Ministry of Education, Xi'an, 710062, Shaanxi, China
| | | | | | | | | | | | | | | |
Collapse
|
25
|
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: 1.3] [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
|
26
|
de Kloet ER, Claessens SEF, Kentrop J. Context modulates outcome of perinatal glucocorticoid action in the brain. Front Endocrinol (Lausanne) 2014; 5:100. [PMID: 25071717 PMCID: PMC4088189 DOI: 10.3389/fendo.2014.00100] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 06/11/2014] [Indexed: 11/13/2022] Open
Abstract
Prematurely born infants may be at risk, because of inadequate maturation of tissues. If there are signs of preterm birth, it has become common practice therefore to treat either antenatally the mother or postnatally the infant with glucocorticoids to accelerate tissue development, particularly of the lung. However, this life-saving early glucocorticoid treatment was found to increase the risk of adverse outcome in later life. In one animal study, the authors reported a 25% shorter lifespan of rats treated as newborns with the synthetic glucocorticoid dexamethasone, but so far this finding has not been replicated. After a brief clinical introduction, we discuss studies in rodents designed to examine how perinatal glucocorticoid action affects the developing brain. It appears that the perinatal action of the glucocorticoid depends on the context and the timing as well as the type of administered steroid. The type of steroid is important because the endogenous glucocorticoids cortisol and corticosterone bind to two distinct receptor populations, i.e., mineralocorticoid and glucocorticoid receptors (GR), while synthetic glucocorticoids predominantly bind to the GR. In addition, if given antenatally hydrocortisone is inactivated in the placenta by 11β-HSD type 2, and dexamethasone is not. With respect to timing, the outcome of glucocorticoid effects is different in early vs. late phases of brain development. The context refers to the environmental input that can affect the susceptibility to glucocorticoid action in the newborn rodent brain; early handling of pups and maternal care obliterate effects of post-natal dexamethasone treatment. Context also refers to coping with environmental conditions in later life, for which the individual may have been programed epigenetically by early-life experience. This knowledge of determinants affecting the outcome of perinatal glucocorticoid exposure may have clinical implications for the treatment of prematurely born infants.
Collapse
Affiliation(s)
- E. Ronald de Kloet
- Department of Medical Pharmacology, Leiden University Medical Center, Leiden University, Leiden, Netherlands
- Department of Endocrinology and Metabolism, Leiden University Medical Center, Leiden, Netherlands
- *Correspondence: E. Ronald de Kloet, Department of Endocrinology and Metabolism, Division of Medical Pharmacology, LACDR, Leiden University Medical Center, Leiden University, PO Box 9503, Leiden 2300 RA, Netherlands e-mail: ;
| | - Sanne E. F. Claessens
- Department of Medical Pharmacology, Leiden University Medical Center, Leiden University, Leiden, Netherlands
| | - Jiska Kentrop
- Department of Medical Pharmacology, Leiden University Medical Center, Leiden University, Leiden, Netherlands
| |
Collapse
|
27
|
Cope JL, Regev L, Chen Y, Korosi A, Rice CJ, Ji S, Rogge GA, Wood MA, Baram TZ. Differential contribution of CBP:CREB binding to corticotropin-releasing hormone expression in the infant and adult hypothalamus. Stress 2014; 17:39-50. [PMID: 23768074 PMCID: PMC3869921 DOI: 10.3109/10253890.2013.806907] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Corticotropin-releasing hormone (CRH) contributes crucially to the regulation of central and peripheral responses to stress. Because of the importance of a finely tuned stress system, CRH expression is tightly regulated in an organ- and brain region-specific manner. Thus, in the hypothalamus, CRH is constitutively expressed and this expression is further enhanced by stress; however, the underlying regulatory mechanisms are not fully understood. The regulatory region of the crh gene contains several elements, including the cyclic-AMP response element (CRE), and the role of the CRE interaction with the cyclic-AMP response element binding protein (CREB) in CRH expression has been a focus of intensive research. Notably, whereas thousands of genes contain a CRE, the functional regulation of gene expression by the CRE:CREB system is limited to ∼100 genes, and likely requires additional proteins. Here, we investigated the role of a member of the CREB complex, CREB binding protein (CBP), in basal and stress-induced CRH expression during development and in the adult. Using mice with a deficient CREB-binding site on CBP, we found that CBP:CREB interaction is necessary for normal basal CRH expression at the mRNA and protein level in the nine-day-old mouse, prior to onset of functional regulation of hypothalamic CRH expression by glucocorticoids. This interaction, which functions directly on crh or indirectly via regulation of other genes, was no longer required for maintenance of basal CRH expression levels in the adult. However, CBP:CREB binding contributed to stress-induced CRH expression in the adult, enabling rapid CRH synthesis in hypothalamus. CBP:CREB binding deficiency did not disrupt basal corticosterone plasma levels or acute stress-evoked corticosterone release. Because dysregulation of CRH expression occurs in stress-related disorders including depression, a full understanding of the complex regulation of this gene is important in both health and disease.
Collapse
Affiliation(s)
- Jessica L. Cope
- Department of Anatomy and Neurobiology, University of California-Irvine, Irvine, CA 92697, USA
| | - Limor Regev
- Department of Anatomy and Neurobiology, University of California-Irvine, Irvine, CA 92697, USA
| | - Yuncai Chen
- Department of Pediatrics, University of California-Irvine, Irvine, CA 92697, USA
| | - Aniko Korosi
- Department of Anatomy and Neurobiology, University of California-Irvine, Irvine, CA 92697, USA
| | - Courtney J. Rice
- Department of Anatomy and Neurobiology, University of California-Irvine, Irvine, CA 92697, USA
| | - Sung Ji
- Department of Anatomy and Neurobiology, University of California-Irvine, Irvine, CA 92697, USA
| | - George A. Rogge
- Department of Neurobiology and Behavior, University of California-Irvine, Irvine, CA 92697, USA
| | - Marcelo A. Wood
- Department of Neurobiology and Behavior, University of California-Irvine, Irvine, CA 92697, USA
| | - Tallie Z. Baram
- Department of Anatomy and Neurobiology, University of California-Irvine, Irvine, CA 92697, USA
- Department of Pediatrics, University of California-Irvine, Irvine, CA 92697, USA
| |
Collapse
|
28
|
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: 12.5] [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]
|
29
|
Krugers HJ, Joëls M. Long-lasting Consequences of Early Life Stress on Brain Structure, Emotion and Cognition. Curr Top Behav Neurosci 2014; 18:81-92. [PMID: 24862989 DOI: 10.1007/7854_2014_289] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
During the perinatal period, the brain undergoes substantial structural changes, synaptic rearrangements, and development of neuronal circuits which ultimately determine brain function and behavior. Environmental factors-such as exposure to adverse experiences-have major impact on brain function and structure during this sensitive period. These alterations can be long-lasting, and have been implicated in psychopathology such as cognitive decline and emotional dysfunction. Here we briefly review how early postnatal adversity determines structure and function of the hippocampus, amygdala, and prefrontal cortex (PFC) areas, which are crucial for proper cognitive and emotional function.
Collapse
Affiliation(s)
- Harm J Krugers
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands,
| | | |
Collapse
|
30
|
Hall FS, Perona MTG. Have studies of the developmental regulation of behavioral phenotypes revealed the mechanisms of gene-environment interactions? Physiol Behav 2012; 107:623-40. [PMID: 22643448 PMCID: PMC3447116 DOI: 10.1016/j.physbeh.2012.05.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Revised: 05/15/2012] [Accepted: 05/15/2012] [Indexed: 12/30/2022]
Abstract
This review addresses the recent convergence of our long-standing knowledge of the regulation of behavioral phenotypes by developmental experience with recent advances in our understanding of mechanisms regulating gene expression. This review supports a particular perspective on the developmental regulation of behavioral phenotypes: That the role of common developmental experiences (e.g. maternal interactions, peer interactions, exposure to a complex environment, etc.) is to fit individuals to the circumstances of their lives within bounds determined by long-standing (evolutionary) mechanisms that have shaped responses to critical and fundamental types of experience via those aspects of gene structure that regulate gene expression. The phenotype of a given species is not absolute for a given genotype but rather variable within bounds that is determined by mechanisms regulated by experience (e.g. epigenetic mechanisms). This phenotypic variation is not necessarily random, or evenly distributed along a continuum of description or measurement, but often highly disjointed, producing distinct, even opposing, phenotypes. The potentiality for these varying phenotypes is itself the product of evolution, the potential for alternative phenotypes itself conveying evolutionary advantage. Examples of such phenotypic variation, resulting from environmental or experiential influences, have a long history of study in neurobiology, and a number of these will be discussed in this review: neurodevelopmental experiences that produce phenotypic variation in visual perception, cognitive function, and emotional behavior. Although other examples will be discussed, particular emphasis will be made on the role of social behavior on neurodevelopment and phenotypic determination. It will be argued that an important purpose of some aspects of social behavior is regulation of neurobehavioral phenotypes by experience via genetic regulatory mechanisms.
Collapse
Affiliation(s)
- F Scott Hall
- Molecular Neurobiology Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, 333 Cassel Drive, Baltimore, MD 21224, United States.
| | | |
Collapse
|
31
|
Ganea K, Menke A, Schmidt MV, Lucae S, Rammes G, Liebl C, Harbich D, Sterlemann V, Storch C, Uhr M, Holsboer F, Binder EB, Sillaber I, Müller MB. Convergent animal and human evidence suggests the activin/inhibin pathway to be involved in antidepressant response. Transl Psychiatry 2012; 2:e177. [PMID: 23092981 PMCID: PMC3565812 DOI: 10.1038/tp.2012.104] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Despite the overt need for improved treatment modalities in depression, efforts to develop conceptually novel antidepressants have been relatively unsuccessful so far. Here we present a translational approach combining results from hypothesis-free animal experiments with data from a genetic association study in depression. Comparing genes regulated by chronic paroxetine treatment in the mouse hippocampus with genes showing nominally significant association with antidepressant treatment response in two pharmacogenetic studies, the activin pathway was the only one to show this dual pattern of association and therefore selected as a candidate. We examined the regulation of activin A and activin receptor type IA mRNA following antidepressant treatment. We investigated the effects of stereotaxic infusion of activin into the hippocampus and the amygdala in a behavioural model of depression. To analyse whether variants in genes in the activin signalling pathway predict antidepressant treatment response, we performed a human genetic association study. Significant changes in the expression of genes in the activin signalling pathway were observed following 1 and 4 weeks of treatment. Injection of activin A into the hippocampus exerts acute antidepressant-like effects. Polymorphisms in the betaglycan gene, a co-receptor mediating functional antagonism of activin signalling, significantly predict treatment outcome in our system-wide pharmacogenetics study in depression. We provide convergent evidence from mouse and human data that genes in the activin signalling pathway are promising novel candidates involved in the neurobiogical mechanisms underlying antidepressant mechanisms of action. Further, our data suggest this pathway to be a target for more rapid-acting antidepressants in the future.
Collapse
Affiliation(s)
- K Ganea
- Max Planck Institute of Psychiatry, Munich, Germany
| | - A Menke
- Max Planck Institute of Psychiatry, Munich, Germany
| | - M V Schmidt
- Max Planck Institute of Psychiatry, Munich, Germany
| | - S Lucae
- Max Planck Institute of Psychiatry, Munich, Germany
| | - G Rammes
- Max Planck Institute of Psychiatry, Munich, Germany,Department of Anesthesiology, Technische Universtität, Munich, Germany
| | - C Liebl
- Max Planck Institute of Psychiatry, Munich, Germany
| | - D Harbich
- Max Planck Institute of Psychiatry, Munich, Germany
| | - V Sterlemann
- Max Planck Institute of Psychiatry, Munich, Germany
| | - C Storch
- Max Planck Institute of Psychiatry, Munich, Germany
| | - M Uhr
- Max Planck Institute of Psychiatry, Munich, Germany
| | - F Holsboer
- Max Planck Institute of Psychiatry, Munich, Germany
| | - E B Binder
- Max Planck Institute of Psychiatry, Munich, Germany
| | - I Sillaber
- Phenoquest AG, Martinsried/Munich, Munich, Germany
| | - M B Müller
- Max Planck Institute of Psychiatry, Munich, Germany,Molecular Stress Physiology, Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804 Munich, Germany. E-mail:
| |
Collapse
|
32
|
Chronic activation of corticotropin-releasing factor type 2 receptors reveals a key role for 5-HT1A receptor responsiveness in mediating behavioral and serotonergic responses to stressful challenge. Biol Psychiatry 2012; 72:437-47. [PMID: 22704666 PMCID: PMC3430862 DOI: 10.1016/j.biopsych.2012.05.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Revised: 04/20/2012] [Accepted: 05/04/2012] [Indexed: 11/30/2022]
Abstract
BACKGROUND The corticotropin-releasing factor type 2 receptor (CRFR2) is suggested to play an important role in aiding recovery from acute stress, but any chronic effects of CRFR2 activation are unknown. CRFR2 in the midbrain raphé nuclei modulate serotonergic activity of this key source of serotonin (5-HT) forebrain innervation. METHODS Transgenic mice overexpressing the highly specific CRFR2 ligand urocortin 3 (UCN3OE) were analyzed for stress-related behaviors and hypothalamic-pituitary-adrenal axis responses. Responses to 5-HT receptor agonist challenge were assessed by local cerebral glucose utilization, while 5-HT and 5-hydroxyindoleacetic acid content were quantified in limbic brain regions. RESULTS Mice overexpressing urocortin 3 exhibited increased stress-related behaviors under basal conditions and impaired retention of spatial memory compared with control mice. Following acute stress, unlike control mice, they exhibited no further increase in these stress-related behaviors and showed an attenuated adrenocorticotropic hormone response. 5-HT and 5-hydroxyindoleacetic acid content of limbic nuclei were differentially regulated by stress in UCN3OE mice as compared with control mice. Responses to 5-HT type 1A receptor challenge were significantly and specifically reduced in UCN3OE mice. The distribution pattern of local cerebral glucose utilization and 5-HT type 1A receptor messenger RNA expression levels suggested this effect was mediated in the raphé nuclei. CONCLUSIONS Chronic activation of CRFR2 promotes an anxiety-like state, yet with attenuated behavioral and hypothalamic-pituitary-adrenal axis responses to stress. This is reminiscent of stress-related atypical psychiatric syndromes such as posttraumatic stress disorder, chronic fatigue, and chronic pain states. This new understanding indicates CRFR2 antagonism as a potential novel therapeutic target for such disorders.
Collapse
|
33
|
Profiling trait anxiety: transcriptome analysis reveals cathepsin B (Ctsb) as a novel candidate gene for emotionality in mice. PLoS One 2011; 6:e23604. [PMID: 21897848 PMCID: PMC3163650 DOI: 10.1371/journal.pone.0023604] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2011] [Accepted: 07/20/2011] [Indexed: 11/19/2022] Open
Abstract
Behavioral endophenotypes are determined by a multitude of counteracting but precisely balanced molecular and physiological mechanisms. In this study, we aim to identify potential novel molecular targets that contribute to the multigenic trait “anxiety”. We used microarrays to investigate the gene expression profiles of different brain regions within the limbic system of mice which were selectively bred for either high (HAB) or low (LAB) anxiety-related behavior, and also show signs of comorbid depression-like behavior. We identified and confirmed sex-independent differences in the basal expression of 13 candidate genes, using tissue from the entire brain, including coronin 7 (Coro7), cathepsin B (Ctsb), muscleblind-like 1 (Mbnl1), metallothionein 1 (Mt1), solute carrier family 25 member 17 (Slc25a17), tribbles homolog 2 (Trib2), zinc finger protein 672 (Zfp672), syntaxin 3 (Stx3), ATP-binding cassette, sub-family A member 2 (Abca2), ectonucleotide pyrophosphatase/phosphodiesterase 5 (Enpp5), high mobility group nucleosomal binding domain 3 (Hmgn3) and pyruvate dehydrogenase beta (Pdhb). Additionally, we confirmed brain region-specific differences in the expression of synaptotagmin 4 (Syt4). Our identification of about 90 polymorphisms in Ctsb suggested that this gene might play a critical role in shaping our mouse model's behavioral endophenotypes. Indeed, the assessment of anxiety-related and depression-like behaviors of Ctsb knock-out mice revealed an increase in depression-like behavior in females. Altogether, our results suggest that Ctsb has significant effects on emotionality, irrespective of the tested mouse strain, making it a promising target for future pharmacotherapy.
Collapse
|
34
|
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: 2.4] [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
|
35
|
Effects of early life stress on neuroendocrine and neurobehavior: mechanisms and implications. Pediatr Neonatol 2011; 52:122-9. [PMID: 21703552 DOI: 10.1016/j.pedneo.2011.03.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2010] [Revised: 11/30/2010] [Accepted: 12/23/2010] [Indexed: 12/12/2022] Open
Abstract
Evidence continues to mount that adverse experiences early in life have an impact on brain functions. Early life stress can program the development of the hypothalamic-pituitary-adrenal axis and cause alterations of neurochemistry and signaling pathways involved in regulating neuroplasticity, with resultant neurobehavioral changes. Early life experiences and genetic factors appear to interact in determining the individual vulnerability to mental health disorders. We reviewed the effects of early life stress on neuroendocrine regulation and the relevance to neurobehavioral development.
Collapse
|
36
|
Noguchi KK, Lau K, Smith DJ, Swiney BS, Farber NB. Glucocorticoid receptor stimulation and the regulation of neonatal cerebellar neural progenitor cell apoptosis. Neurobiol Dis 2011; 43:356-63. [PMID: 21530661 DOI: 10.1016/j.nbd.2011.04.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2011] [Revised: 03/14/2011] [Accepted: 04/07/2011] [Indexed: 01/21/2023] Open
Abstract
Glucocorticoids are used to treat respiratory dysfunction associated with premature birth but have been shown to cause neurodevelopmental deficits when used therapeutically. Recently, we established that acute glucocorticoid exposure at clinically relevant doses produces neural progenitor cell apoptosis in the external granule layer of the developing mouse cerebellum and permanent decreases in the number of cerebellar neurons. As the cerebellum naturally matures and neurogenesis is no longer needed, the external granule layer decreases proliferation and permanently disappears during the second week of life. At this same time, corticosterone (the endogenous rodent glucocorticoid) release increases and a glucocorticoid-metabolizing enzyme that protects the external granule layer against glucocorticoid receptor stimulation (11β-Hydroxysteroid-Dehydrogenase-Type 2; HSD2) naturally disappears. Here we show that HSD2 inhibition and raising corticosterone to adult physiological levels both can independently increase neural progenitor cell apoptosis in the neonatal mouse. Conversely, glucocorticoid receptor antagonism decreases natural physiological apoptosis in this same progenitor cell population suggesting that endogenous glucocorticoid stimulation may regulate apoptosis in the external granule layer. We also found that glucocorticoids which HSD2 can effectively metabolize generate less external granule layer apoptosis than glucocorticoids this enzyme is ineffective at breaking down. This finding may explain why glucocorticoids that this enzyme can metabolize are clinically effective at treating respiratory dysfunction yet seem to produce no neurodevelopmental deficits. Finally, we demonstrate that both acute and chronic glucocorticoid exposures produce external granule layer apoptosis but without appropriate control groups this effect becomes masked. These results are discussed in terms of their implications for glucocorticoid therapy and neurodevelopment during the perinatal period.
Collapse
Affiliation(s)
- Kevin K Noguchi
- Washington University in Saint Louis, School of Medicine, Department of Psychiatry, 660 South Euclid, Box #8134, Saint Louis, MO 63110, USA.
| | | | | | | | | |
Collapse
|
37
|
McClelland S, Korosi A, Cope J, Ivy A, Baram TZ. Emerging roles of epigenetic mechanisms in the enduring effects of early-life stress and experience on learning and memory. Neurobiol Learn Mem 2011; 96:79-88. [PMID: 21338703 DOI: 10.1016/j.nlm.2011.02.008] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2010] [Revised: 02/04/2011] [Accepted: 02/10/2011] [Indexed: 12/25/2022]
Abstract
Epigenetic mechanisms are involved in programming gene expression throughout development. In addition, they are key contributors to the processes by which early-life experience fine-tunes the expression levels of key neuronal genes, governing learning and memory throughout life. Here we describe the long-lasting, bi-directional effects of early-life experience on learning and memory. We discuss how enriched postnatal experience enduringly augments spatial learning, and how chronic early-life stress results in persistent and progressive deficits in the structure and function of hippocampal neurons. The existing and emerging roles of epigenetic mechanisms in these fundamental neuroplasticity phenomena are illustrated.
Collapse
|
38
|
Macrì S, Zoratto F, Laviola G. Early-stress regulates resilience, vulnerability and experimental validity in laboratory rodents through mother-offspring hormonal transfer. Neurosci Biobehav Rev 2011; 35:1534-43. [PMID: 21216260 DOI: 10.1016/j.neubiorev.2010.12.014] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2010] [Revised: 12/15/2010] [Accepted: 12/27/2010] [Indexed: 01/31/2023]
Abstract
The role of early-life stressors in the calibration of individual responses to future challenges has long been investigated in laboratory rodents. Specifically, countless studies show that exposure to early-life stressors - in the form of various periods of maternal separation, administration of exogenous corticosterone and variable feeding conditions - modulate the regulation of defensive responses (e.g. behavioral fearfulness/anxiety and endocrine stress reactivity) in adulthood. Yet, the link between early-life stress and adult defensive responses is not linear. Specifically, while neonatal moderate stress is generally associated with adult subjects characterized by reduced stress reactivity, neonatal elevated stress is often reported to relate to opposite responses. Not only are these findings relevant to the understanding of individual plasticity to contextual features, but also they can have direct implications in the development of rodent models of human disorders. Specifically, these studies demonstrate that the experimental individual responds to early environmental cues with the consequence of adjusting its adaptation to the future environment. If neglected, this aspect may have detrimental consequences in laboratory animal experimentation. For example, neonatal conditions increasing adult responses to moderate stress may result in experimental subjects showing abnormal hypothalamic-pituitary-adrenocortical (HPA) activation to routine husbandry conditions, test environment and general laboratory procedures. The aim of the present review is threefold: (i) propose that neonatal circulating levels of corticosteroids may constitute a potential mediator connecting early and adult defensive systems; (ii) propose that the link between early and adult stress follows a U-shaped curve, with low levels down-regulating individual reactivity to external stressors and high levels exerting opposite effects; (iii) discuss the methodological implications of these considerations in the development of rodent models of human disorders.
Collapse
Affiliation(s)
- Simone Macrì
- Section of Behavioural Neuroscience, Department of Cell Biology & Neuroscience, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Roma, Italy.
| | | | | |
Collapse
|
39
|
Pinheiro ML, Ferraz-de-Paula V, Ribeiro A, Sakai M, Bernardi MM, Palermo-Neto J. Long-term maternal separation differentially alters serum corticosterone levels and blood neutrophil activity in A/J and C57BL/6 mouse offspring. Neuroimmunomodulation 2011; 18:184-90. [PMID: 21311204 DOI: 10.1159/000323516] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2010] [Accepted: 12/02/2010] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVES In this work, we searched for maternal separation effects on serum corticosterone levels and blood neutrophil activity in adult male A/J and C57BL/6 mouse offspring. METHODS 40 male A/J mice and 40 male C57BL/6 mice were divided within each strain into two groups. Mice in the maternal separation group were separated from their mothers (1 h/day) on postnatal days 0-13. Mice in the control group were left undisturbed. On postnatal day 45, blood was drawn from all mice and used to assess neutrophil activity by flow cytometry and serum corticosterone levels by radioimmunoassay. RESULTS The results showed that each mouse strain responded differently to maternal separation, but in both cases, serum corticosterone levels were affected. In both strains, adult mice that experienced maternal separation showed lower serum corticosterone levels than control mice. In relation to control mice kept together with their mothers, the levels of serum corticosterone were 72.7 and 36.36% lower in A/J and C57BL/6 mice submitted to maternal separation, respectively. The current findings showed that maternal separation increased neutrophil activity in mice after reaching adulthood. The observed effects, although in the same direction, differed between A/J and C57BL/6 mice. Maternal separation increased both the percentage and intensity of phagocytosis in C57BL/6 mice, but had no effects on A/J mice. Furthermore, maternal separation increased basal and propidium iodide-labeled Staphylococcus aureus-induced oxidative burst in A/J mice but did not affect oxidative burst in C57BL/6 mice. Finally, phorbol myristate acetate-induced oxidative burst increased in both strains. CONCLUSION These results indicate that early maternal separation increases innate immunity, most likely by modifying hypothalamus-pituitary-adrenal axis activity. This suggests that maternal separation is a good model for stress which produces long-term neuroimmune changes whatever the animal species and strain used.
Collapse
Affiliation(s)
- Milena Lobão Pinheiro
- Laboratory of Neuroimmunomodulation, Department of Pathology, School of Veterinary Medicine, University of São Paulo, São Paulo, Brazil
| | | | | | | | | | | |
Collapse
|
40
|
Gross CM, Flubacher A, Tinnes S, Heyer A, Scheller M, Herpfer I, Berger M, Frotscher M, Lieb K, Haas CA. Early life stress stimulates hippocampal reelin gene expression in a sex-specific manner: evidence for corticosterone-mediated action. Hippocampus 2010; 22:409-20. [PMID: 21136520 DOI: 10.1002/hipo.20907] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/14/2010] [Indexed: 12/12/2022]
Abstract
Early life stress predisposes to the development of psychiatric disorders. In this context the hippocampal formation is of particular interest, because it is affected by stress on the structural and cognitive level. Since little is known how early life stress is translated on the molecular level, we mimicked early life stress in mouse models and analyzed the expression of the glycoprotein Reelin, a master molecule for development and differentiation of the hippocampus. From postnatal day 1 (P1) to P14, mouse pups were subjected to one of the following treatments: nonhandling (NH), handling (H), maternal separation (MS), and early deprivation (ED) followed by immediate (P15) or delayed (P70) real time RT-PCR analysis of reelin mRNA expression. We show that at P15, reelin mRNA levels were significantly increased in male H and ED groups when compared with the NH group. In contrast, no stress-induced alterations of reelin mRNA expression were found in female animals. This sex difference in stress-mediated stimulation of reelin expression was maintained into adulthood, since at P70 intergroup differences were still found in male, but not in female mice. On the cellular level, however, we did not find any significant differences in cell densities of Reelin-immunolabeled neurons between treatment groups or sexes, but an overall reduction of Reelin-expressing neurons in the adult hippocampus when compared to P15. To address the question whether corticosterone mediates the stress-induced up-regulation of reelin gene expression, we used age-matched hippocampal slice cultures derived from male and female mouse pups. Quantitative determination of mRNA levels revealed that corticosterone treatment significantly up-regulated reelin mRNA expression in male, but not in female hippocampi. Taken together, these results show a sex-specific regulation of reelin gene expression by early life experience, most likely mediated by corticosterone.
Collapse
Affiliation(s)
- Claus M Gross
- Experimental Epilepsy Research, Neurocenter, University of Freiburg, Freiburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Skripuletz T, Kruschinski C, Pabst R, Hörsten S, Stephan M. Postnatal experiences influence the behavior in adult male and female Fischer and Lewis rats. Int J Dev Neurosci 2010; 28:561-71. [DOI: 10.1016/j.ijdevneu.2010.07.235] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2010] [Revised: 07/16/2010] [Accepted: 07/27/2010] [Indexed: 11/25/2022] Open
Affiliation(s)
- Thomas Skripuletz
- Department of NeurologyHannover Medical SchoolHannoverGermany
- Institute of Functional and Applied AnatomyHannover Medical SchoolHannoverGermany
| | - Carsten Kruschinski
- Institute of Functional and Applied AnatomyHannover Medical SchoolHannoverGermany
- Institute of General PracticeHannover Medical SchoolHannoverGermany
| | - Reinhard Pabst
- Institute of Functional and Applied AnatomyHannover Medical SchoolHannoverGermany
| | - Stephan Hörsten
- Institute of Functional and Applied AnatomyHannover Medical SchoolHannoverGermany
- Experimental Therapy, Franz‐Penzoldt‐CenterFriedrich‐Alexander‐University Erlangen‐NürnbergErlangenGermany
| | - Michael Stephan
- Institute of Functional and Applied AnatomyHannover Medical SchoolHannoverGermany
- Clinic for Psychosomatics and Psychotherapy, Hannover Medical SchoolHannoverGermany
| |
Collapse
|
42
|
Mabandla MV, Russell VA. Voluntary exercise reduces the neurotoxic effects of 6-hydroxydopamine in maternally separated rats. Behav Brain Res 2010; 211:16-22. [PMID: 20206210 PMCID: PMC2862124 DOI: 10.1016/j.bbr.2010.02.045] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2009] [Revised: 02/23/2010] [Accepted: 02/23/2010] [Indexed: 11/28/2022]
Abstract
Maternal separation has been associated with development of anxiety-like behaviour and learning impairments in adult rats. This has been linked to changes in brain morphology observed after exposure to high levels of circulating glucocorticoids during the stress-hyporesponsive period (P4-P14). In the present study, adult rats that had been subjected to maternal separation (180 min/day for 14 days) during the stress-hyporesponsive period, received unilateral infusions of a small dose of 6-hydroxydopamine (6-OHDA, 5 microg/4 microl saline) into the medial forebrain bundle. The results showed that voluntary exercise had a neuroprotective effect in both non-stressed and maternally separated rats in that there was a decrease in forelimb akinesia (step test) and limb use asymmetry (cylinder test). Maternal separation increased forelimb akinesia and forelimb use asymmetry and reduced the beneficial effect of exercise on forelimb akinesia. It also reduced exploratory behaviour, consistent with anxiety-like behaviour normally associated with maternal separation. Exercise appeared to reduce dopamine neuron destruction in the lesioned substantia nigra when expressed as a percentage of the non-lesioned hemisphere. However, this appeared to be due to a compensatory decrease in completely stained tyrosine hydroxylase-positive neurons in the contralateral, non-lesioned substantia nigra. In agreement with reports that maternal separation increases the 6-OHDA-induced loss of dopamine terminals in the striatum, there was a small increase in dopamine neuron destruction when expressed as a percentage of the non-lesioned hemisphere but there was no difference in dopamine cell number, suggesting that exposure to maternal separation did not exacerbate dopamine cell loss.
Collapse
Affiliation(s)
- Musa Vuyisile Mabandla
- Department of Human Physiology, School of Medical Sciences, Faculty of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa.
| | | |
Collapse
|
43
|
Molecular mechanisms of early life stress—Lessons from mouse models. Neurosci Biobehav Rev 2010; 34:845-52. [DOI: 10.1016/j.neubiorev.2009.05.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2008] [Revised: 03/23/2009] [Accepted: 05/05/2009] [Indexed: 11/22/2022]
|
44
|
Meagher MW, Sieve AN, Johnson RR, Satterlee D, Belyavskyi M, Mi W, Prentice TW, Welsh TH, Welsh CJR. Neonatal maternal separation alters immune, endocrine, and behavioral responses to acute Theiler's virus infection in adult mice. Behav Genet 2010; 40:233-49. [PMID: 20135342 DOI: 10.1007/s10519-010-9333-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2009] [Accepted: 01/09/2010] [Indexed: 12/01/2022]
Abstract
Previous studies have established a link between adverse early life events and subsequent disease vulnerability. The present study assessed the long-term effects of neonatal maternal separation on the response to Theiler's murine encephalomyelitis virus infection, a model of multiple sclerosis. Balb/cJ mouse pups were separated from their dam for 180-min/day (180-min MS), 15-min/day (15-min MS), or left undisturbed from postnatal days 2-14. During adolescence, mice were infected with Theiler's virus and sacrificed at days 14, 21, or 35 post-infection. Prolonged 180-min MS increased viral load and delayed viral clearance in the spinal cords of males and females, whereas brief 15-min MS increased the rate of viral clearance in females. The 15-min and 180-min MS mice exhibited blunted corticosterone responses during infection, suggesting that reduced HPA sensitivity may have altered the immune response to infection. These findings demonstrate that early life events alter vulnerability to CNS infection later in life. Therefore, this model could be used to study gene-environment interactions that contribute to individual differences in susceptibility to infectious and autoimmune diseases of the CNS.
Collapse
Affiliation(s)
- M W Meagher
- Department of Psychology, College of Liberal Arts, Texas A&M University, College Station, TX, 77843-4235, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Susceptibility to stress in transgenic mice overexpressing TrkC, a model of panic disorder. J Psychiatr Res 2010; 44:157-67. [PMID: 19698958 DOI: 10.1016/j.jpsychires.2009.07.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2009] [Revised: 07/24/2009] [Accepted: 07/27/2009] [Indexed: 12/21/2022]
Abstract
Stressful life events increase the susceptibility for subsequent onset of psychiatric disorders in humans. Previous research has implicated neurotrophins in the onset of some stress-related diseases, such as major depression disorder, post-traumatic stress disorder or panic disorder. We have tested the hypothesis that the neurotrophin-3 (NT-3)/TrkC system is a genetic interface mediating the deleterious effects of stress on the initiation of panic disorder and other pathologies. To this aim, we have analyzed the functionality of HPA axis and the behavioral consequences of different types of stressful conditions in a mouse model of panic disorder, which overexpresses TrkC, the high affinity-receptor for NT-3 (TgNTRK3). Our results reveal that TgNTRK3 mice exhibit an altered circadian corticosterone rhythm that is reversed by clonidine treatment, but normal expression of genes involved in the control of the hypothalamus-pituitary-adrenal (HPA) axis (CRH, GR) and normal corticosterone response to acute and chronic stressors. In contrast, they exhibit an altered pattern of activation of stress-related brain areas and showed enhanced anxiety-related behavior and more passive strategies than wild types under some chronic stress conditions. We conclude that TgNTRK3 mice present differences in their response to stress characterized by subtle changes in the HPA axis, marked changes in acute stress-induced brain activation and altered coping strategies, suggesting a key role of TrkC receptor in the stress neural circuitry and in the behavioral consequences of chronic stress.
Collapse
|
46
|
Enthoven L, Schmidt M, Cheung Y, Mark M, Kloet E, Oitzl M. Ontogeny of the HPA axis of the CD1 mouse following 24 h maternal deprivation at pnd 3. Int J Dev Neurosci 2009; 28:217-24. [DOI: 10.1016/j.ijdevneu.2009.10.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2009] [Revised: 10/20/2009] [Accepted: 10/27/2009] [Indexed: 01/08/2023] Open
Affiliation(s)
- L. Enthoven
- Division of Medical PharmacologyLeiden/Amsterdam Center for Drug ResearchLeiden University Medical CenterLeiden UniversityThe Netherlands
| | - M.V. Schmidt
- Division of Medical PharmacologyLeiden/Amsterdam Center for Drug ResearchLeiden University Medical CenterLeiden UniversityThe Netherlands
| | - Y.H. Cheung
- Division of Medical PharmacologyLeiden/Amsterdam Center for Drug ResearchLeiden University Medical CenterLeiden UniversityThe Netherlands
| | - M.H. Mark
- Division of Medical PharmacologyLeiden/Amsterdam Center for Drug ResearchLeiden University Medical CenterLeiden UniversityThe Netherlands
| | - E.R. Kloet
- Division of Medical PharmacologyLeiden/Amsterdam Center for Drug ResearchLeiden University Medical CenterLeiden UniversityThe Netherlands
| | - M.S. Oitzl
- Division of Medical PharmacologyLeiden/Amsterdam Center for Drug ResearchLeiden University Medical CenterLeiden UniversityThe Netherlands
| |
Collapse
|
47
|
Neonatal stress modulates sickness behavior. Brain Behav Immun 2009; 23:977-85. [PMID: 19464359 PMCID: PMC4217217 DOI: 10.1016/j.bbi.2009.05.056] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2009] [Revised: 05/07/2009] [Accepted: 05/15/2009] [Indexed: 11/21/2022] Open
Abstract
The quality of the early environment, especially during the neonatal period, influences the development of individual differences in resistance to stress and illness in adulthood. A previous study demonstrated that neonatal stress augmented proinflammatory cytokine expression and viral replication in influenza virus-infected adult mice. The goal of the following study was to examine the lifelong effects of neonatal stress on the behavioral response to an immune challenge. Neonatal stress consisted of separating mouse pups from their dams (maternal separation, MSP) at critical points of their development. In the first study, pups were separated from the dam daily for 6h between postnatal day 1 and 14. As adults, these mice were infected with influenza A/PR8 virus. In a second study, a similar paradigm of MSP was employed, and as adults mice were injected with lipopolysaccharide (LPS) (ip). In a third study pups were separated from the dam for 24h on postnatal day 4 or 9. As adults, these mice received ip injections of LPS. In all three studies, changes in body weight, food and sweet solution consumption were examined following immune challenge. As previously described, activation of the immune system using influenza virus infection or LPS administration resulted in sickness behavior that consisted of body weight loss, anorexia and reduced consumption of a sweet solution. Furthermore, neonatal stress induced more rapid kinetics of sickness behavior and augmented several aspects of these symptoms. Together with previous studies, these findings suggest that neonatal stress disrupted the regulation of innate resistance to an immune challenge resulting in enhanced immunological and behavioral responses to immune activation. Thus, long lasting effects of early stress events may be the basis for individual differences in health and susceptibility to disease.
Collapse
|
48
|
Oitzl MS, Champagne DL, van der Veen R, de Kloet ER. Brain development under stress: hypotheses of glucocorticoid actions revisited. Neurosci Biobehav Rev 2009; 34:853-66. [PMID: 19631685 DOI: 10.1016/j.neubiorev.2009.07.006] [Citation(s) in RCA: 250] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2008] [Revised: 06/05/2009] [Accepted: 07/16/2009] [Indexed: 12/23/2022]
Abstract
One of the conundrums in today's stress research is why some individuals flourish and others perish under similar stressful conditions. It is recognized that this individual variability in adaptation to stress depends on the outcome of the interaction of genetic and cognitive/emotional inputs in which glucocorticoid hormones and receptors play a crucial role. Hence one approach towards understanding individual variation in stress coping is how glucocorticoid actions can change from protective to harmful. To address this question we focus on four hypotheses that are connected and not mutual exclusive. First, the classical Glucocorticoid Cascade Hypothesis, in which the inability to cope with chronic stress causes a vicious cycle of excess glucocorticoid and downregulation of glucocorticoid receptors (GR) in the hippocampus triggering a feed-forward cascade of degeneration and disease. Second, the Balance Hypothesis, which takes also the limbic mineralocorticoid receptors (MR) into account and proposes that an integral limbic MR:GR imbalance is causal to altered processing of information in circuits underlying fear, reward, social behaviour and resilience, dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis and impairment of behavioural adaptation. The MR:GR balance is altered by gene variants of these receptor complexes and experience-related factors, which can induce lasting epigenetic changes in the expression of these receptors. A particular potent epigenetic stimulus is the maternal environment which is fundamental for the Maternal Mediation Hypothesis. The outcome of perinatal gene x environment interaction, and thus of MR:GR-mediated functions depends however, on the degree of 'matching' with environmental demands in later life. The Predictive Adaptation Hypothesis therefore presents a conceptual framework to examine the role of glucocorticoids in understanding individual phenotypic differences in stress-related behaviours over the lifespan.
Collapse
Affiliation(s)
- Melly S Oitzl
- Division of Medical Pharmacology, Leiden/Amsterdam Centre for Drug Research (LACDR)/Leiden University Medical Centre (LUMC), University of Leiden, Einsteinweg 55, P.O. Box 9502, 2300 RA Leiden, The Netherlands.
| | | | | | | |
Collapse
|
49
|
Schmidt MV, Sterlemann V, Wagner K, Niederleitner B, Ganea K, Liebl C, Deussing JM, Berger S, Schütz G, Holsboer F, Müller MB. Postnatal glucocorticoid excess due to pituitary glucocorticoid receptor deficiency: differential short- and long-term consequences. Endocrinology 2009; 150:2709-16. [PMID: 19213843 DOI: 10.1210/en.2008-1211] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
A tight regulation of hypothalamic-pituitary-adrenal (HPA) axis activity is essential for successful adaptation to stressful stimuli. Disruption of normal HPA axis development is a main risk factor for diseases such as posttraumatic stress disorder or depression, but the molecular mechanisms that lead to these long-term consequences are poorly understood. Here, we test the hypothesis that the pituitary glucocorticoid receptor (GR) is involved in regulating HPA axis function in neonatal and adult animals. Furthermore, we investigate whether postnatal hypercortisolism induced by pituitary GR deficiency is a main factor contributing to the persistent effects of early-life stress. Conditional knockout mice with a deletion of the GR at the pituitary (GR(POMCCre)) show excessive basal corticosterone levels during postnatal development, but not in adulthood. The hypercortisolemic state of neonatal GR(POMCCre) mice is accompanied by central gene expression changes of CRH and vasopressin in the paraventricular nucleus, but these alterations normalize at later ages. In adult mice, pituitary GR deficiency results in impaired glucocorticoid negative feedback. Furthermore, adult GR(POMCCre) mice display a more active coping strategy in the forced swim test, with no alterations in anxiety like behavior or cognitive functions. Postnatal GR antagonist treatment is able to prevent the long-term behavioral effects in GR(POMCCre) mice. In conclusion, we show that pituitary GRs are centrally involved in regulating HPA axis activity in neonates and mediate negative feedback regulation in adult animals. Postnatal glucocorticoid excess results in an altered stress-coping behavior in adult animals, with no effects on anxiety like behavior or cognition.
Collapse
|
50
|
Liebl C, Panhuysen M, Pütz B, Trümbach D, Wurst W, Deussing JM, Müller MB, Schmidt MV. Gene expression profiling following maternal deprivation: involvement of the brain Renin-Angiotensin system. Front Mol Neurosci 2009; 2:1. [PMID: 19506703 PMCID: PMC2691150 DOI: 10.3389/neuro.02.001.2009] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2009] [Accepted: 04/24/2009] [Indexed: 12/19/2022] Open
Abstract
The postnatal development of the mouse is characterized by a stress hypo-responsive period (SHRP), where basal corticosterone levels are low and responsiveness to mild stressors is reduced. Maternal separation is able to disrupt the SHRP and is widely used to model early trauma. In this study we aimed at identifying of brain systems involved in acute and possible long-term effects of maternal separation. We conducted a microarray-based gene expression analysis in the hypothalamic paraventricular nucleus after maternal separation, which revealed 52 differentially regulated genes compared to undisturbed controls, among them are 37 up-regulated and 15 down-regulated genes. One of the prominently up-regulated genes, angiotensinogen, was validated using in-situ hybridization. Angiotensinogen is the precursor of angiotensin II, the main effector of the brain renin-angiotensin system (RAS), which is known to be involved in stress system modulation in adult animals. Using the selective angiotensin type I receptor [AT(1)] antagonist candesartan we found strong effects on CRH and GR mRNA expression in the brain and ACTH release following maternal separation. AT(1) receptor blockade appears to enhance central effects of maternal separation in the neonate, suggesting a suppressing function of brain RAS during the SHRP. Taken together, our results illustrate the molecular adaptations that occur in the paraventricular nucleus following maternal separation and contribute to identifying signaling cascades that control stress system activity in the neonate.
Collapse
|