1
|
Wang H, Liang Q, Wen Z, Ma W, Ji S, Zhang H, Zhang X. Enriched environment alleviates NLRP3 inflammasome mediated neuroinflammation in diabetes complicated with depression rats. Sci Rep 2025; 15:14214. [PMID: 40269100 PMCID: PMC12019150 DOI: 10.1038/s41598-025-98312-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 04/10/2025] [Indexed: 04/25/2025] Open
Abstract
Depression is a serious and common complication of diabetes, with a well-established link between the two conditions. Recent studies indicate that activation of the NLRP3 inflammasome in hippocampal microglia plays a key role in the pathogenesis of diabetes complicated with depression (DD). While environmental enrichment (EE) has demonstrated significant anti-neuroinflammatory effects, its potential to alleviate neuroinflammation specifically induced by DD remains unclear. The DD rat model was established using a combination of a high-fat diet, streptozotocin injection, and chronic unpredictable mild stress. The effects of EE on NLRP3 inflammasome activation in the hippocampal microglia of DD rats were examined through a series of behavioral tests (open field test, forced swim test, and elevated plus maze), along with enzyme-linked immunosorbent assays, hematoxylin and eosin staining, TdT-mediated dUTP nick-end labeling staining, Western blot analysis, and immunofluorescence. Compared to the control group, DD rats exhibited impaired glucose metabolism and increased depressive-like behaviors, alongside hippocampal neuronal damage and elevated apoptosis rates. Activation of the NLRP3 inflammasome in hippocampal microglia was observed, with upregulation of the NLRP3 signaling pathway and inhibition of the PI3K/AKT signaling pathway. EE significantly mitigated these effects, reducing hippocampal neuroinflammation and alleviating depressive behaviors in DD rats. Neuroinflammation in the hippocampus is a key mechanism underlying the pathogenesis of DD. This study demonstrated that EE reduces neuroinflammation, likely by inhibiting the activation of the NLRP3 inflammasome in hippocampal microglia.
Collapse
Affiliation(s)
- Hongyan Wang
- Sichuan Nursing Vocational College, Chengdu, China
- Southwest University, Chongqing, China
| | - Qingfang Liang
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhifei Wen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wenlian Ma
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shuyang Ji
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hong Zhang
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiangeng Zhang
- Sichuan Nursing Vocational College, Chengdu, China.
- Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
2
|
Randell AM, Salia S, Fowler LF, Aung T, Puts DA, Swift-Gallant A. A meta-analysis of sex differences in neonatal rodent ultrasonic vocalizations and the implication for the preclinical maternal immune activation model. Biol Sex Differ 2025; 16:4. [PMID: 39863873 PMCID: PMC11762899 DOI: 10.1186/s13293-025-00685-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
As the earliest measure of social communication in rodents, ultrasonic vocalizations (USVs) in response to maternal separation are critical in preclinical research on neurodevelopmental disorders (NDDs). While sex differences in both USV production and behavioral outcomes are reported, many studies overlook sex as a biological variable in preclinical NDD models. We aimed to evaluate sex differences in USV call parameters and determine if USVs are differently impacted based on sex in the preclinical maternal immune activation (MIA) model. Results indicate that sex differences in USVs vary with developmental stage and are more pronounced in MIA offspring. Specifically, developmental stage is a moderator of sex differences in USV call duration, with control females emitting longer calls than males in early development (up to postnatal day [PND] 8), but this pattern reverses after PND8. MIA leads to a reduction in call numbers for females compared to same-sex controls in early development, with a reversal post-PND8. MIA decreased call duration and increased total call duration in males, but unlike females, developmental stage did not influence these differences. In males, MIA effects varied by species, with decreased call numbers in rats but increased call numbers in mice. MIA timing (gestational day ≤ 12.5 vs. > 12.5) did not significantly affect results. Our findings highlight the importance of considering sex, developmental timing, and species in USVs research. We discuss how analyzing USV call types and incorporating sex as a biological variable can enhance our understanding of neonatal ultrasonic communication and its translational value in NDD research.
Collapse
Affiliation(s)
- Alison M Randell
- Department of Psychology, Memorial University of Newfoundland and Labrador, St. John's NL, Canada
| | - Stephanie Salia
- Department of Psychology, Memorial University of Newfoundland and Labrador, St. John's NL, Canada
| | - Lucas F Fowler
- Department of Psychology, Memorial University of Newfoundland and Labrador, St. John's NL, Canada
- Cognitive and Behavioural Ecology Program, Memorial University of Newfoundland and Labrador, St. John's NL, Canada
| | - Toe Aung
- Department of Psychology and Counseling, Immaculata University, Immaculata, PA, USA
| | - David A Puts
- Department of Anthropology, Pennsylvania State University, University Park, PA, USA
| | - Ashlyn Swift-Gallant
- Department of Psychology, Memorial University of Newfoundland and Labrador, St. John's NL, Canada
| |
Collapse
|
3
|
Martz J, Shelton MA, Langen TJ, Srinivasan S, Seney ML, Kentner AC. Peripubertal antagonism of corticotropin-releasing factor receptor 1 results in sustained changes in behavioral plasticity and the transcriptomic profile of the amygdala. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.08.14.607957. [PMID: 39185241 PMCID: PMC11343213 DOI: 10.1101/2024.08.14.607957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Peripuberty is a significant period of neurodevelopment with long-lasting effects on the brain and behavior. Blocking type 1 corticotropin-releasing factor receptors (CRFR1) in neonatal and peripubertal rats attenuates detrimental effects of early-life stress on neural plasticity, behavior, and stress hormone action, long after exposure to the drug has ended. CRFR1 antagonism can also impact neural and behavioral development in the absence of stressful stimuli, suggesting sustained alterations under baseline conditions. To investigate this further, we administered the CRFR1 antagonist (CRFR1a) R121919 to young adolescent male and female rats across 4 days. Following each treatment, rats were tested for locomotion, social behavior, mechanical allodynia, or prepulse inhibition (PPI). Acute CRFR1 blockade immediately reduced PPI in peripubertal males, but not females. In adulthood, each assay was repeated without CRFR1a exposure to test for persistent effects of the adolescent treatment. Males continued to experience deficits in PPI while females displayed altered locomotion, PPI, and social behavior. The amygdala was collected to measure long-term effects on gene expression. In the adult amygdala, peripubertal CRFR1a induced alterations in pathways related to neural plasticity and stress in males. In females, pathways related to central nervous system myelination, cell junction organization, and glutamatergic regulation of synaptic transmission were affected. Understanding how acute exposure to neuropharmacological agents can have sustained impacts on brain and behavior, in the absence of further exposures, has important clinical implications for developing adolescents.
Collapse
Affiliation(s)
- Julia Martz
- School of Arts & Sciences, Health Psychology Program,
Massachusetts College of Pharmacy and Health Sciences, Boston Massachusetts, United States
02115
| | - Micah A. Shelton
- Department of Psychiatry, University of Pittsburgh, 450
Technology Drive Pittsburgh, PA, 15219
| | - Tristen J. Langen
- School of Arts & Sciences, Health Psychology Program,
Massachusetts College of Pharmacy and Health Sciences, Boston Massachusetts, United States
02115
| | - Sakhi Srinivasan
- School of Arts & Sciences, Health Psychology Program,
Massachusetts College of Pharmacy and Health Sciences, Boston Massachusetts, United States
02115
| | - Marianne L. Seney
- Department of Psychiatry, University of Pittsburgh, 450
Technology Drive Pittsburgh, PA, 15219
| | - Amanda C. Kentner
- School of Arts & Sciences, Health Psychology Program,
Massachusetts College of Pharmacy and Health Sciences, Boston Massachusetts, United States
02115
| |
Collapse
|
4
|
Martz J, Shelton MA, Geist L, Seney ML, Kentner AC. Sex differences in offspring risk and resilience following 11β-hydroxylase antagonism in a rodent model of maternal immune activation. Neuropsychopharmacology 2024; 49:1078-1090. [PMID: 38007547 PMCID: PMC11109257 DOI: 10.1038/s41386-023-01771-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/21/2023] [Accepted: 11/08/2023] [Indexed: 11/27/2023]
Abstract
Maternal immune activation (MIA) puts offspring at greater risk for neurodevelopmental disorders associated with impaired social behavior. While it is known that immune signaling through maternal, placental, and fetal compartments contributes to these phenotypical changes, it is unknown to what extent the stress response to illness is involved and how it can be harnessed for potential interventions. To this end, on gestational day 15, pregnant rat dams were administered the bacterial mimetic lipopolysaccharide (LPS; to induce MIA) alongside metyrapone, a clinically available 11β-hydroxylase (11βHSD) inhibitor used to treat hypercortisolism in pregnant, lactating, and neonatal populations. Maternal, placental, and fetal brain levels of corticosterone and placental 11βHSD enzymes type 1 and 2 were measured 3-hrs post treatment. Offspring social behaviors were evaluated across critical phases of development. MIA was associated with increased maternal, placental, and fetal brain corticosterone concentrations that were diminished with metyrapone exposure. Metyrapone protected against reductions in placental 11βHSD2 in males only, suggesting that less corticosterone was inactivated in female placentas. Behaviorally, metyrapone-exposure attenuated MIA-induced social disruptions in juvenile, adolescent, and adult males, while females were unaffected or performed worse. Metyrapone-exposure reversed MIA-induced transcriptional changes in monoamine-, glutamate-, and GABA-related genes in adult male ventral hippocampus, but not in females. Taken together, these findings illustrate that MIA-induced HPA responses act alongside the immune system to produce behavioral deficits. As a clinically available drug, the sex-specific benefits and constraints of metyrapone should be investigated further as a potential means of reducing neurodevelopmental risks due to gestational MIA.
Collapse
Affiliation(s)
- Julia Martz
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, 02115, USA
| | - Micah A Shelton
- Department of Psychiatry, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA, 15219, USA
| | - Laurel Geist
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, 02115, USA
| | - Marianne L Seney
- Department of Psychiatry, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA, 15219, USA
| | - Amanda C Kentner
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, 02115, USA.
| |
Collapse
|
5
|
Farmer AL, Lewis MH. Reduction of restricted repetitive behavior by environmental enrichment: Potential neurobiological mechanisms. Neurosci Biobehav Rev 2023; 152:105291. [PMID: 37353046 DOI: 10.1016/j.neubiorev.2023.105291] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 06/04/2023] [Accepted: 06/19/2023] [Indexed: 06/25/2023]
Abstract
Restricted repetitive behaviors (RRB) are one of two diagnostic criteria for autism spectrum disorder and common in other neurodevelopmental and psychiatric disorders. The term restricted repetitive behavior refers to a wide variety of inflexible patterns of behavior including stereotypy, self-injury, restricted interests, insistence on sameness, and ritualistic and compulsive behavior. However, despite their prevalence in clinical populations, their underlying causes remain poorly understood hampering the development of effective treatments. Intriguingly, numerous animal studies have demonstrated that these behaviors are reduced by rearing in enriched environments (EE). Understanding the processes responsible for the attenuation of repetitive behaviors by EE should offer insights into potential therapeutic approaches, as well as shed light on the underlying neurobiology of repetitive behaviors. This review summarizes the current knowledge of the relationship between EE and RRB and discusses potential mechanisms for EE's attenuation of RRB based on the broader EE literature. Existing gaps in the literature and future directions are also discussed.
Collapse
Affiliation(s)
- Anna L Farmer
- Department of Psychology, University of Florida, Gainesville, FL, USA.
| | - Mark H Lewis
- Department of Psychology, University of Florida, Gainesville, FL, USA; Department of Psychiatry, University of Florida, Gainesville, FL, USA
| |
Collapse
|
6
|
Sensory-based interventions in the NICU: systematic review of effects on preterm brain development. Pediatr Res 2022; 92:47-60. [PMID: 34508227 DOI: 10.1038/s41390-021-01718-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 07/12/2021] [Accepted: 08/17/2021] [Indexed: 11/08/2022]
Abstract
BACKGROUND Infants born preterm are known to be at risk for abnormal brain development and adverse neurobehavioral outcomes. To improve early neurodevelopment, several non-pharmacological interventions have been developed and implemented in the neonatal intensive care unit (NICU). Sensory-based interventions seem to improve short-term neurodevelopmental outcomes in the inherently stressful NICU environment. However, how this type of intervention affects brain development in the preterm population remains unclear. METHODS A systematic review of the literature was conducted for published studies in the past 20 years reporting the effects of early, non-pharmacological, sensory-based interventions on the neonatal brain after preterm birth. RESULTS Twelve randomized controlled trials (RCT) reporting short-term effects of auditory, tactile, and multisensory interventions were included after the screening of 1202 articles. Large heterogeneity was identified among studies in relation to both types of intervention and outcomes. Three areas of focus for sensory interventions were identified: auditory-based, tactile-based, and multisensory interventions. CONCLUSIONS Diversity in interventions and outcome measures challenges the possibility to perform an integrative synthesis of results and to translate these for evidence-based clinical practice. This review identifies gaps in the literature and methodological challenges for the implementation of RCTs of sensory interventions in the NICU. IMPACT This paper represents the first systematic review to investigate the effect of non-pharmacological, sensory-based interventions in the NICU on neonatal brain development. Although reviewed RCTs present evidence on the impact of such interventions on the neonatal brain following preterm birth, it is not yet possible to formulate clear guidelines for clinical practice. This review integrates existing literature on the effect of sensory-based interventions on the brain after preterm birth and identifies methodological challenges for the conduction of high-quality RCTs.
Collapse
|
7
|
Reemst K, Ruigrok SR, Bleker L, Naninck EFG, Ernst T, Kotah JM, Lucassen PJ, Roseboom TJ, Pollux BJA, de Rooij SR, Korosi A. Sex-dependence and comorbidities of the early-life adversity induced mental and metabolic disease risks: Where are we at? Neurosci Biobehav Rev 2022; 138:104627. [PMID: 35339483 DOI: 10.1016/j.neubiorev.2022.104627] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 02/15/2022] [Accepted: 03/13/2022] [Indexed: 01/02/2023]
Abstract
Early-life adversity (ELA) is a major risk factor for developing later-life mental and metabolic disorders. However, if and to what extent ELA contributes to the comorbidity and sex-dependent prevalence/presentation of these disorders remains unclear. We here comprehensively review and integrate human and rodent ELA (pre- and postnatal) studies examining mental or metabolic health in both sexes and discuss the role of the placenta and maternal milk, key in transferring maternal effects to the offspring. We conclude that ELA impacts mental and metabolic health with sex-specific presentations that depend on timing of exposure, and that human and rodent studies largely converge in their findings. ELA is more often reported to impact cognitive and externalizing domains in males, internalizing behaviors in both sexes and concerning the metabolic dimension, adiposity in females and insulin sensitivity in males. Thus, ELA seems to be involved in the origin of the comorbidity and sex-specific prevalence/presentation of some of the most common disorders in our society. Therefore, ELA-induced disease states deserve specific preventive and intervention strategies.
Collapse
Affiliation(s)
- Kitty Reemst
- University of Amsterdam, Swammerdam Institute of Life Sciences, Center for Neuroscience, Brain Plasticity Group, Amsterdam, The Netherlands
| | - Silvie R Ruigrok
- University of Amsterdam, Swammerdam Institute of Life Sciences, Center for Neuroscience, Brain Plasticity Group, Amsterdam, The Netherlands
| | - Laura Bleker
- Amsterdam University Medical Center, University of Amsterdam, Department of Epidemiology and Data Science, Amsterdam, The Netherlands
| | - Eva F G Naninck
- University of Amsterdam, Swammerdam Institute of Life Sciences, Center for Neuroscience, Brain Plasticity Group, Amsterdam, The Netherlands
| | - Tiffany Ernst
- Wageningen University, Department of Animal Sciences, Experimental Zoology &Evolutionary Biology Group, Wageningen, The Netherlands
| | - Janssen M Kotah
- University of Amsterdam, Swammerdam Institute of Life Sciences, Center for Neuroscience, Brain Plasticity Group, Amsterdam, The Netherlands
| | - Paul J Lucassen
- University of Amsterdam, Swammerdam Institute of Life Sciences, Center for Neuroscience, Brain Plasticity Group, Amsterdam, The Netherlands; Centre for Urban Mental Health, University of Amsterdam, The Netherlands
| | - Tessa J Roseboom
- Amsterdam University Medical Center, University of Amsterdam, Department of Epidemiology and Data Science, Amsterdam, The Netherlands
| | - Bart J A Pollux
- Wageningen University, Department of Animal Sciences, Experimental Zoology &Evolutionary Biology Group, Wageningen, The Netherlands
| | - Susanne R de Rooij
- Amsterdam University Medical Center, University of Amsterdam, Department of Epidemiology and Data Science, Amsterdam, The Netherlands
| | - Aniko Korosi
- University of Amsterdam, Swammerdam Institute of Life Sciences, Center for Neuroscience, Brain Plasticity Group, Amsterdam, The Netherlands.
| |
Collapse
|
8
|
DeRosa H, Richter T, Wilkinson C, Hunter RG. Bridging the Gap Between Environmental Adversity and Neuropsychiatric Disorders: The Role of Transposable Elements. Front Genet 2022; 13:813510. [PMID: 35711940 PMCID: PMC9196244 DOI: 10.3389/fgene.2022.813510] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 04/13/2022] [Indexed: 12/21/2022] Open
Abstract
Long regarded as “junk DNA,” transposable elements (TEs) have recently garnered much attention for their role in promoting genetic diversity and plasticity. While many processes involved in mammalian development require TE activity, deleterious TE insertions are a hallmark of several psychiatric disorders. Moreover, stressful events including exposure to gestational infection and trauma, are major risk factors for developing psychiatric illnesses. Here, we will provide evidence demonstrating the intersection of stressful events, atypical TE expression, and their epigenetic regulation, which may explain how neuropsychiatric phenotypes manifest. In this way, TEs may be the “bridge” between environmental perturbations and psychopathology.
Collapse
Affiliation(s)
- Holly DeRosa
- Psychology Department, Developmental Brain Sciences Program, College of Liberal Arts, University of Massachusetts Boston, Boston, MA, United States
| | - Troy Richter
- Psychology Department, Developmental Brain Sciences Program, College of Liberal Arts, University of Massachusetts Boston, Boston, MA, United States
| | - Cooper Wilkinson
- Psychology Department, Developmental Brain Sciences Program, College of Liberal Arts, University of Massachusetts Boston, Boston, MA, United States
| | - Richard G Hunter
- Psychology Department, Developmental Brain Sciences Program, College of Liberal Arts, University of Massachusetts Boston, Boston, MA, United States
| |
Collapse
|
9
|
Zhao X, Erickson M, Mohammed R, Kentner AC. Maternal immune activation accelerates puberty initiation and alters mechanical allodynia in male and female C57BL6/J mice. Dev Psychobiol 2022; 64:e22278. [DOI: 10.1002/dev.22278] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/13/2022] [Accepted: 03/09/2022] [Indexed: 12/17/2022]
Affiliation(s)
- Xin Zhao
- School of Arts & Sciences, Health Psychology Program Massachusetts College of Pharmacy and Health Sciences Boston Massachusetts USA
| | - Mary Erickson
- School of Arts & Sciences, Health Psychology Program Massachusetts College of Pharmacy and Health Sciences Boston Massachusetts USA
| | - Ruqayah Mohammed
- School of Arts & Sciences, Health Psychology Program Massachusetts College of Pharmacy and Health Sciences Boston Massachusetts USA
| | - Amanda C. Kentner
- School of Arts & Sciences, Health Psychology Program Massachusetts College of Pharmacy and Health Sciences Boston Massachusetts USA
| |
Collapse
|
10
|
Granata LE, Valentine A, Hirsch JL, Brenhouse HC. Infant ultrasonic vocalizations predict adolescent social behavior in rats: Effects of early life adversity. Dev Psychobiol 2022; 64:e22260. [PMID: 35312059 PMCID: PMC9340574 DOI: 10.1002/dev.22260] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 01/27/2022] [Accepted: 01/31/2022] [Indexed: 12/16/2022]
Abstract
Early life adversity (ELA) increases risk for psychopathologies that often manifest during adolescence and involve disrupted social functioning. ELA affects development of the prefrontal cortex (PFC), which plays a role in social behavior. PFC oxytocin and vasopressin are important regulators of, first, mother-infant attachment, and, later, social behavior, and are implicated in psychiatric disorders. Here, we tested whether infant social communication is predictive of PFC development and adolescent social behavior. We used the limited bedding (LB) ELA model in rats during postnatal days (P)2-14, and measured isolation-induced ultrasonic vocalizations (USVs) at P10 to characterize differences in an early social response. Rats were tested for dyadic social interaction in adolescence (P34). Adolescent oxytocin receptor (Oxtr) and arginine-vasopressin receptor 1a mRNA were measured in the PFC. Relationships between infant USVs, adolescent behavior, and gene expression were assessed. LB-reared rats exhibited fewer USVs at P10. While social behaviors were not robustly affected by rearing, fewer total and complex-type infant USVs predicted fewer interactions in adolescence. LB increased Oxtr in both sexes but Oxtr was not directly predicted by USVs. Findings support the use of USVs as indicators of differential early life experience in rodents, toward further characterization of early factors associated with vulnerability.
Collapse
Affiliation(s)
| | | | - Jason L. Hirsch
- Department of Psychology Northeastern University Boston MA USA
| | | |
Collapse
|
11
|
Kim YM, Lim HH. Association of Early Pubertal Onset in Female Rats With Inhalation of Lavender Oil. J Korean Med Sci 2022; 37:e9. [PMID: 35014224 PMCID: PMC8748666 DOI: 10.3346/jkms.2022.37.e9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 11/12/2021] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Central precocious puberty (CPP) is caused by early activation of the hypothalamic-pituitary-gonadal axis but its major cause remains unclear. Studies have indicated an association between chronic environmental exposure to endocrine-disrupting chemicals and pubertal onset. Essential oil is widely used in homes worldwide for relief of respiratory symptoms, stress, and/or sleep disturbance. METHODS To evaluate this association, we compared the hormone levels and timing of vaginal opening (VO) in female rats exposed to lavender oil (LO) through different routes (study groups: control, LO nasal spray [LS], and indoor exposure to LO [LE]) during the prepubertal period. The body weights of the animals were also compared every 3 days until the day of VO, at which time gonadotropin levels and internal organ weights were assessed. RESULTS The LS group showed early VO at 33.8 ± 1.8 days compared with the control (38.4 ± 2.9 days) and LE (36.6 ± 1.5 days) groups. Additionally, luteinizing hormone levels were significantly higher in the LE and LS groups than those in the control group. Body weights did not differ significantly among the groups. CONCLUSION Inhalation exposure to an exogenic simulant during the prepubertal period might trigger early pubertal onset in female rats. Further evaluation of exposure to other endocrine-disrupting chemicals capable of inducing CPP through the skin, orally, and/or nasally is warranted.
Collapse
Affiliation(s)
- Yoo-Mi Kim
- Department of Pediatrics, College of Medicine, Chungnam National University, Daejeon, Korea
- Department of Pediatrics, Chungnam National University Sejong Hospital, Sejong, Korea.
| | - Han Hyuk Lim
- Department of Pediatrics, College of Medicine, Chungnam National University, Daejeon, Korea
- Department of Pediatrics, Chungnam National University Hospital, Daejeon, Korea
| |
Collapse
|
12
|
Adcock SJJ. Early Life Painful Procedures: Long-Term Consequences and Implications for Farm Animal Welfare. FRONTIERS IN ANIMAL SCIENCE 2021. [DOI: 10.3389/fanim.2021.759522] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Farm animals routinely undergo painful husbandry procedures early in life, including disbudding and castration in calves and goat kids, tail docking and castration in piglets and lambs, and beak trimming in chicks. In rodents, inflammatory events soon after birth, when physiological systems are developing and sensitive to perturbation, can profoundly alter phenotypic outcomes later in life. This review summarizes the current state of research on long-term phenotypic consequences of neonatal painful procedures in rodents and farm animals, and discusses the implications for farm animal welfare. Rodents exposed to early life inflammation show a hypo-/hyper-responsive profile to pain-, fear-, and anxiety-inducing stimuli, manifesting as an initial attenuation in responses that transitions into hyperresponsivity with increasing age or cumulative stress. Neonatal inflammation also predisposes rodents to cognitive, social, and reproductive deficits, and there is some evidence that adverse effects may be passed to offspring. The outcomes of neonatal inflammation are modulated by injury etiology, age at the time of injury and time of testing, sex, pain management, and rearing environment. Equivalent research examining long-term phenotypic consequences of early life painful procedures in farm animals is greatly lacking, despite obvious implications for welfare and performance. Improved understanding of how these procedures shape phenotypes will inform efforts to mitigate negative outcomes through reduction, replacement, and refinement of current practices.
Collapse
|
13
|
Strzelewicz AR, Vecchiarelli HA, Rondón-Ortiz AN, Raneri A, Hill MN, Kentner AC. Interactive effects of compounding multidimensional stressors on maternal and male and female rat offspring outcomes. Horm Behav 2021; 134:105013. [PMID: 34171577 PMCID: PMC8403628 DOI: 10.1016/j.yhbeh.2021.105013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 10/21/2022]
Abstract
Exposure to adverse childhood experiences (ACEs) is a risk factor for the development of psychiatric disorders in addition to cardiovascular associated diseases. This risk is elevated when the cumulative burden of ACEs is increased. Laboratory animals can be used to model the changes (as well as the underlying mechanisms) that result in response to adverse events. In this study, using male and female Sprague Dawley rats, we examined the impact of increasing stress burden, utilizing both two adverse early life experiences (parental/offspring high fat diet + limited bedding exposure) and three adverse early life experiences (parental/offspring high fat diet + limited bedding exposure + neonatal inflammation), on maternal care quality and offspring behavior. Additionally, we measured hormones and hippocampal gene expression related to stress. We found that the adverse perinatal environment led to a compensatory increase in maternal care. Moreover, these dams had reduced maternal expression of oxytocin receptor, compared to standard housed dams, in response to acute stress on postnatal day (P)22. In offspring, the two-hit and three-hit models resulted in a hyperlocomotor phenotype and increased body weights. Plasma leptin and hippocampal gene expression of corticotropin releasing hormone (Chrh)1 and Crhr2 were elevated (males) while expression of oxytocin was reduced (females) following acute stress. On some measures (e.g., hyperlocomotion, leptin), the magnitude of change was lower in the three-hit compared to the two-hit model. This suggests that multiple early adverse events can have interactive, and often unpredictable, impacts, highlighting the importance of modeling complex interactions amongst stressors during development.
Collapse
Affiliation(s)
- Arielle R Strzelewicz
- School of Pharmacy, Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115, United States
| | - Haley A Vecchiarelli
- Divisions of Medical Sciences, University of Victoria, BC V8P 5C2, Canada; Neuroscience Graduate Program, Hotchkiss Brain Institute, Mathison Centre for Mental Health, Research and Education, Cumming School of Medicine, University of Calgary, AB T2N 1N4, Canada
| | - Alejandro N Rondón-Ortiz
- School of Pharmacy, Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115, United States
| | - Anthony Raneri
- School of Arts & Sciences, Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115, United States
| | - Matthew N Hill
- Neuroscience Graduate Program, Hotchkiss Brain Institute, Mathison Centre for Mental Health, Research and Education, Cumming School of Medicine, University of Calgary, AB T2N 1N4, Canada
| | - Amanda C Kentner
- School of Arts & Sciences, Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115, United States.
| |
Collapse
|
14
|
Zhao X, Mohammed R, Tran H, Erickson M, Kentner AC. Poly (I:C)-induced maternal immune activation modifies ventral hippocampal regulation of stress reactivity: prevention by environmental enrichment. Brain Behav Immun 2021; 95:203-215. [PMID: 33766701 PMCID: PMC8187276 DOI: 10.1016/j.bbi.2021.03.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/28/2021] [Accepted: 03/17/2021] [Indexed: 12/14/2022] Open
Abstract
Environmental enrichment (EE) has been successfully implemented in human rehabilitation settings. However, the mechanisms underlying its success are not understood. Incorporating components of EE protocols into our animal models allows for the exploration of these mechanisms and their role in mitigation. Using a mouse model of maternal immune activation (MIA), the present study explored disruptions in social behavior and associated hypothalamic pituitary adrenal (HPA) axis functioning, and whether a supportive environment could prevent these effects. We show that prenatal immune activation of toll-like receptor 3, by the viral mimetic polyinosinic-polycytidylic acid (poly(I:C)), led to disrupted maternal care in that dams built poorer quality nests, an effect corrected by EE housing. Standard housed male and female MIA mice engaged in higher rates of repetitive rearing and had lower levels of social interaction, alongside sex-specific expression of several ventral hippocampal neural stress markers. Moreover, MIA males had delayed recovery of plasma corticosterone in response to a novel social encounter. Enrichment housing, likely mediated by improved maternal care, protected against these MIA-induced effects. We also evaluated c-Fos immunoreactivity associated with the novel social experience and found MIA to decrease neural activation in the dentate gyrus. Activation in the hypothalamus was blunted in EE housed animals, suggesting that the putative circuits modulating social behaviors may be different between standard and complex housing environments. These data demonstrate that augmentation of the environment supports parental care and offspring safety/security, which can offset effects of early health adversity by buffering HPA axis dysregulation. Our findings provide further evidence for the viability of EE interventions in maternal and pediatric settings.
Collapse
Affiliation(s)
| | | | | | | | - Amanda C. Kentner
- Corresponding author: Amanda Kentner, , Office #617-274-3360, Fax # 617-732-2959
| |
Collapse
|
15
|
Wöhr M, Kisko TM, Schwarting RK. Social Behavior and Ultrasonic Vocalizations in a Genetic Rat Model Haploinsufficient for the Cross-Disorder Risk Gene Cacna1c. Brain Sci 2021; 11:brainsci11060724. [PMID: 34072335 PMCID: PMC8229447 DOI: 10.3390/brainsci11060724] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/24/2021] [Accepted: 05/26/2021] [Indexed: 01/27/2023] Open
Abstract
The top-ranked cross-disorder risk gene CACNA1C is strongly associated with multiple neuropsychiatric dysfunctions. In a recent series of studies, we applied a genomically informed approach and contributed extensively to the behavioral characterization of a genetic rat model haploinsufficient for the cross-disorder risk gene Cacna1c. Because deficits in processing social signals are associated with reduced social functioning as commonly seen in neuropsychiatric disorders, we focused on socio-affective communication through 22-kHz and 50-kHz ultrasonic vocalizations (USV). Specifically, we applied a reciprocal approach for studying socio-affective communication in sender and receiver by including rough-and-tumble play and playback of 22-kHz and 50-kHz USV. Here, we review the findings obtained in this recent series of studies and link them to the key features of 50-kHz USV emission during rough-and-tumble play and social approach behavior evoked by playback of 22-kHz and 50-kHz USV. We conclude that Cacna1c haploinsufficiency in rats leads to robust deficits in socio-affective communication through 22-kHz and 50-kHz USV and associated alterations in social behavior, such as rough-and-tumble play behavior.
Collapse
Affiliation(s)
- Markus Wöhr
- Social and Affective Neuroscience Research Group, Laboratory of Biological Psychology, Research Unit Brain and Cognition, Faculty of Psychology and Educational Sciences, KU Leuven, B-3000 Leuven, Belgium
- Leuven Brain Institute, KU Leuven, B-3000 Leuven, Belgium
- Faculty of Psychology, Experimental and Biological Psychology, Behavioral Neuroscience, Philipps-University of Marburg, D-35032 Marburg, Germany; (T.M.K.); (R.K.W.S.)
- Center for Mind, Brain, and Behavior, Philipps-University of Marburg, D-35032 Marburg, Germany
- Correspondence: ; Tel.: +32-16-19-45-57
| | - Theresa M. Kisko
- Faculty of Psychology, Experimental and Biological Psychology, Behavioral Neuroscience, Philipps-University of Marburg, D-35032 Marburg, Germany; (T.M.K.); (R.K.W.S.)
- Center for Mind, Brain, and Behavior, Philipps-University of Marburg, D-35032 Marburg, Germany
| | - Rainer K.W. Schwarting
- Faculty of Psychology, Experimental and Biological Psychology, Behavioral Neuroscience, Philipps-University of Marburg, D-35032 Marburg, Germany; (T.M.K.); (R.K.W.S.)
- Center for Mind, Brain, and Behavior, Philipps-University of Marburg, D-35032 Marburg, Germany
| |
Collapse
|
16
|
Morris AS, Hays-Grudo J, Kerr KL, Beasley LO. The heart of the matter: Developing the whole child through community resources and caregiver relationships. Dev Psychopathol 2021; 33:533-544. [PMID: 33955346 PMCID: PMC8108681 DOI: 10.1017/s0954579420001595] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Numerous developmental scholars have been influenced by the research, policies, and thinking of the late Edward Zigler, who was instrumental in founding Head Start and Early Head Start. In line with the research and advocacy work of Zigler, we discuss two models that support the development of the whole child. We begin by reviewing how adverse and protective experiences "get under the skin" and affect developmental trajectories and risk and resilience processes. We then present research and examples of how experiences affect the whole child, the heart and the head (social, emotional, cognitive, and physical development), and consider development within context and across domains. We discuss examples of interventions that strengthen nurturing relationships as the mechanism of change. We offer a public health perspective on promoting optimal development through nurturing relationships and access to resources during early childhood. We end with a discussion of the myth that our current society is child-focused and argue for radical, essential change to make promoting optimal development for all children the cornerstone of our society.
Collapse
|
17
|
Zhang H, Wong TY, Broekman BFP, Chong YS, Shek LP, Gluckman PD, Tan KH, Meaney MJ, Fortier MV, Qiu A. Maternal Adverse Childhood Experience and Depression in Relation with Brain Network Development and Behaviors in Children: A Longitudinal Study. Cereb Cortex 2021; 31:4233-4244. [PMID: 33825872 DOI: 10.1093/cercor/bhab081] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 02/14/2021] [Accepted: 03/10/2021] [Indexed: 01/28/2023] Open
Abstract
Maternal childhood maltreatment and depression increase risks for the psychopathology of the offspring. This study employed a longitudinal dataset of mother-child dyads to investigate the developmental trajectories of brain functional networks and behaviors of children in relation with maternal childhood adverse experience and depression. Maternal childhood trauma was retrospectively assessed via childhood trauma questionnaire, whereas maternal depressive symptoms were prospectively evaluated during pregnancy and after delivery (n = 518). Child brain scans were acquired at age of 4.5, 6, and 7.5 years (n = 163) and behavioral problems were measured at 7.5 years using the Child Behavior Checklist. We found the functional connectivity of the language network with the sensorimotor, frontal, and attentional networks as a function of maternal adverse experience that interacted with sex and age. Girls exposed to mothers with depressive symptoms or childhood abuse showed the increased development of the functional connectivity of the language network with the visual networks, which was associated with social problems. Girls exposed to mothers with depressive symptoms showed the slower growth of the functional connectivity of the language network with the sensorimotor networks. Our findings, in a community sample, suggest the language network organization as neuroendophenotypes for maternal childhood trauma and depression.
Collapse
Affiliation(s)
- Han Zhang
- School of Computer Engineering and Science, Shanghai University, Shanghai 200444, China.,Department of Biomedical Engineering, National University of Singapore, Singapore 117583, Singapore
| | - Ting-Yat Wong
- Department of Biomedical Engineering, National University of Singapore, Singapore 117583, Singapore
| | - Birit F P Broekman
- Singapore Institute for Clinical Sciences, Singapore 117609, Singapore.,Department of Psychiatry, OLVG and Amsterdam UMC, VU University, Amsterdam 1081 HJ, the Netherlands
| | - Yap-Seng Chong
- Singapore Institute for Clinical Sciences, Singapore 117609, Singapore.,Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore 119228, Singapore
| | - Lynette P Shek
- Department of Pediatrics, Khoo Teck Puat - National University Children's Medical Institute, National University of Singapore, Singapore 119228, Singapore
| | - Peter D Gluckman
- Singapore Institute for Clinical Sciences, Singapore 117609, Singapore
| | - Kok Hian Tan
- Department of Maternal Fetal Medicine, KK Women's and Children's Hospital, Singapore 229899, Singapore
| | - Michael J Meaney
- Singapore Institute for Clinical Sciences, Singapore 117609, Singapore.,Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore.,Douglas Mental Health University Institute, McGill University, Montreal H4H 1R3, Canada
| | - Marielle V Fortier
- Department of Diagnostic and Interventional Imaging, KK Women's and Children's Hospital, Singapore 229899, Singapore
| | - Anqi Qiu
- Department of Biomedical Engineering, National University of Singapore, Singapore 117583, Singapore.,The N.1 Institute for Health, National University of Singapore, Singapore 117456, Singapore.,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| |
Collapse
|
18
|
Henderson HJM, Etem G, Bjorni M, Belnap MA, Rosellini B, Halladay LR. Sex-dependent and ontogenetic effects of low dose ethanol on social behavioral deficits induced by mouse maternal separation. Behav Brain Res 2021; 406:113241. [PMID: 33727047 DOI: 10.1016/j.bbr.2021.113241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 02/06/2021] [Accepted: 03/09/2021] [Indexed: 10/21/2022]
Abstract
Early life stress can induce lifelong emotional and social behavioral deficits that may in some cases be alleviated by drugs or alcohol. A model for early life stress, rodent maternal separation, recapitulates these behavioral sequelae, which are not limited to potentiated anxiety-like behavior, attenuated social motivation, and altered reward-seeking. Here we employed mouse maternal separation with early weaning (MSEW), consisting of pup-dam separation lasting 4-8 hours on postnatal days (PD) 2-16, with early weaning on PD 17. Prior MSEW studies have limited subjects by age or sex, so we more comprehensively investigated MSEW effects in both sexes, during adolescence and adulthood. We found universal effects of MSEW to include lifelong enhancement of anxiety-like and despair behavior, as well as deficits in social motivation. We also observed some sex-dependent effects of MSEW, namely that female MSEW mice exhibited social habituation to a greater degree than their male counterparts. Low dose ethanol administration had no major effects on the social behavior of non-stressed mice. But interestingly, MSEW-induced social habituation was counteracted by low dose ethanol in adolescent female mice, and potentiated in adolescent male mice. These effects were absent in adult animals, suggesting that ethanol may exert differential effects on the developing brain in such a manner to produce age-, sex-, and stress-dependent effects upon social behavior. Together, results indicate that MSEW reliably produces long-lasting impairments in emotional and social behaviors in both sexes and across the lifespan, but may exert more salient social behavioral effects on female animals.
Collapse
Affiliation(s)
- Hannah J M Henderson
- Department of Psychology, Santa Clara University, 500 El Camino Real, Santa Clara, CA, 95053, USA
| | - Gabrielle Etem
- Department of Psychology, Santa Clara University, 500 El Camino Real, Santa Clara, CA, 95053, USA
| | - Max Bjorni
- Department of Psychology, Santa Clara University, 500 El Camino Real, Santa Clara, CA, 95053, USA
| | - Malia A Belnap
- Department of Psychology, Santa Clara University, 500 El Camino Real, Santa Clara, CA, 95053, USA
| | - Bryce Rosellini
- Department of Psychology, Santa Clara University, 500 El Camino Real, Santa Clara, CA, 95053, USA
| | - Lindsay R Halladay
- Department of Psychology, Santa Clara University, 500 El Camino Real, Santa Clara, CA, 95053, USA.
| |
Collapse
|
19
|
Kestering-Ferreira E, Tractenberg SG, Lumertz FS, Orso R, Creutzberg KC, Wearick-Silva LE, Viola TW, Grassi-Oliveira R. Long-term Effects of Maternal Separation on Anxiety-Like Behavior and Neuroendocrine Parameters in Adult Balb/c Mice. CHRONIC STRESS (THOUSAND OAKS, CALIF.) 2021; 5:24705470211067181. [PMID: 34993376 PMCID: PMC8725222 DOI: 10.1177/24705470211067181] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 11/30/2021] [Indexed: 11/17/2022]
Abstract
Introduction: Disruption of maternal care using maternal separation (MS) models has provided significant evidence of the deleterious long-term effects of early life stress. Several preclinical studies investigating MS showed multiple behavioral and biomolecular alterations. However, there is still conflicting results from MS studies, which represents a challenge for reliability and replicability of those findings. Objective: To address that, this study was conducted to investigate whether MS would affect anxiety-like behaviors using a battery of classical tasks, as well as central and peripheral stress-related biomarkers. Methods: Male Balb/c mice were exposed to MS from postnatal day (PND) 2 to 14 for 180-min per day. Two independent cohorts were performed to evaluate both baseline and anxiety-like behavior responses to MS at PND60. We performed composite scores to evaluate MS effects on anxiety and risk assessment phenotypes. Also, we assessed mRNA gene expression in the medial pre-frontal cortex (mPFC) of glucocorticoid and mineralocorticoid receptors (GR and MR) using real-time PCR and peripheral corticosterone levels (CORT) to investigate possible neurobiological correlates to anxiety behaviors. Results: We found increased anxiety-like behavior and decreased risk assessment and exploratory behaviors in MS mice. The animals exposed to MS also presented a decrease in MR mRNA expression and higher levels of CORT compared to controls. Conclusions: Our findings reinforce the body of evidence suggesting that long-term MS induces effects on anxiety and risk assessment phenotypes following the exposure to a standardized MS protocol. Moreover, MS affected the expression of MR mRNA and induced significant changes on CORT response. This data highlights that the reprograming MS effects on HPA axis could be mediate by MR gene expression in mPFC and chronic overactivity of peripheral CORT levels.
Collapse
Affiliation(s)
- Erika Kestering-Ferreira
- Developmental Cognitive Neuroscience Lab
(DCNL), Pontifical University Catholic of Rio Grande do Sul
| | - Saulo Gantes Tractenberg
- Developmental Cognitive Neuroscience Lab
(DCNL), Pontifical University Catholic of Rio Grande do Sul
| | | | - Rodrigo Orso
- Developmental Cognitive Neuroscience Lab
(DCNL), Pontifical University Catholic of Rio Grande do Sul
| | | | | | - Thiago Wendt Viola
- Developmental Cognitive Neuroscience Lab
(DCNL), Pontifical University Catholic of Rio Grande do Sul
| | - Rodrigo Grassi-Oliveira
- Developmental Cognitive Neuroscience Lab
(DCNL), Pontifical University Catholic of Rio Grande do Sul
- Aarhus University, Denmark
| |
Collapse
|
20
|
Neonatal immune challenge induces female-specific changes in social behavior and somatostatin cell number. Brain Behav Immun 2020; 90:332-345. [PMID: 32860938 PMCID: PMC7556772 DOI: 10.1016/j.bbi.2020.08.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 08/11/2020] [Accepted: 08/12/2020] [Indexed: 12/20/2022] Open
Abstract
Decreases in social behavior are a hallmark aspect of acute "sickness behavior" in response to infection. However, immune insults that occur during the perinatal period may have long-lasting consequences for adult social behavior by impacting the developmental organization of underlying neural circuits. Microglia, the resident immune cells of the central nervous system, are sensitive to immune stimulation and play a critical role in the developmental sculpting of neural circuits, making them likely mediators of this process. Here, we investigated the impact of a postnatal day (PND) 4 lipopolysaccharide (LPS) challenge on social behavior in adult mice. Somewhat surprisingly, neonatal LPS treatment decreased sociability in adult female, but not male mice. LPS-treated females also displayed reduced social interaction and social memory in a social discrimination task as compared to saline-treated females. Somatostatin (SST) interneurons within the anterior cingulate cortex (ACC) have recently been suggested to modulate a variety of social behaviors. Interestingly, the female-specific changes in social behavior observed here were accompanied by an increase in SST interneuron number in the ACC. Finally, these changes in social behavior and SST cell number do not appear to depend on microglial inflammatory signaling, because microglia-specific genetic knock-down of myeloid differentiation response protein 88 (MyD88; the removal of which prevents LPS from increasing proinflammatory cytokines such as TNFα and IL-1β) did not prevent these LPS-induced changes. This study provides novel evidence for enduring effects of neonatal immune activation on social behavior and SST interneurons in females, largely independent of microglial inflammatory signaling.
Collapse
|
21
|
Emmons R, Sadok T, Rovero NG, Belnap MA, Henderson HJM, Quan AJ, Del Toro NJ, Halladay LR. Chemogenetic manipulation of the bed nucleus of the stria terminalis counteracts social behavioral deficits induced by early life stress in C57BL/6J mice. J Neurosci Res 2020; 99:90-109. [PMID: 32476178 DOI: 10.1002/jnr.24644] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 03/23/2020] [Accepted: 04/25/2020] [Indexed: 12/11/2022]
Abstract
Trauma during critical periods of development can induce long-lasting adverse effects. To study neural aberrations resulting from early life stress (ELS), many studies utilize rodent maternal separation, whereby pups are intermittently deprived of maternal care necessary for proper development. This can produce adulthood behavioral deficits related to anxiety, reward, and social behavior. The bed nucleus of the stria terminalis (BNST) encodes aspects of anxiety-like and social behaviors, and also undergoes developmental maturation during the early postnatal period, rendering it vulnerable to effects of ELS. Mice underwent maternal separation (separation 4 hr/day during postnatal day (PD)2-5 and 8 hr/day on PD6-16) with early weaning on PD17, which induced behavioral deficits in adulthood performance on two-part social interaction task designed to test social motivation (choice between a same-sex novel conspecific or an empty cup) and social novelty preference (choice between the original-novel conspecific vs. a new-novel conspecific). We used chemogenetics to non-selectively silence or activate neurons in the BNST to examine its role in social motivation and social novelty preference, in mice with or without the history of ELS. Manipulation of BNST produced differing social behavior effects in non-stressed versus ELS mice; social motivation was decreased in non-stressed mice following BNST activation, but unchanged following BNST silencing, while ELS mice showed no change in social behavior after BNST activation, but exhibited enhancement of social motivation-for which they were deficient prior-following BNST silencing. Findings emphasize the BNST as a potential therapeutic target for social anxiety disorders instigated by childhood trauma.
Collapse
Affiliation(s)
- Randi Emmons
- Department of Psychology, Santa Clara University, Santa Clara, CA, USA
| | - Tasneem Sadok
- Department of Psychology, Santa Clara University, Santa Clara, CA, USA
| | - Natalie G Rovero
- Department of Psychology, Santa Clara University, Santa Clara, CA, USA
| | - Malia A Belnap
- Department of Psychology, Santa Clara University, Santa Clara, CA, USA
| | | | - Alex J Quan
- Department of Psychology, Santa Clara University, Santa Clara, CA, USA
| | - Noël J Del Toro
- Department of Psychology, Santa Clara University, Santa Clara, CA, USA
| | | |
Collapse
|
22
|
Synthesizing Views to Understand Sex Differences in Response to Early Life Adversity. Trends Neurosci 2020; 43:300-310. [PMID: 32353334 DOI: 10.1016/j.tins.2020.02.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 02/11/2020] [Accepted: 02/18/2020] [Indexed: 02/06/2023]
Abstract
Sex as a biological variable (SABV) is critical for understanding the broad range of physiological, neurobiological, and behavioral consequences of early life adversity(ELA). The study of the interaction of SABV and ELA ties into several current debates, including the importance of taking into account SABV in research, differing strategies employed by males and females in response to adversity, and the possible evolutionary and developmental mechanisms of altered development in response to adversity. This review highlights the importance of studying both sexes, of understanding sex differences (and similarities) in response to ELA, and provides a context for the debate surrounding whether the response to ELA may be an adaptive process.
Collapse
|
23
|
Environmental influences on placental programming and offspring outcomes following maternal immune activation. Brain Behav Immun 2020; 83:44-55. [PMID: 31493445 PMCID: PMC6906258 DOI: 10.1016/j.bbi.2019.08.192] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 08/15/2019] [Accepted: 08/27/2019] [Indexed: 02/06/2023] Open
Abstract
Adverse experiences during pregnancy induce placental programming, affecting the fetus and its developmental trajectory. However, the influence of 'positive' maternal experiences on the placenta and fetus remain unclear. In animal models of early life stress, environmental enrichment (EE) has ameliorated and even prevented associated impairments in brain and behavior. Here, using a maternal immune activation (MIA) model in rats, we test whether EE attenuates maternal, placental and/or fetal responses to an inflammatory challenge, thereby offering a mechanism by which fetal programming may be prevented. Moreover, we evaluate life-long EE exposure on offspring development and examine a constellation of genes and epigenetic writers that may protect against MIA challenges. In our model, maternal plasma corticosterone and interleukin-1β were elevated 3 h after MIA, validating the maternal inflammatory response. Evidence for developmental programming was demonstrated by a simultaneous decrease in the placental enzymes Hsd11b2 and Hsd11b2/Hsd11b1, suggesting disturbances in glucocorticoid metabolism. Reductions of Hsd11b2 in response to challenge is thought to result in excess glucocorticoid exposure to the fetus and altered glucocorticoid receptor expression, increasing susceptibility to behavioral impairments later in life. The placental, but not maternal, glucocorticoid implications of MIA were attenuated by EE. There were also sustained changes in epigenetic writers in both placenta and fetal brain as a consequence of environmental experience and sex. Following MIA, both male and female juvenile animals were impaired in social discrimination ability. Life-long EE mitigated these impairments, in addition to the sex specific MIA associated disruptions in central Fkbp5 and Oprm1. These data provide the first evidence that EE protects placental functioning during stressor exposure, underscoring the importance of addressing maternal health and well-being throughout pregnancy. Future work must evaluate critical periods of EE use to determine if postnatal EE experience is necessary, or if prenatal exposure alone is sufficient to confer protection.
Collapse
|
24
|
Strzelewicz AR, Ordoñes Sanchez E, Rondón-Ortiz AN, Raneri A, Famularo ST, Bangasser DA, Kentner AC. Access to a high resource environment protects against accelerated maturation following early life stress: A translational animal model of high, medium and low security settings. Horm Behav 2019; 111:46-59. [PMID: 30708031 PMCID: PMC6527488 DOI: 10.1016/j.yhbeh.2019.01.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 12/18/2018] [Accepted: 01/10/2019] [Indexed: 12/21/2022]
Abstract
Early life exposure to a low security setting, characterized by a scarcity of resources and limited food access, increases the risk for psychiatric illness and metabolic dysfunction. We utilized a translational rat model to mimic a low security environment and determined how this manipulation affected offspring behavior, metabolism, and puberty. Because food insecurity in humans is associated with reduced access to healthy food options the "low security" rat manipulation combined a Western diet with exposure to a limited bedding and nesting manipulation (WD-LB). In this setting, dams were provided with limited nesting materials during the pups' early life (P2-P10). This manipulation was contrasted with standard rodent caging (SD) and environmental enrichment (EE), to model "medium security" and "high security" environments, respectively. To determine if transitioning from a low to high security environment improved outcomes, some juvenile WD-LB offspring were exposed to EE. Maternal care was impacted by these environments such that EE dams engaged in high quality care when on the nest, but spent less time on the nest than SD dams. Although WD-LB dams excessively chased their tails, they were very attentive to their pups, perhaps to compensate for limited resources. Offspring exposed to WD-LB only displayed subtle changes in behavior. However, WD-LB exposure resulted in significant metabolic dysfunction characterized by increased body weight, precocious puberty and alterations in the hypothalamic kisspeptin system. These negative effects of WD-LB on puberty and weight regulation were mitigated by EE exposure. Collectively, these studies suggest that both compensatory maternal care and juvenile enrichment can reduce the impact of a low security environment. Moreover, they highlight how utilizing diverse models of resource (in)stability can reveal mechanisms that confer vulnerability and resilience to early life stress.
Collapse
Affiliation(s)
- Arielle R Strzelewicz
- School of Pharmacy, Massachusetts College of Pharmacy and Health Sciences, Boston,MA 02115, United States
| | | | - Alejandro N Rondón-Ortiz
- School of Pharmacy, Massachusetts College of Pharmacy and Health Sciences, Boston,MA 02115, United States
| | - Anthony Raneri
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115, United States
| | - Sydney T Famularo
- Department of Psychology, Temple University, Philadelphia, PA 19122, United States
| | - Debra A Bangasser
- Department of Psychology, Temple University, Philadelphia, PA 19122, United States
| | - Amanda C Kentner
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115, United States.
| |
Collapse
|
25
|
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
|