1
|
Zafar S, Williams C, Joo J, Himes BE, Schneider JS. Developmental lead exposure and aggression in male rats: Influences of maternal care and environmental enrichment. Toxicol Rep 2025; 14:101937. [PMID: 39989979 PMCID: PMC11847132 DOI: 10.1016/j.toxrep.2025.101937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/18/2025] [Accepted: 01/29/2025] [Indexed: 02/25/2025] Open
Abstract
Developmental lead (Pb) exposure results in a variety of cognitive deficits and behavioral issues including increased antisocial behavior and aggression. This study investigated the effect of developmental Pb exposure on aggression and violent behavior in male rats and the potential modulatory influences of quality of maternal care and enriched/non-enriched housing conditions. Long-Evans male rats with or without Pb exposure (perinatal or early postnatal) from low or high maternal care mothers (based on amounts of licking/grooming and arched-back nursing) were randomly assigned to live in enriched or non-enriched environments at weaning. At postnatal day 120-190, offensive aggression was assessed using a resident intruder test. Clinch attack (CAK) frequency and latency, and occurrence of biting events were observed to determine violent behavior. Both perinatal and postnatal Pb-exposed rats were significantly more aggressive and showed more violent behavior, compared to non-Pb-exposed animals, regardless of level of maternal care and environmental enrichment. High maternal care significantly lowered the proportion of animals with short CAK latencies and enriched housing significantly lowered the occurrence of biting events. These results suggest that high maternal care and enriched housing may potentially modify expression of violent aggressive behavior in rats with early life Pb exposure.
Collapse
Affiliation(s)
- Shamaila Zafar
- Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Courtney Williams
- Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Jaehyun Joo
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
- Center of Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Blanca E. Himes
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
- Center of Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Jay S. Schneider
- Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA 19107, United States
- Center of Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| |
Collapse
|
2
|
La Loggia O, Antunes DF, Aubin-Horth N, Taborsky B. Social Complexity During Early Development has Long-Term Effects on Neuroplasticity in the Social Decision-Making Network. Mol Ecol 2025; 34:e17738. [PMID: 40116137 DOI: 10.1111/mec.17738] [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: 07/23/2024] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 03/23/2025]
Abstract
In social species, early social experience shapes the development of appropriate social behaviours during conspecific interactions referred to as social competence. However, the underlying neuronal mechanisms responsible for the acquisition of social competence are largely unknown. A key candidate to influence social competence is neuroplasticity, which functions to restructure neural networks in response to novel experiences or alterations of the environment. One important mediator of this restructuring is the neurotrophin BDNF, which is well conserved among vertebrates. We studied the highly social fish Neolamprologus pulcher, in which the impact of early social experience on social competence has been previously shown. We investigated experimentally how variation in the early social environment impacts markers of neuroplasticity by analysing the relative expression of the bdnf gene and its receptors p75NTR and TrkB across nodes of the social decision-making network. In fish raised in larger groups, bdnf and TrkB were upregulated in the anterior tuberal nucleus, compared to fish raised in smaller groups, while TrkB was downregulated and bdnf was upregulated in the lateral part of the dorsal telencephalon. In the preoptic area (POA), all three genes were upregulated in fish raised in large groups, suggesting that early social experiences might lead to changes of the neuronal connectivity in the POA. Our results highlight the importance of early social experience in programming the constitutive expression of neuroplasticity markers, suggesting that the effects of early social experience on social competence might be due to changes in neuroplasticity.
Collapse
Affiliation(s)
- Océane La Loggia
- Institute for Ecology and Evolution, Behavioural Ecology Division, University of Bern, Bern, Switzerland
| | - Diogo F Antunes
- Institute for Ecology and Evolution, Behavioural Ecology Division, University of Bern, Bern, Switzerland
| | - Nadia Aubin-Horth
- Département de Biologie and Institut de Biologie Intégrative et Des Systèmes, Université Laval, Quebec, Canada
| | - Barbara Taborsky
- Institute for Ecology and Evolution, Behavioural Ecology Division, University of Bern, Bern, Switzerland
| |
Collapse
|
3
|
Lauby SC, Agarwal I, Lapp HE, Salazar M, Semyrenko S, Chauhan D, Margolis AE, Champagne FA. Interplay between prenatal bisphenol exposure, postnatal maternal care, and offspring sex in predicting DNA methylation relevant to anxiety-like behavior in rats. Horm Behav 2025; 172:105745. [PMID: 40273582 DOI: 10.1016/j.yhbeh.2025.105745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 04/04/2025] [Accepted: 04/14/2025] [Indexed: 04/26/2025]
Abstract
Prenatal exposure to endocrine disrupting chemicals, such as bisphenols, can alter neurodevelopmental trajectories and have a lasting neurobehavioral impact through epigenetic pathways. However, outcomes associated with prenatal bisphenol exposure may also be shaped by the postnatal environment and collectively these environmental effects may be sex-specific. Thus, an integrative research design that includes multiple early life exposures and considers sex differences may be essential for predicting outcomes. In the current study, we use a multivariate approach to examine the contributions of prenatal bisphenol exposure, postnatal maternal care, and offspring sex to variation in DNA methylation of well-studied candidate genes (NR3C1, BDNF, OXTR) in the ventral hippocampus and amygdala of adult Long-Evans rats. Main effects of postnatal maternal care and interactions with prenatal bisphenol exposure were consistently found for DNA methylation within the NR3C1 gene (ventral hippocampus) and within the BDNF and OXTR genes (amygdala). Sex-specific effects were also found across all analyses. Overall, our findings suggest that both early-life factors (prenatal and postnatal) and offspring sex contribute to variation in DNA methylation in genes and brain regions relevant for the expression of anxiety-like behavior. These results highlight the need to consider the brain region-specific effects of multiple exposures in males and females to understand the lasting effects of early environments.
Collapse
Affiliation(s)
- Samantha C Lauby
- Department of Psychology, College of Liberal Arts, University of Texas at Austin, Austin, TX, United States of America; Center for Molecular Carcinogenesis and Toxicology, University of Texas at Austin, Austin, TX, United States of America
| | - Isha Agarwal
- Department of Psychology, College of Liberal Arts, University of Texas at Austin, Austin, TX, United States of America
| | - Hannah E Lapp
- Department of Psychology, College of Liberal Arts, University of Texas at Austin, Austin, TX, United States of America
| | - Melissa Salazar
- Department of Psychology, College of Liberal Arts, University of Texas at Austin, Austin, TX, United States of America
| | - Sofiia Semyrenko
- Department of Psychology, College of Liberal Arts, University of Texas at Austin, Austin, TX, United States of America
| | - Danyal Chauhan
- Department of Psychology, College of Liberal Arts, University of Texas at Austin, Austin, TX, United States of America
| | - Amy E Margolis
- Department of Psychiatry, The Ohio State Univeristy, Columbus, OH, United States of America
| | - Frances A Champagne
- Department of Psychology, College of Liberal Arts, University of Texas at Austin, Austin, TX, United States of America; Center for Molecular Carcinogenesis and Toxicology, University of Texas at Austin, Austin, TX, United States of America.
| |
Collapse
|
4
|
Harris S, Kodila Z, Salberg S, Sgro M, Vlassopoulos E, Li CN, Smith MJ, Shultz SR, Yamakawa GR, Noel M, Mychasiuk R. Maternal oxytocin administration mitigates nociceptive, social, and epigenetic impairments in adolescent offspring exposed to perinatal trauma. Neurotherapeutics 2025:e00598. [PMID: 40268660 DOI: 10.1016/j.neurot.2025.e00598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 04/07/2025] [Accepted: 04/09/2025] [Indexed: 04/25/2025] Open
Abstract
Adverse childhood experiences (ACEs) alter brain development, leading to vulnerability for chronic pain, mental health disorders, and suicidality. These effects often emerge during adolescence. Importantly, ACEs can occur prenatally, including when exposed to in utero intimate partner violence (IPV) or postnatally as maternal neglect. Maternal social support has demonstrated promise in the mitigation of ACE-related deficits. Oxytocin, which has a role in social-bonding and stress regulation, serves as a suitable surrogate for social support in preclinical studies. Therefore, we aimed to explore the effects of oxytocin on alleviating social deficits, nociception, and epigenetic changes resulting from models that aimed to mimic the stress normally induced following exposure to two ACEs: IPV in utero and maternal neglect. During pregnancy, dams were randomly assigned to experience the model of IPV or a sham insult. Following birth, offspring from the IPV group underwent 10 days of maternal separation. Dams received three days of oxytocin therapy while nursing. In adolescence, half of the offspring underwent a plantar surgery to induce pain. Overall, in adolescence, rats exposed to the ACEs exhibited increased nociceptive sensitivity and aberrant social interactions, particularly among males, further suggesting that ACEs can increase an individual's risk for chronic pain. The ACEs changed gene expression related to social behaviour and neuroplasticity. Maternal oxytocin normalized pain, social, and gene changes, while oxytocin levels in offspring correlated with nociceptive sensitivity. Although ACEs have enduring consequences, the outcomes are modifiable, and oxytocin may be a robust and implementable therapeutic capable of attenuating early adversity.
Collapse
Affiliation(s)
- Sydney Harris
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Zoe Kodila
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Sabrina Salberg
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Marissa Sgro
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Elaina Vlassopoulos
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Crystal N Li
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Madeleine J Smith
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Sandy R Shultz
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia; Centre for Trauma and Mental Health Research, Vancouver Island University, Nanaimo, B.C., Canada
| | - Glenn R Yamakawa
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Melanie Noel
- Department of Psychology, Alberta Children's Hospital, Hotchkiss Brain Institute, University of Calgary, AB, Canada
| | - Richelle Mychasiuk
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
5
|
Abd-Elmonsif NM, Gamal S, Barsoom SA. Chronic stress and depression impact on tongue and major sublingual gland histology and the potential protective role of Thymus vulgaris: An animal study. Arch Oral Biol 2025; 172:106182. [PMID: 39864188 DOI: 10.1016/j.archoralbio.2025.106182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/12/2024] [Accepted: 01/18/2025] [Indexed: 01/28/2025]
Abstract
OBJECTIVES Reporting the histological effects of chronic stress on certain oral tissues, as well as the capacity of Thymus vulgaris (thyme) to protect tissues from stress and link both serum cortisol and serotonin levels. METHODS 30 rats were randomly divided into a trio of groups: normal control (no treatment), stress group (chronic stress without treatment), and treatment group (chronic stress treated with thyme at a dose of 200 mg/kg BW orally via needle gavage daily for 21 days). At the end of the experiment, tongues and major sublingual glands (SLGs) were surgically removed and processed for histological and histochemical studies. Blood samples were taken shortly before scarification for the biochemical study of cortisol and serotonin serum levels. RESULTS Examination of tongue and SLG sections of the stress group indicated significant alterations in histology and changes in SLG secretion. An examination of tongue and SLG histological sections of the thyme-treated group are showed an improvement. Chronic stress raises cortisol serum levels and lowers serotonin serum levels. CONCLUSIONS Chronic stress causes alteration of the tongue and major SLG histology, as well as changes in SLG secretion. Thyme may protect tissues from stress, and there is a relation between cortisol and serotonin levels.
Collapse
Affiliation(s)
- Nehad M Abd-Elmonsif
- Department of Oral Biology, Faculty of Oral and Dental Medicine, Future University in Egypt, Cairo, Egypt.
| | - Sherif Gamal
- Research Labs Supervisor, Faculty of pharmacy, Future University in Egypt, Cairo, Egypt
| | | |
Collapse
|
6
|
Grígelová A, Mikulecká A, Kubová H. Behavioral comorbidities of early-life seizures: Insights from developmental studies in rats. Epilepsy Behav 2025; 165:110307. [PMID: 40015055 DOI: 10.1016/j.yebeh.2025.110307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 02/01/2025] [Accepted: 02/04/2025] [Indexed: 03/01/2025]
Abstract
Childhood epilepsy is frequently associated with neurobehavioral comorbidities such as depression, anxiety, cognitive impairments, and social dysfunction, as revealed by both clinical and experimental studies. Despite extensive neurophysiological research, behavioral studies in developing animals remain limited and underreported. Here, we review the behavioral impact of early-life seizures (ELSs) in commonly used rat models in developmental studies. We outline suitable tests and provide guidance on how traditional tests should be adapted and interpreted in this context. Finally, we examine factors influencing behavioral analysis in developmental studies, exploring confounding variables and offering strategies to minimize their impact.
Collapse
Affiliation(s)
- Andrea Grígelová
- Developmental Epileptology Institute of Physiology of the Czech Academy of Science Prague Czech Republic; Department of Physiology Faculty of Science Charles University Prague Czech Republic.
| | - Anna Mikulecká
- Developmental Epileptology Institute of Physiology of the Czech Academy of Science Prague Czech Republic
| | - Hana Kubová
- Developmental Epileptology Institute of Physiology of the Czech Academy of Science Prague Czech Republic
| |
Collapse
|
7
|
Hasegawa R, Nakaya K, Kanazawa M, Fukudo S. Corticotropin-releasing hormone receptor-1 antagonist attenuates visceral hypersensitivity induced by trinitrobenzene sulfonic acid colitis and maternal separation in rats. Biopsychosoc Med 2025; 19:5. [PMID: 40155981 PMCID: PMC11951537 DOI: 10.1186/s13030-025-00324-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 01/21/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND The prevailing paradigm for the etiology of irritable bowel syndrome is that transient noxious events lead to long-lasting sensitization of the neural pain circuit, despite complete resolution of the initiating event. In this study, we tested the hypotheses that (1) the combination of maternal separation (MS) and previous colorectal inflammation induces extensive visceral hypersensitivity in rats and (2) visceral hypersensitivity induced by maternal separation and previous colorectal inflammation in rats is mediated via the corticotropin-releasing hormone receptor-1 (CRH-R1) pathway. METHODS Male rat pups were separated from their dams from postnatal day 2 to postnatal day 21. Acute colitis was induced by colorectal administration of trinitrobenzene sulfonic acid (TNBS) or vehicle on postnatal day 8. On postnatal day 50, the visceromotor response was evaluated by electromyography of the abdominal muscle in response to graded (10-80 mmHg) and phasic colorectal distention (CRD) one time. The same experiments were repeated after administration of the selective CRH-R1 antagonist CP-154,526 (20 mg/kg) or vehicle at 45 min before CRD. RESULTS Compared with control rats, visceral perception was increased in MS + TNBS rats. MS + TNBS rats showed a significantly larger visceromotor response to phasic CRD with 40 mmHg, 60 mmHg, and 80 mmHg. Compared with vehicle administration in MS + TNBS rats, administration of CP-154,526 significantly attenuated this visceromotor response to CRD with 40 mmHg, 60 mmHg, and 80 mmHg. CONCLUSIONS These findings suggest that the combination of previous colitis and early life stress induce visceral hypersensitivity, and that the CRH-R1 pathway may play a role in this sensitization.
Collapse
Affiliation(s)
- Ryoko Hasegawa
- Department of Behavioral Medicine, Tohoku University Graduate School of Medicine, 2-1 Seiryo, Aoba, Sendai, 980-8575, Japan
| | - Kumi Nakaya
- Department of Behavioral Medicine, Tohoku University Graduate School of Medicine, 2-1 Seiryo, Aoba, Sendai, 980-8575, Japan
- Division of Epidemiology, School of Public Health, Tohoku University Graduate School of Medicine, Sendai, Japan
- Division of Personalized Prevention and Epidemiology Department of Preventive Medicine and Epidemiology Tohoku Medical Megabank Organization, Sendai, Japan
| | - Motoyori Kanazawa
- Department of Behavioral Medicine, Tohoku University Graduate School of Medicine, 2-1 Seiryo, Aoba, Sendai, 980-8575, Japan
- Department of Psychosomatic Medicine, Tohoku University Hospital, Sendai, Japan
| | - Shin Fukudo
- Department of Behavioral Medicine, Tohoku University Graduate School of Medicine, 2-1 Seiryo, Aoba, Sendai, 980-8575, Japan.
- Department of Psychosomatic Medicine, Tohoku University Hospital, Sendai, Japan.
- Research Center for Accelerator and Radioisotope Science, Tohoku University, Sendai, Japan.
- Department of Psychosomatic Medicine, Japanese Red Cross Ishinomaki Hospital, Ishinomaki, Japan.
| |
Collapse
|
8
|
Taylor WW, Korobkova L, Bhinderwala N, Dias BG. Toward Understanding and Halting Legacies of Trauma. Biol Psychiatry 2025:S0006-3223(25)00108-8. [PMID: 39956254 DOI: 10.1016/j.biopsych.2025.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 02/02/2025] [Accepted: 02/10/2025] [Indexed: 02/18/2025]
Abstract
Echoes of natural and anthropogenic stressors not only reverberate within the physiology, biology, and neurobiology of the generation directly exposed to them but also within the biology of future generations. With the intent of understanding this phenomenon, significant efforts have been made to establish how exposure to psychosocial stress, chemicals, over- and undernutrition, and chemosensory experiences exert multigenerational influences. From these studies, we are gaining new appreciation for how negative environmental events experienced by one generation impact future generations. In this review, we first outline the need to operationally define dimensions of negative environmental events in the laboratory and the routes by which the impact of such events are felt through generations. Next, we discuss molecular processes that cause the effects of negative environmental events to be initiated in the exposed generation and then perpetuated across generations. Finally, we discuss how legacies of flourishing can be engineered to halt or reverse multigenerational influences of negative environmental events. In summary, this review synthesizes our current understanding of the concept, causes, and consequences of multigenerational echoes of stress and looks for opportunities to halt them.
Collapse
Affiliation(s)
- William Wesley Taylor
- Neuroscience Graduate Program, University of Southern California, Los Angeles, California; Developmental Neuroscience and Neurogenetics Program, The Saban Research Institute, Los Angeles, California
| | - Laura Korobkova
- Neuroscience Graduate Program, University of Southern California, Los Angeles, California; Developmental Neuroscience and Neurogenetics Program, The Saban Research Institute, Los Angeles, California
| | - Nabeel Bhinderwala
- Developmental Neuroscience and Neurogenetics Program, The Saban Research Institute, Los Angeles, California
| | - Brian George Dias
- Developmental Neuroscience and Neurogenetics Program, The Saban Research Institute, Los Angeles, California; Division of Endocrinology, Diabetes and Metabolism, Children's Hospital Los Angeles, Los Angeles, California; Department of Pediatrics, Keck School of Medicine of the University of Southern California, Los Angeles, California.
| |
Collapse
|
9
|
Vernick J, Martin C, Montelpare W, Dunham AE, Overall KL. Understanding the Influence of Early-Life Stressors on Social Interaction, Telomere Length, and Hair Cortisol Concentration in Homeless Kittens. Animals (Basel) 2025; 15:446. [PMID: 39943216 PMCID: PMC11815723 DOI: 10.3390/ani15030446] [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: 12/02/2024] [Revised: 01/14/2025] [Accepted: 02/04/2025] [Indexed: 02/16/2025] Open
Abstract
The early postnatal period is a critical neurodevelopmental stage characterized by rapid neural maturation and is adversely affected by early-life stressors. This study explored the behavioural, physiological, and epigenetic consequences of early-life stress in a population of homeless rescue kittens. This longitudinal study included 50 kittens rescued and placed into foster care by the Prince Edward Island Humane Society. They underwent behavioural testing at 8, 10, and 12 weeks of age. Hair cortisol concentration was measured at 8 weeks and served as a physiological marker of the previous 3 months' cumulative stress response, which, for these kittens, included the late gestation period. A blood sample for relative telomere length measurement was taken at 10-12 weeks to estimate epigenetic changes as young kittens. Data were analyzed with respect to age and performance in all repeated measures tests, status as a stray or a surrender, and the presence of the dam in their foster homes. As expected, the performance of kittens in all tests changed over the 5 weeks of testing. Kittens separated from their mothers exhibited significantly higher hair cortisol concentrations (p = 0.02) and elongated relative telomere lengths (p = 0.04). No correlation was found between hair cortisol concentration and relative telomere lengths (p = 0.99). These results support the need for further study on the effects of epigenetics and early-life stress, both in kittens and across species.
Collapse
Affiliation(s)
- Jennifer Vernick
- Department of Health Management, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, PE C1A 4P3, Canada
| | - Chelsea Martin
- Department of Microbiology and Pathology, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, PE C1A 4P3, Canada;
| | - William Montelpare
- Department of Applied Human Sciences, Faculty of Science and Faculty of Nursing, University of Prince Edward Island, Charlottetown, PE C1A 4P3, Canada;
| | - Arthur E. Dunham
- Biology Department, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Karen L. Overall
- Department of Health Management, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, PE C1A 4P3, Canada
| |
Collapse
|
10
|
Takahashi A. The role of social isolation stress in escalated aggression in rodent models. Neurosci Res 2025; 211:75-84. [PMID: 35917930 DOI: 10.1016/j.neures.2022.07.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 07/14/2022] [Accepted: 07/27/2022] [Indexed: 11/26/2022]
Abstract
Anti-social behavior and violence are major public health concerns. Globally, violence contributes to more than 1.6 million deaths each year. Previous studies have reported that social rejection or neglect exacerbates aggression. In rodent models, social isolation stress is used to demonstrate the adverse effects of social deprivation on physiological, endocrinological, immunological, and behavioral parameters, including aggressive behavior. This review summarizes recent rodent studies on the effect of social isolation stress during different developmental periods on aggressive behavior and the underlying neural mechanisms. Social isolation during adulthood affects the levels of neurosteroids and neuropeptides and increases aggressive behavior. These changes are ethologically relevant for the adaptation to changes in local environmental conditions in the natural habitats. Chronic deprivation of social interaction after weaning, especially during the juvenile to adolescent periods, leads to the disruption of the development of appropriate social behavior and the maladaptive escalation of aggressive behavior. The understanding of neurobiological mechanisms underlying social isolation-induced escalated aggression will aid in the development of therapeutic interventions for escalated aggression.
Collapse
Affiliation(s)
- Aki Takahashi
- Laboratory of Behavioral Neurobiology, Faculty of Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan.
| |
Collapse
|
11
|
Bentele UU, Strobel P, Meier M, Benz ABE, Gaertner RJ, Klink ESC, Denk BF, Dimitroff SJ, Unternaehrer E, Pruessner JC. The effect of mortality salience and early-life maternal care on neuroendocrine, autonomic, and psychological stress responses. Sci Rep 2025; 15:1349. [PMID: 39779773 PMCID: PMC11711282 DOI: 10.1038/s41598-025-85380-w] [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: 08/28/2024] [Accepted: 01/02/2025] [Indexed: 01/11/2025] Open
Abstract
Adverse early-life experiences alter the regulation of major stress systems such as the hypothalamic-pituitary-adrenal (HPA) axis. Low early-life maternal care (MC) has repeatedly been related to blunted cortisol stress responses. Likewise, an acutely increased awareness of mortality (mortality salience [MS]) also has been shown to blunt cortisol responses. In this study we investigated the effects of early-life MC and a potential interaction with MS on HPA axis responsivity, as well as autonomic and subjective stress responses. Seventy-three women (Mage=21.56, SDage=2.85) with self-reported low (n = 30) or high (n = 43) early-life MC, underwent the Trier Social Stress Test for groups. Before, they were asked to briefly contemplate either death (mortality condition, n = 38) or sleep (control condition, n = 35). Salivary cortisol and alpha amylase, heart rate variability and subjective stress levels were assessed repeatedly. Multilevel mixed models confirmed an effect of MC on stress system regulation, indicated by blunted cortisol responses and overall reduced heart rate variability in low versus high MC individuals. Moreover, we found an interaction between MS and MC concerning subjective stress and autonomic measures. Specifically, low MC individuals in the control compared to the mortality condition showed both overall higher subjective stress levels, and less increase in heart rate variability following stress. These findings demonstrate the enduring impact of low early-life MC and the potential role of acute mortality primes on the regulation of stress systems in healthy women.
Collapse
Affiliation(s)
- Ulrike U Bentele
- Department of Psychology, Division of Neuropsychology, University of Constance, Fach 905, Universitaetsstrasse 10, 78464, Constance, Germany.
| | - Paula Strobel
- Department of Psychology, Division of Neuropsychology, University of Constance, Fach 905, Universitaetsstrasse 10, 78464, Constance, Germany
| | - Maria Meier
- Department of Psychology, Division of Neuropsychology, University of Constance, Fach 905, Universitaetsstrasse 10, 78464, Constance, Germany
- Child- and Adolescent Research Department, University Psychiatric Clinics Basel (UPK), University of Basel, Basel, Switzerland
| | - Annika B E Benz
- Department of Psychology, Division of Neuropsychology, University of Constance, Fach 905, Universitaetsstrasse 10, 78464, Constance, Germany
| | - Raphaela J Gaertner
- Department of Psychology, Division of Neuropsychology, University of Constance, Fach 905, Universitaetsstrasse 10, 78464, Constance, Germany
| | - Elea S C Klink
- Department of Psychology, Division of Neuropsychology, University of Constance, Fach 905, Universitaetsstrasse 10, 78464, Constance, Germany
| | - Bernadette F Denk
- Department of Psychology, Division of Neuropsychology, University of Constance, Fach 905, Universitaetsstrasse 10, 78464, Constance, Germany
- Centre for the Advanced Study of Collective Behaviour, University of Constance, Constance, Germany
| | | | - Eva Unternaehrer
- Department of Psychology, Division of Neuropsychology, University of Constance, Fach 905, Universitaetsstrasse 10, 78464, Constance, Germany
- Child- and Adolescent Research Department, University Psychiatric Clinics Basel (UPK), University of Basel, Basel, Switzerland
| | - Jens C Pruessner
- Department of Psychology, Division of Neuropsychology, University of Constance, Fach 905, Universitaetsstrasse 10, 78464, Constance, Germany
- Centre for the Advanced Study of Collective Behaviour, University of Constance, Constance, Germany
| |
Collapse
|
12
|
Frasch MG, Wakefield C, Janoschek B, Frank YS, Karp F, Reyes N, Desrochers A, Wallingford MC, Antonelli MC, Metz GAS. Perinatal Psychoneuroimmunology of Prenatal Stress and Its Effects on Fetal and Postnatal Brain Development. Methods Mol Biol 2025; 2868:303-332. [PMID: 39546237 DOI: 10.1007/978-1-0716-4200-9_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
Prenatal stress (PS) impacts early behavioral, neuroimmune, and cognitive development. Pregnant rat models have been very valuable in examining the mechanisms of such fetal programming. A pregnant sheep model of maternal stress offers the unique advantages of chronic in utero monitoring and manipulation. This chapter presents the techniques used to model single and multigenerational stress exposures and their pleiotropic effects on the offspring.
Collapse
Affiliation(s)
- Martin G Frasch
- Department of Obstetrics and Gynecology and Institute on Human Development and Disability, University of Washington, Seattle, WA, USA.
| | - Colin Wakefield
- Department of Obstetrics and Gynecology and Institute on Human Development and Disability, University of Washington, Seattle, WA, USA
| | - Ben Janoschek
- Department of Obstetrics and Gynecology and Institute on Human Development and Disability, University of Washington, Seattle, WA, USA
| | - Yael S Frank
- Department of Obstetrics and Gynecology and Institute on Human Development and Disability, University of Washington, Seattle, WA, USA
| | - Floyd Karp
- Departments of Pharmacy and Bioengineering, University of Washington, Seattle, WA, USA
| | - Nicholas Reyes
- Department of Comparative Medicine, University of Washington, Seattle, WA, USA
| | - Andre Desrochers
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC, Canada
| | - Mary C Wallingford
- Mother Infant Research Institute, Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
- Department of Obstetrics and Gynecology, Tufts University School of Medicine, Boston, MA, USA
| | - Marta C Antonelli
- Department of Obstetrics and Gynecology, Klinikum Rechts Der Isar, Technical University of Munich, Munich, Germany
- Instituto de Biología Celular y Neurociencia "Prof. Eduardo De Robertis", Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Gerlinde A S Metz
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
| |
Collapse
|
13
|
Núñez-Murrieta MA, Rendón-Candanedo E, Angeles-Torres D, Noguez P, Coria-Avila GA, Bolado-García VE, Corona-Morales AA. Maternal Sensitization in Virgin Female Rats: Behavioral Effects of Enriched Environment. Dev Psychobiol 2025; 67:e70010. [PMID: 39648287 DOI: 10.1002/dev.70010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 11/05/2024] [Accepted: 11/18/2024] [Indexed: 12/10/2024]
Abstract
In virgin female rats, the continuous presence of pups causes them to express typical maternal behaviors, a process known as maternal sensitization. Previous experience with pups accelerates maternal sensitization. It is also known that in primiparous rats, enriched environment (EE) increases the expression of maternal behaviors. Here, we investigated whether experience, other than pup exposure, affects the process of maternal sensitization and hypothesized that EE increases the expression of maternal behaviors and maternal motivation in virgin rats. Virgin adult females were housed in standard conditions or physical and social EE for 21 days. Then, females were exposed daily to pups until they expressed full maternal behaviors. Thereafter, females performed pup preference, pup motivation in a novel context, and resident-intruder tests. We found that initial pup rejection was higher in EE rats, but eventually, both groups became maternally sensitized simultaneously. The frequency and duration of pup licking were higher in EE rats. In the other tests, EE rats exhibited more entries to the open arms, retrieved more pups toward the closed arms, and were more aggressive towards an intruder. We conclude that housing in a social EE modulates aspects of the pup-induced maternal sensitization, particularly increasing pup licking and motivation in both familiar and novel circumstances.
Collapse
Affiliation(s)
- Mauricio A Núñez-Murrieta
- Maestría en Neuroetología y Doctorado en Investigaciones Cerebrales, Universidad Veracruzana, Xalapa, Veracruz, México
| | | | - Diana Angeles-Torres
- Doctorado en Ciencias Biomédicas, Universidad Veracruzana, Xalapa, Veracruz, México
| | - Paula Noguez
- Tecnológico Nacional de México, Instituto Tecnológico Superior de Xalapa, Xalapa, Veracruz, México
| | - Genaro A Coria-Avila
- Instituto de Investigaciones Cerebrales, Universidad Veracruzana, Xalapa, Veracruz, México
| | - Victoria E Bolado-García
- Laboratorio de Investigación Genómica y Fisiológica, Facultad de Nutrición, Universidad Veracruzana, Xalapa, Veracruz, México
| | - Aleph A Corona-Morales
- Laboratorio de Investigación Genómica y Fisiológica, Facultad de Nutrición, Universidad Veracruzana, Xalapa, Veracruz, México
| |
Collapse
|
14
|
Gildawie KR, Budge KE, Vassoler FM, Yen E, Byrnes EM. Differential Effects of Prenatal Buprenorphine and Methadone on Postnatal Growth and Gene Expression in the Nucleus Accumbens. Dev Psychobiol 2025; 67:e70015. [PMID: 39648276 PMCID: PMC11709121 DOI: 10.1002/dev.70015] [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: 07/30/2024] [Revised: 10/27/2024] [Accepted: 11/20/2024] [Indexed: 12/10/2024]
Abstract
Methadone and buprenorphine are commonly prescribed during pregnancy to maintain recovery and prevent symptoms of withdrawal in women with opioid use disorder. Infants prenatally exposed to medications for opioid use disorder (MOUD), however, commonly show signs of neonatal opioid withdrawal syndrome (NOWS), which can include feeding-related issues like hyperphagia. To investigate the effects of prenatal MOUD exposure on feeding behavior, female Sprague-Dawley rats were implanted with osmotic minipumps filled with methadone, buprenorphine, or saline and subsequently mated. On postnatal day (PND) 1, buprenorphine- and methadone-exposed offspring weighed less than saline-exposed subjects. Throughout early postnatal development (PND2, 7, and 12), this reduction in weight persisted in buprenorphine, but not methadone, offspring. RNAscope in situ hybridization was then used to measure expression of genes in the nucleus accumbens (NAc) previously associated with hyperphagia in NOWS infants, including proopiomelanocortin (Pomc), neuropeptide Y2 receptors (Npy2r), and dopamine type 2 receptors (Drd2). Distinct developmental expression patterns were noted across the postnatal period, with few effects of MOUD; however, significantly lower Pomc expression was observed in methadone-exposed but not buprenorphine-exposed offspring. These findings demonstrate differential effects of methadone and buprenorphine on offspring development and gene expression, highlighting differences in offspring outcomes associated with these two MOUDs.
Collapse
Affiliation(s)
- Kelsea R Gildawie
- Department of Comparative Pathobiology, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, USA
- Department of Psychology, Simmons University, Boston, Massachusetts, USA
| | - Kerri E Budge
- Department of Comparative Pathobiology, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, USA
| | - Fair M Vassoler
- Department of Comparative Pathobiology, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, USA
| | - Elizabeth Yen
- Department of Pediatrics, Tufts University School of Medicine, Boston, Massachusetts, USA
- Mother Infant Research Institute, Tufts Medical Center, Boston, Massachusetts, USA
| | - Elizabeth M Byrnes
- Department of Comparative Pathobiology, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, USA
| |
Collapse
|
15
|
Zhang S, Han T, Yang R, Song Y, Jiang W, Tian Z. Unraveling the influence of childhood emotional support on adult aging: Insights from the UK Biobank. Arch Gerontol Geriatr 2024; 127:105600. [PMID: 39151235 DOI: 10.1016/j.archger.2024.105600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 08/05/2024] [Accepted: 08/09/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND Exploring the association between Childhood Emotional Support (CES) and the mechanisms of aging is pivotal for understanding its potential to lessen the incidence of age-related pathologies and promote a milieu for healthy aging. METHODS Utilizing data from the UK Biobank comprising nearly 160,000 individuals, comprehensive analyses were conducted to explore associations between CES levels and age-related diseases, biological age and aging hallmarks. Cox proportional hazards regression models were used to investigate the relationship between CES and the risk of hospitalization for age-related diseases. Linear regression models were employed to explore the associations between CES and the frailty index (FI), Klemera-Doubal method (KDM) biological age acceleration, homeostatic dysregulation (HD), C-reactive protein (CRP), white blood cell (WBC) count, and telomere length. RESULTS The analyses revealed a significant association between higher CES levels and a decreased risk of hospitalization for age-related diseases in later life. After adjustments for covariates, the hazard ratio for age-related diseases was 0.87 (95 % confidence interval, 0.83-0.91, p < 0.001) in those with the highest CES level compared to those with the lowest CES level. Participants with the highest CES level exhibited lower FI scores (coefficient = -0.033, p < 0.001), reduced CRP level (coefficient = -0.097, p < 0.05) and lower WBC counts (coefficient = -0.034, p < 0.05). Stratified analyses based on genetic susceptibility further elucidated the protective role of CES against age-related diseases. CONCLUSION These findings underscore the potential of early interventions targeting CES to promote healthy aging and alleviating the burden of age-related diseases.
Collapse
Affiliation(s)
- Shibo Zhang
- Department of Pediatrics, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, China
| | - Tianshu Han
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Harbin, Heilongjiang, China
| | - Ruiming Yang
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Harbin, Heilongjiang, China
| | - Yuxin Song
- Department of Pediatrics, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, China
| | - Wenbo Jiang
- Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Harbin, Heilongjiang, China.
| | - Zhiliang Tian
- Department of Pediatrics, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, China.
| |
Collapse
|
16
|
Arusha KS, Boadi KD, Ellah SS, Kim D, Bauer CM. Sibling presence during fostering ameliorates endocrine stress profile changes in a social rodent species (Octodon degus) in a sex-specific manner. Horm Behav 2024; 166:105660. [PMID: 39500218 DOI: 10.1016/j.yhbeh.2024.105660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 10/30/2024] [Accepted: 11/01/2024] [Indexed: 11/29/2024]
Abstract
During early life, disruption of the parent-offspring bond can substantially impact development of offspring physiology and behavior. In rodents, it has been well-documented that parental separation, reduction in parental care, and cross-fostering can affect development of the endocrine stress response. For social species, however, several social factors may mitigate the stress of cross-fostering, such as remaining with other known adult caregivers or siblings. In this study, we cross-fostered a social rodent species (Octodon degus) with or without their siblings at postnatal day (PND) 8 and measured their endocrine stress response immediately after fostering (PND9) and at weaning (PND28). We found that female singly-fostered offspring displayed elevated baseline cortisol levels and reduced weight gain immediately after fostering. At weaning, female singly-fostered offspring continued to display elevated baseline cortisol levels compared to non-fostered female offspring, while singly-fostered males demonstrated weaker cortisol negative feedback strength compared to male offspring that were not fostered or were fostered with their siblings. These results suggest that sibling presence may help mitigate the stress of fostering, and that future studies should further examine other social conditions that may help reduce developmental consequences of long-term parental bond disruption.
Collapse
Affiliation(s)
- Kaja S Arusha
- Department of Biology, Swarthmore College, Swarthmore, PA, USA
| | - Krystle D Boadi
- Department of Biology, Swarthmore College, Swarthmore, PA, USA
| | - Sabrina S Ellah
- Department of Biology, Swarthmore College, Swarthmore, PA, USA
| | - Daniela Kim
- Department of Biology, Swarthmore College, Swarthmore, PA, USA
| | - Carolyn M Bauer
- Department of Biology, Swarthmore College, Swarthmore, PA, USA.
| |
Collapse
|
17
|
Cassiano LMG, de Queiroz KB, Martins TVDA, Maia ALA, Rachid MA, Spencer SJ, Coimbra RS. Neonatal overfeeding attenuates microgliosis and hippocampal damage in an infant rat model of pneumococcal meningitis. Front Immunol 2024; 15:1429157. [PMID: 39469711 PMCID: PMC11513286 DOI: 10.3389/fimmu.2024.1429157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 09/30/2024] [Indexed: 10/30/2024] Open
Abstract
Background Pneumococcal meningitis (PM) triggers apoptotic neuronal and progenitor cell death in the hippocampal dentate gyrus (DG), resulting in subsequent cognitive impairment. Microglia play a crucial role in PM-induced hippocampal damage. While the lasting effects of neonatal nutrition on health are well documented, the influence of early-life overfeeding on the host response to neuroinfections remains uncertain. This study aimed to examine whether neonatal overfeeding affects the outcome of PM in the hippocampus (HC). Material and methods Overfeeding was induced by adjusting litter size immediately after birth. On the eleventh day of life, rats were intracisternally injected with Streptococcus pneumoniae or saline, followed by euthanasia after 24 hours for brain dissection. Histological analysis evaluated apoptosis in the DG and the extent of inflammatory infiltrate in the hippocampal fissure, while microgliosis was assessed by immunohistochemistry. The hippocampal transcriptome was analyzed using RNAseq, and the mRNA levels of specific inflammatory biomarkers were evaluated via RT-qPCR. Results Overfed rats exhibited 40.5% greater body mass compared to their normal-fed counterparts. Intriguingly, PM-induced apoptosis in the DG was 50% lower in overfed rats. This effect was accompanied by significant alterations in the hippocampal transcriptional profile, particularly the lack of activation of the Programmed cell death pathway in overfed infected animals. RT-qPCR analysis of Aif1 and examination of Iba1-immunostained cells revealed mild microgliosis in the HC of infected-overfed animals. This reduced microglial reaction may be attributed to the diminished activation of interferon signaling pathways, as disclosed by the transcriptome analysis, potentially preventing microglial priming. Additionally, evidence of reduced neuroinflammation in overfed rats with PM was observed through the milder activation of pathways associated with Toll-like receptors, interleukins, and chemokine signaling. Furthermore, overfed animals exhibited increased transcription of proinflammatory Il6 and anti-inflammatory Il10 genes, with the latter showing higher expression even in the absence of PM, suggesting a priming effect of overfeeding on hippocampal immune cells. Conclusion This study sheds light on the complex interplay between early-life overfeeding, immune response, and neuroprotection in infant rats with PM. The findings demonstrate the neuroprotective impact of early-life overfeeding in the context of PM, linked to the modulation of microglial function.
Collapse
Affiliation(s)
- Larissa Marcely Gomes Cassiano
- Neurogenômica, Imunopatologia, Instituto René Rachou (IRR), Fiocruz, Belo Horizonte, Brazil
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | | | - Thais Veronez de Andrade Martins
- Serviço de Animais de Laboratório (SAL), Instituto Nacional de Controle de Qualidade em Saúde (INCQS), Fiocruz, Rio de Janeiro, Brazil
| | - Ana Luiza Azevedo Maia
- Neurogenômica, Imunopatologia, Instituto René Rachou (IRR), Fiocruz, Belo Horizonte, Brazil
| | - Milene Alvarenga Rachid
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Sarah J. Spencer
- School of Health Sciences and Biomedical Sciences, Royal Melbourne Institute of Technology (RMIT) University, Melbourne, VIC, Australia
| | - Roney Santos Coimbra
- Neurogenômica, Imunopatologia, Instituto René Rachou (IRR), Fiocruz, Belo Horizonte, Brazil
| |
Collapse
|
18
|
Schneider JS, Williams C, Zafar S, Joo J, Himes BE. Influences of quality of maternal care and environmental enrichment on associative memory function in rats with early life lead exposure. Brain Behav 2024; 14:e70040. [PMID: 39295102 PMCID: PMC11410876 DOI: 10.1002/brb3.70040] [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: 04/04/2024] [Revised: 07/11/2024] [Accepted: 08/02/2024] [Indexed: 09/21/2024] Open
Abstract
INTRODUCTION Children in low socioeconomic status (SES) communities are at higher risk of exposure to lead (Pb) and potentially more severe adverse outcomes from Pb exposures. While the factors encompassing SES are complex, low SES households often have less enriching home environments and parent-child interactions. This study investigated the extent to which environmental/behavioral factors (quality of maternal care and richness of the postnatal environment) may modify adverse effects from Pb exposure. METHODS Long-Evans female rats were randomly assigned to Control (no Pb), Early Postnatal (EPN: birth through weaning), or Perinatal (PERI: 14 days pre-mating through weaning) Pb exposure groups. From postnatal days (PNDs) 2-9, maternal care behaviors were observed, and dams were classified as low or high maternal care based on amounts of licking/grooming and arched back nursing. At weaning, pups were randomly assigned to enriched or non-enriched environments. At PND 55, animals began trace fear conditioning and associative memory was tested on days 1, 2, and 10 postconditioning. RESULTS Control offspring showed no significant effects of maternal care or enrichment on task performance. Females with EPN-Pb exposure and males with PERI-Pb exposure living in the non-enriched environment and having an LMC mother had significant memory impairments at days 2 and 10 that were not observed in comparably housed animals with HMC mothers. Enriched animals had no deficits, regardless of maternal care status. CONCLUSION These results show the potential for modulatory influences of maternal care and housing environment on protecting against or reversing at least one aspect of Pb-induced cognitive/behavioral dysfunction.
Collapse
Affiliation(s)
- Jay S. Schneider
- Department of Pathology and Genomic MedicineThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
- Center of Excellence in Environmental Toxicology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Courtney Williams
- Department of Pathology and Genomic MedicineThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Shamaila Zafar
- Department of Pathology and Genomic MedicineThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Jaehyun Joo
- Center of Excellence in Environmental Toxicology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Blanca. E. Himes
- Center of Excellence in Environmental Toxicology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
19
|
Abrahamyan A, Lucas R, Severo M, Talih M, Fraga S. Association between adverse childhood experiences and bodily pain in early adolescence. Stress Health 2024; 40:e3383. [PMID: 38358262 DOI: 10.1002/smi.3383] [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: 09/27/2023] [Revised: 12/19/2023] [Accepted: 02/05/2024] [Indexed: 02/16/2024]
Abstract
We aimed to examine the relationship between lifetime exposure to adverse childhood experiences (ACEs) during the first decade of life and recent pain features reported in early adolescence. We conducted a prospective study using data from 4564 adolescent Generation XXI birth cohort participants recruited in 2005-2006. Adverse childhood experiences were reported by children at ages 10 and 13 years using a 15-item questionnaire. Recent pain features (e.g., any pain, pain sites, recurrent pain intensity, and recurrent pain duration) were measured using structured questionnaires, including the Luebeck pain screening questionnaire at age 13. Using hierarchical binary and multinomial logistic regression analyses with progressive adjustments for confounders, we estimated the associations [adjused odds ratios (aOR) with their 95% confidence intervals (95% CI)] between exposure to ACEs at 10 and pain features at 13 years. The study revealed a statistically significant association between exposure to ACEs reported at age 10 and any pain experienced at age 13 (OR = 1.09; 95% CI [1.07, 1.12]). Even after accounting for the newly reported ACEs at age 13, the association with ACEs at age 10, remained significant (aOR = 1.11 [95% CI, 1.08-1.14]). Consistent patterns were observed when the number of pain sites, recurrent pain intensity, or recurrent pain duration were used as outcome variables instead of any pain at age 13. Adverse childhood experiences occurring during the first decade of life predict the onset of pain features during early adolescence. Consequently, childhood exposure to adversity should be considered a pivotal initial exposure in a pathway leading to chronic pain later in life.
Collapse
Affiliation(s)
- Armine Abrahamyan
- EPIUnit - Instituto de Saúde Pública da Universidade do Porto, Porto, Portugal
- ITR- Laboratório para a Investigação Integrativa e Translacional, Porto, Portugal
| | - Raquel Lucas
- EPIUnit - Instituto de Saúde Pública da Universidade do Porto, Porto, Portugal
- ITR- Laboratório para a Investigação Integrativa e Translacional, Porto, Portugal
| | - Milton Severo
- EPIUnit - Instituto de Saúde Pública da Universidade do Porto, Porto, Portugal
- ITR- Laboratório para a Investigação Integrativa e Translacional, Porto, Portugal
| | - Makram Talih
- EPIUnit - Instituto de Saúde Pública da Universidade do Porto, Porto, Portugal
- ITR- Laboratório para a Investigação Integrativa e Translacional, Porto, Portugal
| | - Sílvia Fraga
- EPIUnit - Instituto de Saúde Pública da Universidade do Porto, Porto, Portugal
- ITR- Laboratório para a Investigação Integrativa e Translacional, Porto, Portugal
| |
Collapse
|
20
|
Taff CC, McNew SM, Campagna L, Vitousek MN. Corticosterone exposure is associated with long-term changes in DNA methylation, physiology and breeding decisions in a wild bird. Mol Ecol 2024; 33:e17456. [PMID: 38953311 DOI: 10.1111/mec.17456] [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: 07/31/2023] [Revised: 06/07/2024] [Accepted: 06/24/2024] [Indexed: 07/04/2024]
Abstract
When facing challenges, vertebrates activate a hormonal stress response that can dramatically alter behaviour and physiology. Although this response can be costly, conceptual models suggest that it can also recalibrate the stress response system, priming more effective responses to future challenges. Little is known about whether this process occurs in wild animals, particularly in adulthood, and if so, how information about prior experience with stressors is encoded. One potential mechanism is hormonally mediated changes in DNA methylation. We simulated the spikes in corticosterone that accompany a stress response using non-invasive dosing in tree swallows (Tachycineta bicolor) and monitored the phenotypic effects 1 year later. In a subset of individuals, we characterized DNA methylation using reduced representation bisulfite sequencing shortly after treatment and a year later. The year after treatment, experimental females had stronger negative feedback and initiated breeding earlier-traits that are associated with stress resilience and reproductive performance in our population-and higher baseline corticosterone. We also found that natural variation in corticosterone predicted patterns of DNA methylation. Finally, corticosterone treatment influenced methylation on short (1-2 weeks) and long (1 year) time scales; however, these changes did not have clear links to functional regulation of the stress response. Taken together, our results are consistent with corticosterone-induced priming of future stress resilience and support DNA methylation as a potential mechanism, but more work is needed to demonstrate functional consequences. Uncovering the mechanisms linking experience with the response to future challenges has implications for understanding the drivers of stress resilience.
Collapse
Affiliation(s)
- Conor C Taff
- Department of Ecology & Evolutionary Biology and Cornell Lab of Ornithology, Cornell University, Ithaca, New York, USA
- Department of Biology, Colby College, Waterville, Maine, USA
| | - Sabrina M McNew
- Department of Ecology & Evolutionary Biology, University of Arizona, Tucson, Arizona, USA
| | - Leonardo Campagna
- Department of Ecology & Evolutionary Biology and Cornell Lab of Ornithology, Cornell University, Ithaca, New York, USA
| | - Maren N Vitousek
- Department of Ecology & Evolutionary Biology and Cornell Lab of Ornithology, Cornell University, Ithaca, New York, USA
| |
Collapse
|
21
|
Hiura LC, Lazaro VA, Ophir AG. Paternal absence and increased caregiving independently and interactively shape the development of male prairie voles at subadult and adult life stages. Horm Behav 2024; 164:105605. [PMID: 39032207 PMCID: PMC11330720 DOI: 10.1016/j.yhbeh.2024.105605] [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: 04/19/2024] [Revised: 07/11/2024] [Accepted: 07/12/2024] [Indexed: 07/22/2024]
Abstract
The influence of maternal caregiving is a powerful force on offspring development. The absence of a father during early life in biparental species also has profound implications for offspring development, although it is far less studied than maternal influences. Moreover, we have limited understanding of the interactive forces that maternal and paternal caregiving impart on offspring. We investigated if behaviorally upregulating maternal care compensates for paternal absence on prairie vole (Microtus ochrogaster) pup development. We used an established handling manipulation to increase levels of caregiving in father-absent and biparental families, and later measured male offspring behavioral outcomes at sub-adulthood and adulthood. Male offspring raised without fathers were more prosocial (or possibly less socially anxious) than those raised biparentally. Defensive behavior and responses to contextual novelty were also influenced by the absence of fathers, but only in adulthood. Offensive aggression and movement in the open field test changed as a function of life-stage but not parental exposure. Notably, adult pair bonding was not impacted by our manipulations. Boosting parental care produced males that moved more in the open field test. Parental handling also increased oxytocin immunoreactive cells within the supraoptic nucleus of the hypothalamus (SON), and in the paraventricular nucleus (PVN) of biparentally-reared males. We found no differences in vasopressinergic cell groups. We conclude that male prairie voles are contextually sensitive to the absence of fathers and caregiving intensity. Our study highlights the importance of considering the ways early experiences synergistically shape offspring behavioral and neural phenotypes across the lifespan.
Collapse
Affiliation(s)
- Lisa C Hiura
- Department of Psychology, Cornell University, Ithaca, NY 14853, USA
| | - Vanessa A Lazaro
- Department of Psychology, Cornell University, Ithaca, NY 14853, USA
| | - Alexander G Ophir
- Department of Psychology, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
22
|
Sanguino-Gómez J, Krugers HJ. Early-life stress impairs acquisition and retrieval of fear memories: sex-effects, corticosterone modulation, and partial prevention by targeting glucocorticoid receptors at adolescent age. Neurobiol Stress 2024; 31:100636. [PMID: 38883213 PMCID: PMC11177066 DOI: 10.1016/j.ynstr.2024.100636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 03/11/2024] [Accepted: 04/20/2024] [Indexed: 06/18/2024] Open
Abstract
The early postnatal period is a sensitive time window that is characterized by several neurodevelopmental processes that define neuronal architecture and function later in life. Here, we examined in young adult mice, using an auditory fear conditioning paradigm, whether stress during the early postnatal period 1) impacts fear acquisition and memory consolidation in male and female mice; 2) alters the fear responsiveness to corticosterone and 3) whether effects of early-life stress (ELS) can be prevented by treating mice with a glucocorticoid (GR) antagonist at adolescence. Male and female mice were exposed to a limited nesting and bedding model of ELS from postnatal day (PND) 2-9 and injected i.p with RU38486 (RU486) at adolescent age (PND 28-30). At two months of age, mice were trained in the fear conditioning (FC) paradigm (with and without post training administration of corticosterone - CORT) and freezing behavior during fear acquisition and contextual and auditory memory retrieval was scored. We observed that ELS impaired fear acquisition specifically in male mice and reduced both contextual and auditory memory retrieval in male and female mice. Acute post-training administration of CORT increased freezing levels during auditory memory retrieval in female mice but reduced freezing levels during the tone presentation in particular in control males. Treatment with RU486 prevented ELS-effects in acquisition in male mice and in females during auditory memory retrieval. In conclusion, this study highlights the long-lasting consequences of early-life stress on fear memory processing and further illustrates 1) the potential of a glucocorticoid antagonist intervention during adolescence to mitigate these effects and 2) the partial modulation of the auditory retrieval upon post training administration of CORT, with all these effects being sex-dependent.
Collapse
Affiliation(s)
| | - Harm J Krugers
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
23
|
Manav N, Jit BP, Kataria B, Sharma A. Cellular and epigenetic perspective of protein stability and its implications in the biological system. Epigenomics 2024; 16:879-900. [PMID: 38884355 PMCID: PMC11370918 DOI: 10.1080/17501911.2024.2351788] [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: 11/29/2023] [Accepted: 04/30/2024] [Indexed: 06/18/2024] Open
Abstract
Protein stability is a fundamental prerequisite in both experimental and therapeutic applications. Current advancements in high throughput experimental techniques and functional ontology approaches have elucidated that impairment in the structure and stability of proteins is intricately associated with the cause and cure of several diseases. Therefore, it is paramount to deeply understand the physical and molecular confounding factors governing the stability of proteins. In this review article, we comprehensively investigated the evolution of protein stability, examining its emergence over time, its relationship with organizational aspects and the experimental methods used to understand it. Furthermore, we have also emphasized the role of Epigenetics and its interplay with post-translational modifications (PTMs) in regulating the stability of proteins.
Collapse
Affiliation(s)
- Nisha Manav
- Department of Biochemistry, All India Institute of Medical Sciences New Delhi, Ansari Nagar, 110029, India
| | - Bimal Prasad Jit
- Department of Biochemistry, All India Institute of Medical Sciences New Delhi, Ansari Nagar, 110029, India
| | - Babita Kataria
- Department of Medical Oncology, National Cancer Institute, All India Institute of Medical Sciences, Jhajjar, 124105, India
| | - Ashok Sharma
- Department of Biochemistry, All India Institute of Medical Sciences New Delhi, Ansari Nagar, 110029, India
- Department of Biochemistry, National Cancer Institute, All India Institute of Medical Sciences, Jhajjar, 124105, India
| |
Collapse
|
24
|
Parks BJ, Salazar P, Morrison L, McGraw MK, Gunnell M, Tobacyk J, Brents LK, Berquist MD. Limited bedding and nesting increases ethanol drinking in female rats. Pharmacol Biochem Behav 2024; 239:173756. [PMID: 38555037 PMCID: PMC11088506 DOI: 10.1016/j.pbb.2024.173756] [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: 12/20/2023] [Revised: 02/29/2024] [Accepted: 03/25/2024] [Indexed: 04/02/2024]
Abstract
Prenatal opioid exposure (POE) and postnatal adverse experiences are early life adversities (ELA) that often co-occur and increase problematic alcohol (EtOH) drinking during adolescence. We investigated the relationship between POE, postnatal adversity, and adolescent EtOH drinking in rats. We also sought to determine whether ELAs affect alpha-adrenoceptor density in the brain because the noradrenergic system is involved in problematic alcohol drinking and its treatment. We hypothesized that the combination of POE and postnatal adversity will increase alcohol drinking in rats compared to rats with exposure to either adversity alone or to control. We also predicted that POE and postnatal adversity would increase α1-adrenoceptor density and decrease α2-adrenoceptor density in brain to confer a stress-responsive phenotype. Pregnant rats received morphine (15 mg/kg/day) or saline via subcutaneous minipumps from gestational day 9 until birth. Limited bedding and nesting (LBN) procedures were introduced from postnatal day (PD) 3-11 to mimic early life adversity-scarcity. Offspring rats (PD 31-33) were given opportunities to drink EtOH (20 %, v/v) using intermittent-access, two-bottle choice (with water) procedures. Rats given access to EtOH were assigned into sub-groups that were injected with either yohimbine (1 mg/kg, ip) or vehicle (2 % DMSO, ip) 30 min prior to each EtOH access session to determine the effects of α2-adrenoceptor inhibition on alcohol drinking. We harvested cortices, brainstems, and hypothalami from EtOH-naïve littermates on either PD 30 or PD 70 and conducted radioligand receptor binding assays to quantify α1- and α2-adrenoceptor densities. Contrary to our hypothesis, only LBN alone increased EtOH intake in female adolescent rats compared to female rats with POE. Neither POE nor LBN affected α1- or α2-adrenoceptor densities in the cortex, brainstem, or hypothalamus of early- or late-aged adolescent rats. These results suggest a complex interaction between ELA type and sex on alcohol drinking.
Collapse
Affiliation(s)
- B J Parks
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Mail Slot 611, Little Rock, AR 72205, United States of America
| | - P Salazar
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Mail Slot 611, Little Rock, AR 72205, United States of America
| | - L Morrison
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Mail Slot 611, Little Rock, AR 72205, United States of America
| | - M K McGraw
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Mail Slot 611, Little Rock, AR 72205, United States of America
| | - M Gunnell
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Mail Slot 611, Little Rock, AR 72205, United States of America
| | - J Tobacyk
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Mail Slot 611, Little Rock, AR 72205, United States of America
| | - L K Brents
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Mail Slot 611, Little Rock, AR 72205, United States of America
| | - M D Berquist
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Mail Slot 611, Little Rock, AR 72205, United States of America.
| |
Collapse
|
25
|
Lucas ME, Hemsworth LM, Butler KL, Morrison RS, Tilbrook AJ, Marchant JN, Rault JL, Galea RY, Hemsworth PH. Early human contact and housing for pigs - part 3: ability to cope with the environment. Animal 2024; 18:101166. [PMID: 38772077 DOI: 10.1016/j.animal.2024.101166] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 05/23/2024] Open
Abstract
Early experiences can have long-term impacts on stress adaptability. This paper is the last of three in a series on early experiences and stress in pigs, and reports on the effects of early human contact and housing on the ability of pigs to cope with their general environment. Using a 2 × 2 factorial design, 48 litters of pigs were reared in either a farrowing crate (FC) or a loose farrowing pen (LP; PigSAFE pen) which was larger, more physically complex and allowed the sow to move freely. Piglets were provided with either routine contact from stockpeople (C), or routine contact plus regular opportunities for positive human contact (+HC) involving 5 min of scratching, patting and stroking imposed to the litter 5 days/week from 0 to 4 weeks of age. At 4 weeks of age (preweaning), C piglets that were reared in FC had considerably lower concentrations of serum brain-derived neurotrophic factor (BDNF) than piglets from the other treatment combinations. Compared to C pigs, +HC pigs had fewer injuries at 4 weeks of age. There were no clear effects of human contact on BDNF concentrations or injuries after weaning, or on basal cortisol or immunoglobulin-A concentrations, behavioural time budgets, tear staining, growth, or piglet survival. Compared to FC piglets, LP piglets showed more play behaviour and interactions with the dam and less repetitive nosing towards pen mates during lactation. There was no evidence that early housing affected pigs' behavioural time budgets or physiology after weaning. Tear staining severity was greater in LP piglets at 4 weeks of age, but this may have been associated with the higher growth rates of LP piglets preweaning. There was no effect of lactation housing on growth after weaning. Preweaning piglet mortality was higher in the loose system. The findings on BDNF concentrations, injuries and play behaviour suggest improved welfare during the treatment period in +HC and LP piglets compared to C and FC piglets, respectively. These results together with those from the other papers in this series indicate that positive human interaction early in life promotes stress adaptability in pigs. Furthermore, while the farrowing crate environment deprives piglets of opportunities for play behaviour and sow interaction, there was no evidence that rearing in crates negatively affected pig welfare or stress resilience after weaning. Whether these findings are specific to the two housing systems studied here, or can be generalised to other housing designs, warrants further research.
Collapse
Affiliation(s)
- M E Lucas
- The Animal Welfare Science Centre, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia.
| | - L M Hemsworth
- The Animal Welfare Science Centre, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - K L Butler
- The Animal Welfare Science Centre, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - R S Morrison
- Rivalea Australia Pty Ltd, Corowa, Victoria 2464, Australia
| | - A J Tilbrook
- Centre for Animal Science, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St Lucia 4072, Australia; School of Veterinary Science, The University of Queensland, Gatton Campus, Gatton, Queensland 4343, Australia
| | - J N Marchant
- Organic Plus Trust, Alexandria, VA 22302, USA; A World of Good Initiative Inc., Dover, DE 19901, USA
| | - J-L Rault
- Institute of Animal Welfare Science, University of Veterinary Medicine, Vienna A-1210, Austria
| | - R Y Galea
- The Animal Welfare Science Centre, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - P H Hemsworth
- The Animal Welfare Science Centre, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
26
|
Baud O, Knoop M. [Oxytocin as a neuroprotective strategy in neonates: concept and preclinical evidence]. GYNECOLOGIE, OBSTETRIQUE, FERTILITE & SENOLOGIE 2024; 52:418-424. [PMID: 38145743 DOI: 10.1016/j.gofs.2023.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 12/20/2023] [Indexed: 12/27/2023]
Abstract
OBJECTIVE Prematurity and intra-uterine growth retardation are responsible for brain damage associated with various neurocognitive and behavioral disorders in more than 9 million children each year. Most pharmacological strategies aimed at preventing perinatal brain injury have not demonstrated substantial clinical benefits so far. In contrast, enrichment of the newborn's environment appears to have positive effects on brain structure and function, influences newborn hormonal responses, and has lasting neurobehavioral consequences during infancy and adulthood. Oxytocin (OT), a neuropeptide released by the hypothalamus, may represent the hormonal basis for these long-term effects. METHOD This review of the literature summarizes the knowledge concerning the effect of OT in the newborn and the preclinical data supporting its neuroprotective effect. RESULTS OT plays a role during the perinatal period, in parent-child attachment and in social behavior. Furthermore, preclinical studies strongly suggest that endogenous and synthetic OT is capable of regulating the inflammatory response of the central nervous system in response to situations of prematurity or more generally insults to the developing brain. The long-term effect of synthetic OT administration during labor is also discussed. CONCLUSION All the conceptual and experimental data converge to indicate that OT would be a promising candidate for neonatal neuroprotection, in particular through the regulation of neuroinflammation.
Collapse
Affiliation(s)
- Olivier Baud
- Laboratoire du développement, Université de Genève, Genève, Suisse; Inserm U1141, Université Paris Cité, Paris, France; Service de Soins Intensifs Pédiatriques et Néonatologie, Hôpitaux Universitaires de Genève, Genève, Suisse.
| | - Marit Knoop
- Laboratoire du développement, Université de Genève, Genève, Suisse
| |
Collapse
|
27
|
Takahashi A. Toward understanding the neural mechanisms involved in early life stress-induced aggression: A Highlight for "Maternal separation early in life induces excessive activity of the central amygdala related to abnormal aggression". J Neurochem 2024; 168:957-960. [PMID: 38413201 DOI: 10.1111/jnc.16050] [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: 12/24/2023] [Revised: 12/25/2023] [Accepted: 01/01/2024] [Indexed: 02/29/2024]
Abstract
Early life stress, such as childhood abuse and neglect, is one of the major risk factors for the development of antisocial behavior. In rat models, repeated maternal separation (MS) stress, in which the pups are separated from the dams for a few hours each day during the first 2-3 weeks of life, increases aggressive behavior in adult males. This Editorial highlights an article in the current issue of the Journal of Neurochemistry that demonstrates the involvement of the central nucleus of the amygdala (CeA) in the escalation of aggressive behavior in the MS model. The authors show that MS rats exhibit higher c-Fos expression in the CeA during an aggressive encounter compared to non-isolated control rats. Unexpectedly, other amygdala subnuclei did not show differential activation between MS and control groups. Using optogenetics, they provide direct evidence that activation of CeA neurons increases intermale aggressive behavior and that bilateral CeA activation shifts behavioral patterns toward more qualitatively intense aggressive behavior than unilateral CeA activation. These findings highlight the important role of the CeA in the development of abnormal aggression and indicate that this region may be an important therapeutic target for human aggression induced by early life stress.
Collapse
Affiliation(s)
- Aki Takahashi
- Laboratory of Behavioral Neurobiology, Institute of Human Sciences, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
28
|
Hou W, Ma H, Huang C, Li Y, Li L, Zhang L, Qu Y, Xun Y, Yang Q, He Z, Tai F. Effects of paternal deprivation on empathetic behavior and the involvement of oxytocin receptors in the anterior cingulate cortex. Horm Behav 2024; 162:105536. [PMID: 38522143 DOI: 10.1016/j.yhbeh.2024.105536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 03/04/2024] [Accepted: 03/18/2024] [Indexed: 03/26/2024]
Abstract
Paternal deprivation (PD) impairs social cognition and sociality and increases levels of anxiety-like behavior. However, whether PD affects the levels of empathy in offspring and its underlying mechanisms remain unknown. The present study found that PD increased anxiety-like behavior in mandarin voles (Microtus mandarinus), impaired sociality, reduced the ability of emotional contagion, and the level of consolation behavior. Meanwhile, PD reduced OT neurons in the paraventricular nucleus (PVN) in both male and female mandarin voles. PD decreased the level of OT receptor (OTR) mRNA in the anterior cingulate cortex (ACC) of male and female mandarin voles. Besides, OTR overexpression in the ACC reversed the PD-induced changes in anxiety-like behavior, social preference, emotional contagion, and consolation behavior. Interference of OTR expression in the ACC increased levels of anxiety-like behaviors, while it reduced levels of sociality, emotional contagion, and consolation. These results revealed that the OTR in the ACC is involved in the effects of PD on empathetic behaviors, and provide mechanistic insight into how social experiences affect empathetic behaviors.
Collapse
Affiliation(s)
- Wenjuan Hou
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an 710062, China; School of Environmental and Material Engineering, Yantai University, 264005, China
| | - Huan Ma
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an 710062, China
| | - Caihong Huang
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an 710062, China
| | - Yin Li
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an 710062, China
| | - Lu Li
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an 710062, China
| | - Lizi Zhang
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an 710062, China
| | - Yishan Qu
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an 710062, China
| | - Yufeng Xun
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an 710062, China
| | - Qixuan Yang
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an 710062, China
| | - Zhixiong He
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an 710062, China.
| | - Fadao Tai
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an 710062, China.
| |
Collapse
|
29
|
Kuroda KO, Fukumitsu K, Kurachi T, Ohmura N, Shiraishi Y, Yoshihara C. Parental brain through time: The origin and development of the neural circuit of mammalian parenting. Ann N Y Acad Sci 2024; 1534:24-44. [PMID: 38426943 DOI: 10.1111/nyas.15111] [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] [Indexed: 03/02/2024]
Abstract
This review consolidates current knowledge on mammalian parental care, focusing on its neural mechanisms, evolutionary origins, and derivatives. Neurobiological studies have identified specific neurons in the medial preoptic area as crucial for parental care. Unexpectedly, these neurons are characterized by the expression of molecules signaling satiety, such as calcitonin receptor and BRS3, and overlap with neurons involved in the reproductive behaviors of males but not females. A synthesis of comparative ecology and paleontology suggests an evolutionary scenario for mammalian parental care, possibly stemming from male-biased guarding of offspring in basal vertebrates. The terrestrial transition of tetrapods led to prolonged egg retention in females and the emergence of amniotes, skewing care toward females. The nocturnal adaptation of Mesozoic mammalian ancestors reinforced maternal care for lactation and thermal regulation via endothermy, potentially introducing metabolic gate control in parenting neurons. The established maternal care may have served as the precursor for paternal and cooperative care in mammals and also fostered the development of group living, which may have further contributed to the emergence of empathy and altruism. These evolution-informed working hypotheses require empirical validation, yet they offer promising avenues to investigate the neural underpinnings of mammalian social behaviors.
Collapse
Affiliation(s)
- Kumi O Kuroda
- RIKEN Center for Brain Science, Saitama, Japan
- School of Life Sciences and Technologies, Tokyo Institute of Technology, Kanagawa, Japan
| | - Kansai Fukumitsu
- RIKEN Center for Brain Science, Saitama, Japan
- Department of Physiology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Takuma Kurachi
- RIKEN Center for Brain Science, Saitama, Japan
- Department of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Nami Ohmura
- RIKEN Center for Brain Science, Saitama, Japan
- Center for Brain, Mind and Kansei Sciences Research, Hiroshima University, Hiroshima, Japan
| | - Yuko Shiraishi
- RIKEN Center for Brain Science, Saitama, Japan
- Kawamura Gakuen Woman's University, Chiba, Japan
| | - Chihiro Yoshihara
- RIKEN Center for Brain Science, Saitama, Japan
- School of Life Sciences and Technologies, Tokyo Institute of Technology, Kanagawa, Japan
| |
Collapse
|
30
|
Tabbaa M, Levitt P. Chd8 haploinsufficiency impacts rearing experience in C57BL/6 mice. GENES, BRAIN, AND BEHAVIOR 2024; 23:e12892. [PMID: 38560770 PMCID: PMC10982810 DOI: 10.1111/gbb.12892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/14/2024] [Accepted: 03/05/2024] [Indexed: 04/04/2024]
Abstract
Mutations in CHD8 are one of the highest genetic risk factors for autism spectrum disorder. Studies in mice that investigate underlying mechanisms have shown Chd8 haploinsufficient mice display some trait disruptions that mimic clinical phenotypes, although inconsistencies have been reported in some traits across different models on the same strain background. One source of variation across studies may be the impact of Chd8 haploinsufficiency on maternal-offspring interactions. While differences in maternal care as a function of Chd8 genotype have not been studied directly, a previous study showed that pup survival was reduced when reared by Chd8 heterozygous dams compared with wild-type (WT) dams, suggesting altered maternal care as a function of Chd8 genotype. Through systematic observation of the C57BL/6 strain, we first determined the impact of Chd8 haploinsufficiency in the offspring on WT maternal care frequencies across preweaning development. We next determined the impact of maternal Chd8 haploinsufficiency on pup care. Compared with litters with all WT offspring, WT dams exhibited less frequent maternal behaviors toward litters consisting of offspring with mixed Chd8 genotypes, particularly during postnatal week 1. Dam Chd8 haploinsufficiency decreased litter survival and increased active maternal care also during postnatal week 1. Determining the impact of Chd8 haploinsufficiency on early life experiences provides an important foundation for interpreting offspring outcomes and determining mechanisms that underlie heterogeneous phenotypes.
Collapse
Affiliation(s)
- Manal Tabbaa
- Children's Hospital Los AngelesThe Saban Research InstituteLos AngelesCaliforniaUSA
- Keck School of Medicine of the University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Pat Levitt
- Children's Hospital Los AngelesThe Saban Research InstituteLos AngelesCaliforniaUSA
- Keck School of Medicine of the University of Southern CaliforniaLos AngelesCaliforniaUSA
| |
Collapse
|
31
|
Burenkova OV, Grigorenko EL. The role of epigenetic mechanisms in the long-term effects of early-life adversity and mother-infant relationship on physiology and behavior of offspring in laboratory rats and mice. Dev Psychobiol 2024; 66:e22479. [PMID: 38470450 PMCID: PMC10959231 DOI: 10.1002/dev.22479] [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: 09/15/2023] [Revised: 01/23/2024] [Accepted: 02/16/2024] [Indexed: 03/13/2024]
Abstract
Maternal care during the early postnatal period of altricial mammals is a key factor in the survival and adaptation of offspring to environmental conditions. Natural variations in maternal care and experimental manipulations with maternal-child relationships modeling early-life adversity (ELA) in laboratory rats and mice have a strong long-term influence on the physiology and behavior of offspring in rats and mice. This literature review is devoted to the latest research on the role of epigenetic mechanisms in these effects of ELA and mother-infant relationship, with a focus on the regulation of hypothalamic-pituitary-adrenal axis and brain-derived neurotrophic factor. An important part of this review is dedicated to pharmacological interventions and epigenetic editing as tools for studying the causal role of epigenetic mechanisms in the development of physiological and behavioral profiles. A special section of the manuscript will discuss the translational potential of the discussed research.
Collapse
Affiliation(s)
- Olga V. Burenkova
- Department of Psychology, University of Houston, Houston, Texas, USA
- Texas Institute for Measurement, Evaluation, and Statistics, University of Houston, Houston, Texas, USA
- Department of Integrative Biology, University of Guelph, Guelph, Ontario, Canada
| | - Elena L. Grigorenko
- Department of Psychology, University of Houston, Houston, Texas, USA
- Texas Institute for Measurement, Evaluation, and Statistics, University of Houston, Houston, Texas, USA
- Center for Cognitive Sciences, Sirius University of Science and Technology, Sochi, Russia
- Departments of Molecular and Human Genetics and Pediatrics, Baylor College of Medicine, Houston, Texas, USA
- Child Study Center, Yale University, New Haven, Connecticut, USA
- Research Administration, Moscow State University for Psychology and Education, Moscow, Russia
| |
Collapse
|
32
|
Bhattacharya A, Chakraborty M, Chanda A, Alqahtani T, Kumer A, Dhara B, Chattopadhyay M. Neuroendocrine and cellular mechanisms in stress resilience: From hormonal influence in the CNS to mitochondrial dysfunction and oxidative stress. J Cell Mol Med 2024; 28:e18220. [PMID: 38509751 PMCID: PMC10955164 DOI: 10.1111/jcmm.18220] [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: 12/17/2023] [Revised: 02/12/2024] [Accepted: 02/22/2024] [Indexed: 03/22/2024] Open
Abstract
Recent advancements in neuroendocrinology challenge the long-held belief that hormonal effects are confined to perivascular tissues and do not extend to the central nervous system (CNS). This paradigm shift, propelled by groundbreaking research, reveals that synthetic hormones, notably in anti-inflammatory medications, significantly influence steroid psychosis, behavioural, and cognitive impairments, as well as neuropeptide functions. A seminal development in this field occurred in 1968 with McEven's proposal that rodent brains are responsive to glucocorticoids, fundamentally altering the understanding of how anxiety impacts CNS functionality and leading to the identification of glucocorticosteroids and mineralocorticoids as distinct corticotropic receptors. This paper focuses on the intricate roles of the neuroendocrine, immunological, and CNS in fostering stress resilience, underscored by recent animal model studies. These studies highlight active, compensatory, and passive strategies for resilience, supporting the concept that anxiety and depression are systemic disorders involving dysregulation across both peripheral and central systems. Resilience is conceptualized as a multifaceted process that enhances psychological adaptability to stress through adaptive mechanisms within the immunological system, brain, hypothalamo-pituitary-adrenal axis, and ANS Axis. Furthermore, the paper explores oxidative stress, particularly its origin from the production of reactive oxygen species (ROS) in mitochondria. The mitochondria's role extends beyond ATP production, encompassing lipid, heme, purine, and steroidogenesis synthesis. ROS-induced damage to biomolecules can lead to significant mitochondrial dysfunction and cell apoptosis, emphasizing the critical nature of mitochondrial health in overall cellular function and stress resilience. This comprehensive synthesis of neuroendocrinological and cellular biological research offers new insights into the systemic complexity of stress-related disorders and the imperative for multidisciplinary approaches in their study and treatment.
Collapse
Affiliation(s)
- Arghya Bhattacharya
- Department of PharmacologyCalcutta Institute of Pharmaceutical Technology and AHSUluberiaWest BengalIndia
| | - Manas Chakraborty
- Department of Pharmaceutical BiotechnologyCalcutta institute of pharmaceutical technology and AHSUluberiaWest BengalIndia
| | - Ananya Chanda
- Department of Pharmaceutical ScienceAdamas UniversityBarasatWest BengalIndia
| | - Taha Alqahtani
- Department of Pharmacology, College of PharmacyKing Khalid UniversityAbhaSaudi Arabia
| | - Ajoy Kumer
- Department of ChemistryCollege of Arts and Sciences, IUBAT‐International University of Business Agriculture and TechnologyDhakaBangladesh
| | - Bikram Dhara
- Center for Global Health ResearchSaveetha Medical College and Hospital, Saveetha Institute of Medical and Technical SciencesChennaiIndia
- Department of Health SciencesNovel Global Community and Educational FoundationHebershamNew South WalesAustralia
| | - Moitreyee Chattopadhyay
- Department of Pharmaceutical TechnologyMaulana Abul Kalam Azad University of TechnologyKolkataWest BengalIndia
| |
Collapse
|
33
|
da Cunha Nones DC, Novais CO, Rojas VCT, de Paula Franco P, da Silva Estevam E, Silva MS, Giusti-Paiva A, Dos Anjos-Garcia T, Vilela FC. Litter reduction-induced obesity promotes early depressive-like behavior and elevated prefrontal cortex GFAP expression in male offspring. Behav Brain Res 2024; 461:114839. [PMID: 38154508 DOI: 10.1016/j.bbr.2023.114839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/22/2023] [Accepted: 12/23/2023] [Indexed: 12/30/2023]
Abstract
AIMS The present study was developed to investigate how litter reduction-induced obesity promotes early depressive-related behaviors in rodent offspring. MAIN METHODS We employed a standardized litter size reduction protocol, dividing litters into groups: normal litters (NL), consisting of six males and six females pups and small litters (SL), comprising two males and two females pups. Maternal behavior was monitored during the initial week of lactation. Subsequently, we assessed the pups for weight gain, locomotor activity, social play behavior, and performance in forced swimming test. We further evaluated the weights of retroperitoneal and perigonadal fat tissues, along with the expression of glial fibrillary acidic pprotein (GFAP) in the hippocampus and prefrontal cortex of the offspring. KEY FINDINGS Our results indicated that litter size reduction led to an increased the maternal behavior. In contrast, offspring from the SL group displayed greater weight gain and increased, retroperitoneal and perigonadal fat. Both male and female rodents in the SL group exhibited decreased social play behavior, and male offspring spent more time immobile during the forced swimming test, suggesting a depressive-like phenotype. Notably, we observed an increase in the GFAP expression in the prefrontal cortex of male rodents, with a trend toward increased expression in the hippocampus. SIGNIFICANCE Obesity may facilitate the development of early depressive-like behaviors, potentially associated with elevated GFAP expression in the prefrontal cortex.
Collapse
Affiliation(s)
- Débora Cristina da Cunha Nones
- Programa de Pós-Graduação em Biociências Aplicadas à Saúde (PPGB), Universidade Federal de Alfenas (Unifal-MG), Alfenas, Minas Gerais, Brazil; Departamento de Ciências Fisiológicas, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas (Unifal-MG), Alfenas, Minas Gerais, Brazil
| | - Cíntia Onofra Novais
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas (PPGMCF), Universidade Federal de Alfenas (Unifal-MG), Alfenas, Minas Gerais, Brazil; Departamento de Ciências Fisiológicas, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas (Unifal-MG), Alfenas, Minas Gerais, Brazil
| | - Viviana Carolina Trujillo Rojas
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas (PPGMCF), Universidade Federal de Alfenas (Unifal-MG), Alfenas, Minas Gerais, Brazil; Departamento de Ciências Fisiológicas, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas (Unifal-MG), Alfenas, Minas Gerais, Brazil
| | - Priscila de Paula Franco
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas (PPGMCF), Universidade Federal de Alfenas (Unifal-MG), Alfenas, Minas Gerais, Brazil; Departamento de Ciências Fisiológicas, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas (Unifal-MG), Alfenas, Minas Gerais, Brazil
| | - Elisa da Silva Estevam
- Departamento de Ciências Fisiológicas, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas (Unifal-MG), Alfenas, Minas Gerais, Brazil
| | - Mariana Santos Silva
- Departamento de Ciências Fisiológicas, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas (Unifal-MG), Alfenas, Minas Gerais, Brazil
| | - Alexandre Giusti-Paiva
- Departamento de Ciências Fisiológicas, Centro de Ciências Biológicas da Universidade Federal de Santa Catarina (UFSC), Florianópolis, Santa Catarina, Brazil
| | - Tayllon Dos Anjos-Garcia
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas (PPGMCF), Universidade Federal de Alfenas (Unifal-MG), Alfenas, Minas Gerais, Brazil; Departamento de Ciências Fisiológicas, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas (Unifal-MG), Alfenas, Minas Gerais, Brazil.
| | - Fabiana Cardoso Vilela
- Programa de Pós-Graduação em Biociências Aplicadas à Saúde (PPGB), Universidade Federal de Alfenas (Unifal-MG), Alfenas, Minas Gerais, Brazil; Centro de Inovação e Ensaios Pré-Clínicos (CIEnP), Florianópolis, Santa Catarina, Brazil
| |
Collapse
|
34
|
Abstract
In recent years, the impact of prenatal sound on development, notably for programming individual phenotypes for postnatal conditions, has increasingly been revealed. However, the mechanisms through which sound affects physiology and development remain mostly unexplored. Here, I gather evidence from neurobiology, developmental biology, cellular biology and bioacoustics to identify the most plausible modes of action of sound on developing embryos. First, revealing often-unsuspected plasticity, I discuss how prenatal sound may shape auditory system development and determine individuals' later capacity to receive acoustic information. I also consider the impact of hormones, including thyroid hormones, glucocorticoids and androgen, on auditory plasticity. Second, I review what is known about sound transduction to other - non-auditory - brain regions, and its potential to input on classical developmental programming pathways. Namely, the auditory pathway has direct anatomical and functional connectivity to the hippocampus, amygdala and/or hypothalamus, in mammals, birds and anurans. Sound can thus trigger both immediate and delayed responses in these limbic regions, which are specific to the acoustic stimulus and its biological relevance. Third, beyond the brain, I briefly consider the possibility for sound to directly affect cellular functioning, based on evidence in earless organisms (e.g. plants) and cell cultures. Together, the multi-disciplinary evidence gathered here shows that the brain is wired to allow multiple physiological and developmental effects of sound. Overall, there are many unexplored, but possible, pathways for sound to impact even primitive or immature organisms. Throughout, I identify the most promising research avenues for unravelling the processes of acoustic developmental programming.
Collapse
Affiliation(s)
- Mylene M Mariette
- Doñana Biological Station EBD-CSIC, 41092 Seville, Spain
- School of Life and Environmental Sciences, Deakin University, Geelong, VIC 3216, Australia
| |
Collapse
|
35
|
Siller Wilks SJ, Heidinger BJ, Westneat DF, Solomon J, Rubenstein DR. The impact of parental and developmental stress on DNA methylation in the avian hypothalamic-pituitary-adrenal axis. Mol Ecol 2024; 33:e17291. [PMID: 38343177 DOI: 10.1111/mec.17291] [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: 09/30/2023] [Revised: 12/29/2023] [Accepted: 01/23/2024] [Indexed: 03/07/2024]
Abstract
The hypothalamic-pituitary-adrenal (HPA) axis coordinates an organism's response to environmental stress. The responsiveness and sensitivity of an offspring's stress response may be shaped not only by stressors encountered in their early post-natal environment but also by stressors in their parent's environment. Yet, few studies have considered how stressors encountered in both of these early life environments may function together to impact the developing HPA axis. Here, we manipulated stressors in the parental and post-natal environments in a population of house sparrows (Passer domesticus) to assess their impact on changes in DNA methylation (and corresponding gene expression) in a suite of genes within the HPA axis. We found that nestlings that experienced early life stress across both life-history periods had higher DNA methylation in a critical HPA axis gene, the glucocorticoid receptor (NR3C1). In addition, we found that the life-history stage when stress was encountered impacted some genes (HSD11B1, NR3C1 and NR3C2) differently. We also found evidence for the mitigation of parental stress by post-natal stress (in HSD11B1 and NR3C2). Finally, by assessing DNA methylation in both the brain and blood, we were able to evaluate cross-tissue patterns. While some differentially methylated regions were tissue-specific, we found cross-tissue changes in NR3C2 and NR3C1, suggesting that blood is a suitable tissue for assessing DNA methylation as a biomarker of early life stress. Our results provide a crucial first step in understanding the mechanisms by which early life stress in different life-history periods contributes to changes in the epigenome of the HPA axis.
Collapse
Affiliation(s)
- Stefanie J Siller Wilks
- Department of Ecology Evolution and Environmental Biology, Columbia University, New York, New York, USA
| | - Britt J Heidinger
- Biological Sciences Department, North Dakota State University, Fargo, North Dakota, USA
| | - David F Westneat
- Department of Biology, University of Kentucky, Lexington, Kentucky, USA
| | - Joseph Solomon
- Department of Ecology Evolution and Environmental Biology, Columbia University, New York, New York, USA
| | - Dustin R Rubenstein
- Department of Ecology Evolution and Environmental Biology, Columbia University, New York, New York, USA
| |
Collapse
|
36
|
Rice RC, Gil DV, Baratta AM, Frawley RR, Hill SY, Farris SP, Homanics GE. Inter- and transgenerational heritability of preconception chronic stress or alcohol exposure: Translational outcomes in brain and behavior. Neurobiol Stress 2024; 29:100603. [PMID: 38234394 PMCID: PMC10792982 DOI: 10.1016/j.ynstr.2023.100603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 01/19/2024] Open
Abstract
Chronic stress and alcohol (ethanol) use are highly interrelated and can change an individual's behavior through molecular adaptations that do not change the DNA sequence, but instead change gene expression. A recent wealth of research has found that these nongenomic changes can be transmitted across generations, which could partially account for the "missing heritability" observed in genome-wide association studies of alcohol use disorder and other stress-related neuropsychiatric disorders. In this review, we summarize the molecular and behavioral outcomes of nongenomic inheritance of chronic stress and ethanol exposure and the germline mechanisms that could give rise to this heritability. In doing so, we outline the need for further research to: (1) Investigate individual germline mechanisms of paternal, maternal, and biparental nongenomic chronic stress- and ethanol-related inheritance; (2) Synthesize and dissect cross-generational chronic stress and ethanol exposure; (3) Determine cross-generational molecular outcomes of preconception ethanol exposure that contribute to alcohol-related disease risk, using cancer as an example. A detailed understanding of the cross-generational nongenomic effects of stress and/or ethanol will yield novel insight into the impact of ancestral perturbations on disease risk across generations and uncover actionable targets to improve human health.
Collapse
Affiliation(s)
- Rachel C. Rice
- Center for Neuroscience at the University of Pittsburgh, Pittsburgh, PA, USA
| | - Daniela V. Gil
- Center for Neuroscience at the University of Pittsburgh, Pittsburgh, PA, USA
| | - Annalisa M. Baratta
- Center for Neuroscience at the University of Pittsburgh, Pittsburgh, PA, USA
| | - Remy R. Frawley
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Shirley Y. Hill
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sean P. Farris
- Center for Neuroscience at the University of Pittsburgh, Pittsburgh, PA, USA
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Gregg E. Homanics
- Center for Neuroscience at the University of Pittsburgh, Pittsburgh, PA, USA
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
37
|
Xia X, Chen K, Chen Y. Change in function and homeostasis of HPA axis: The role of vitamin family. Chem Biol Interact 2024; 391:110899. [PMID: 38325521 DOI: 10.1016/j.cbi.2024.110899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 01/24/2024] [Accepted: 02/04/2024] [Indexed: 02/09/2024]
Abstract
With the improvement of living quality, people pay more and more attention to vitamin supplements. The vitamins in the daily diet can meet the needs of the body. Whether additional vitamin supplementation is necessary still needs to be further explored. Many studies have reported that vitamin deficiency and excessive vitamin supplementation could lead to abnormal development in the body or increase the risk of diseases. Here, we summarize the abnormal levels of vitamins can cause the homeostasis imbalance of hypothalamus-pituitary-adrenal (HPA) axis by affecting its development and function. It can lead to abnormal synthesis and secretion of glucocorticoid in the body, which mediates the occurrence and development of metabolic diseases and psychoneurotic diseases. In addition, vitamin has a strong antioxidant effect, which can eliminate oxygen free radicals. Thereby, vitamins can alter HPA axis function and homeostasis maintenance by combating oxidative stress. This review provides a theoretical basis for clarifying the role of abnormal levels of vitamin in the occurrence and development of multiple diseases and its intervention strategy, and also provides reference value and guiding significance for rational use of vitamins.
Collapse
Affiliation(s)
- Xuan Xia
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Kaiqi Chen
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yawen Chen
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
38
|
Siddi C, Cosentino S, Tamburini E, Concas L, Pisano MB, Ardu R, Deplano M, Follesa P, Maciocco E, Porcu P, Serra M, Pisu MG. Parental Social Isolation during Adolescence Alters Gut Microbiome in Rat Male Offspring. Biomolecules 2024; 14:172. [PMID: 38397408 PMCID: PMC10886888 DOI: 10.3390/biom14020172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/23/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
Previous work from our laboratory demonstrated that parental stress, induced by social isolation starting at puberty, leads to behavioral, endocrine, and biochemical changes in the male, but not female, offspring (ISO-O) of Sprague-Dawley rats. Here, we report alterations in the gut microbiota composition of ISO-O vs. grouped-housed offspring (GH-O), although all animals received the same diet and were housed in the same conditions. Analysis of bacterial communities by next-generation sequencing (NGS) of 16S rRNA gene revealed alterations at family and order levels within the main phyla of Bacteroides, Proteobacteria, and Firmicutes, including an almost total deficit in Limosilactobacillus reuteri (formerly Lactobacillus reuteri) and a significant increase in Ligilactobacillus murinus (formerly Lactobacillus murinus). In addition, we found an increase in the relative abundance of Rhodospirillales and Clostridiales in the families of Lachnospiraceae and Ruminococcaceae, and Bacteroidales in the family of Prevotellaceae. Furthermore, we examined plasma levels of the proinflammatory cytokines interleukin-1-beta and tumor necrosis factor-alpha, which did not differ between the two groups, while corticosterone concentrations were significantly increased in ISO-O rats. Our findings suggest that adverse environmental conditions experienced by parents may have an impact on the likelihood of disease development in the subsequent generations.
Collapse
Affiliation(s)
- Carlotta Siddi
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato, 09042 Cagliari, Italy; (C.S.); (L.C.); (P.F.)
| | - Sofia Cosentino
- Department of Medical Sciences and Public Health, University of Cagliari, Monserrato, 09042 Cagliari, Italy; (S.C.); (M.B.P.); (M.D.)
| | - Elena Tamburini
- Department of Biomedical Sciences, University of Cagliari, Monserrato, 09042 Cagliari, Italy; (E.T.); (R.A.)
| | - Luca Concas
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato, 09042 Cagliari, Italy; (C.S.); (L.C.); (P.F.)
| | - Maria Barbara Pisano
- Department of Medical Sciences and Public Health, University of Cagliari, Monserrato, 09042 Cagliari, Italy; (S.C.); (M.B.P.); (M.D.)
| | - Riccardo Ardu
- Department of Biomedical Sciences, University of Cagliari, Monserrato, 09042 Cagliari, Italy; (E.T.); (R.A.)
| | - Maura Deplano
- Department of Medical Sciences and Public Health, University of Cagliari, Monserrato, 09042 Cagliari, Italy; (S.C.); (M.B.P.); (M.D.)
| | - Paolo Follesa
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato, 09042 Cagliari, Italy; (C.S.); (L.C.); (P.F.)
| | - Elisabetta Maciocco
- Neuroscience Institute, National Research Council of Italy (CNR), Monserrato, 09042 Cagliari, Italy; (E.M.); (P.P.); (M.G.P.)
| | - Patrizia Porcu
- Neuroscience Institute, National Research Council of Italy (CNR), Monserrato, 09042 Cagliari, Italy; (E.M.); (P.P.); (M.G.P.)
| | - Mariangela Serra
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato, 09042 Cagliari, Italy; (C.S.); (L.C.); (P.F.)
| | - Maria Giuseppina Pisu
- Neuroscience Institute, National Research Council of Italy (CNR), Monserrato, 09042 Cagliari, Italy; (E.M.); (P.P.); (M.G.P.)
| |
Collapse
|
39
|
Nicolaides NC, Kanaka-Gantenbein C, Pervanidou P. Developmental Neuroendocrinology of Early-Life Stress: Impact on Child Development and Behavior. Curr Neuropharmacol 2024; 22:461-474. [PMID: 37563814 PMCID: PMC10845081 DOI: 10.2174/1570159x21666230810162344] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 02/22/2023] [Accepted: 03/15/2023] [Indexed: 08/12/2023] Open
Abstract
Our internal balance, or homeostasis, is threatened or perceived as threatened by stressful stimuli, the stressors. The stress system is a highly conserved system that adjusts homeostasis to the resting state. Through the concurrent activation of the hypothalamic-pituitary-adrenal axis and the locus coeruleus/norepinephrine-autonomic nervous systems, the stress system provides the appropriate physical and behavioral responses, collectively termed as "stress response", to restore homeostasis. If the stress response is prolonged, excessive or even inadequate, several acute or chronic stress-related pathologic conditions may develop in childhood, adolescence and adult life. On the other hand, earlylife exposure to stressors has been recognized as a major contributing factor underlying the pathogenesis of non-communicable disorders, including neurodevelopmental disorders. Accumulating evidence suggests that early-life stress has been associated with an increased risk for attention deficit hyperactivity disorder and autism spectrum disorder in the offspring, although findings are still controversial. Nevertheless, at the molecular level, early-life stressors alter the chemical structure of cytosines located in the regulatory regions of genes, mostly through the addition of methyl groups. These epigenetic modifications result in the suppression of gene expression without changing the DNA sequence. In addition to DNA methylation, several lines of evidence support the role of non-coding RNAs in the evolving field of epigenetics. In this review article, we present the anatomical and functional components of the stress system, discuss the proper, in terms of quality and quantity, stress response, and provide an update on the impact of early-life stress on child development and behavior.
Collapse
Affiliation(s)
- Nicolas C. Nicolaides
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens, School of Medicine, ‘Aghia Sophia’ Children's Hospital, Athens, 11527, Greece
- Division of Endocrinology and Metabolism, Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, 11527, Greece
- School of Medicine, University Research Institute of Maternal and Child Health and Precision Medicine, National and Kapodistrian University of Athens, Athens, Greece
- Department of Molecular Genetics, Function and Therapy, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Christina Kanaka-Gantenbein
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens, School of Medicine, ‘Aghia Sophia’ Children's Hospital, Athens, 11527, Greece
| | - Panagiota Pervanidou
- Unit of Developmental and Behavioral Pediatrics, First Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, “Aghia Sophia” Children's Hospital, Athens, Greece
| |
Collapse
|
40
|
Maayan L, Maayan M. Inflammatory mediation of the relationship between early adversity and major depressive disorder: A systematic review. J Psychiatr Res 2024; 169:364-377. [PMID: 38154266 DOI: 10.1016/j.jpsychires.2023.11.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 08/30/2023] [Accepted: 11/15/2023] [Indexed: 12/30/2023]
Abstract
Early adverse experience is related to psychiatric illness that occurs decades later. The mechanisms underlying this phenomenon have not been fully identified. There is a translational and clinical literature linking early adversity with Major Depressive Disorder (MDD) and inflammation. We reviewed articles that examine whether inflammation mediates this relationship. METHODS Literature review of PUB MED, CINAHL and APA Psycinfo articles that explicitly examine inflammation as a mediator between early adversity and depression using ((((((((((adversity) OR (trauma)) OR (maltreatment)) OR (child abuse)) AND (inflammation)) OR (inflammatory cytokines)) OR (crp)) OR (il-6)) OR (tnf)) AND (mediates)) AND (depression))))))))) as key words. RESULTS 2842 articles were initially identified. 1338 non-human studies were excluded and 512 more were filtered out as reviews. The remaining 992 titles and, when necessary, abstracts and manuscripts were reviewed and 956 were removed as being of other non-related phenomena. Four additional studies were added by hand searching the references of remaining studies. Out of these 40, 15 explicitly examined inflammation as a mediator of the relationship between early adversity and later depression. Approximately half (8/15) showed evidence that inflammation mediated the relationship between early adversity and depression. Sensitivity analyses showed that studies taking place in clinical populations, in youth and those that used the Adverse Childhood Events Scale to measure adversity, and IL-6 and TNF-α (as opposed to CRP) to measure inflammation were most likely to show mediation. CONCLUSIONS There is evidence to support the model of inflammation mediating the relationship between early adversity and depression. Certain measures in clinical populations appear more likely to support this model. Further study with more standardized, robust methods will help to answer this question more definitively and may elucidate a subtype of depression related to early adversity by alterations in immune function.
Collapse
Affiliation(s)
- Lawrence Maayan
- New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY, 10032, USA.
| | - Michal Maayan
- Skidmore College, 815 N. Broadway, Saratoga Springs, NY, 12866, USA
| |
Collapse
|
41
|
Lloyd KM, Gabard-Durnam L, Beaudry K, De Lisio M, Raine LB, Bernard-Willis Y, Watrous JNH, Whitfield-Gabrieli S, Kramer AF, Hillman CH. Cross-sectional analysis reveals COVID-19 pandemic community lockdown was linked to dysregulated cortisol and salivary alpha amylase in children. Front Public Health 2023; 11:1210122. [PMID: 38169630 PMCID: PMC10758420 DOI: 10.3389/fpubh.2023.1210122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 11/27/2023] [Indexed: 01/05/2024] Open
Abstract
The COVID-19 pandemic altered everyday life starting in March 2020. These alterations extended to the lives of children as their normal routines were disrupted by community lockdowns, online learning, limited in-person social contact, increased screen time, and reduced physical activity. Considerable research has investigated the physical health impact of COVID-19 infection, but far fewer studies have investigated the physiological impact of stressful pandemic-related changes to daily life, especially in children. The purpose of this study was to leverage an ongoing clinical trial to investigate physiological consequences associated with chronic stress of pandemic community lockdown on children. As a part of the clinical trial, children provided saliva samples. Saliva samples were analyzed for cortisol and salivary alpha amylase (sAA) content. This secondary cross-sectional analysis included 94 preadolescent children located within the Greater Boston, Massachusetts community. Children participated in the study either before, during, or following the pandemic community lockdown to form three groups for comparison. In response to chronic stress caused by the pandemic community lockdown, participants demonstrated dysregulation of fast-acting catecholamine response of the locus-coeruleus-norepinephrine system and slower-acting glucocorticoid response, resulting in an asymmetrical relationship of hypocortisolism (M = 0.78 ± 0.19 μg/mL, p < 0.001) paired with higher sAA (M = 12.73 ± 4.06 U/mL, p = 0.01). Results suggest that the abrupt COVID-19 disruption to daily life, including the stressful experience of community lockdown, had physiological effects on typically developing children. Further research is required to investigate mental health outcomes of children following the chronic stress of the pandemic community lockdown.
Collapse
Affiliation(s)
- Katherine M. Lloyd
- Department of Psychology, Northeastern University, Boston, MA, United States
| | | | - Kayleigh Beaudry
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Michael De Lisio
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Lauren B. Raine
- Department of Physical Therapy, Movement, and Rehabilitation Sciences, Northeastern University, Boston, MA, United States
| | - Ysabeau Bernard-Willis
- Department of Psychology, Northeastern University, Boston, MA, United States
- Division of Cognitive and Behavioral Neurology at Brigham and Women’s Hospital, Boston, MA, United States
| | | | | | - Arthur F. Kramer
- Department of Psychology, Northeastern University, Boston, MA, United States
- University of Illinois Beckman Institute, Champaign-Urbana, IL, United States
| | - Charles H. Hillman
- Department of Psychology, Northeastern University, Boston, MA, United States
- Department of Physical Therapy, Movement, and Rehabilitation Sciences, Northeastern University, Boston, MA, United States
| |
Collapse
|
42
|
Gundacker A, Glat M, Wais J, Stoehrmann P, Pollak A, Pollak DD. Early-life iron deficiency persistently disrupts affective behaviour in mice. Ann Med 2023; 55:1265-1277. [PMID: 37096819 PMCID: PMC10132221 DOI: 10.1080/07853890.2023.2191003] [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: 12/20/2022] [Accepted: 03/09/2023] [Indexed: 04/26/2023] Open
Abstract
BACKGROUND/OBJECTIVE Iron deficiency (ID) is the most common nutrient deficiency, affecting two billion people worldwide, including about 30% of pregnant women. During gestation, the brain is particularly vulnerable to environmental insults, which can irrevocably impair critical developmental processes. Consequently, detrimental consequences of early-life ID for offspring brain structure and function have been described. Although early life ID has been associated with an increased long-term risk for several neuropsychiatric disorders, the effect on depressive disorders has remained unresolved. MATERIALS AND METHODS A mouse model of moderate foetal and neonatal ID was established by keeping pregnant dams on an iron-deficient diet throughout gestation until postnatal day 10. The ensuing significant decrease of iron content in the offspring brain, as well as the impact on maternal behaviour and offspring vocalization was determined in the first postnatal week. The consequences of early-life ID for depression- and anxiety-like behaviour in adulthood were revealed employing dedicated behavioural assays. miRNA sequencing of hippocampal tissue of offspring revealed specific miRNAs signatures accompanying the behavioural deficits of foetal and neonatal ID in the adult brain. RESULTS Mothers receiving iron-deficient food during pregnancy and lactation exhibited significantly less licking and grooming behaviour, while active pup retrieval and pup ultrasonic vocalizations were unaltered. Adult offspring with a history of foetal and neonatal ID showed an increase in depression- and anxiety-like behaviour, paralleled by a deranged miRNA expression profile in the hippocampus, specifically levels of miR200a and miR200b. CONCLUSION ID during the foetal and neonatal periods has life-long consequences for affective behaviour in mice and leaves a specific and persistent mark on the expression of miRNAs in the brain. Foetal and neonatal ID needs to be further considered as risk factor for the development of depression and anxiety disorders later in life.Key MessagesMarginal reduction of gestational alimentary iron intake decreases brain iron content of the juvenile offspring.Early-life ID is associated with increased depression- and anxiety-like behaviour in adulthood.Reduction of maternal alimentary iron intake during pregnancy is reflected in an alteration of miRNA signatures in the adult offspring brain.
Collapse
Affiliation(s)
- Anna Gundacker
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Micaela Glat
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Jonathan Wais
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
- Department of Neurosurgery, Medical University of Vienna, Vienna, Austria
| | - Peter Stoehrmann
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Arnold Pollak
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Daniela D. Pollak
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
43
|
Abstract
Early life experiences can have an enduring impact on the brain and behavior, with implications for stress reactivity, cognition, and social behavior. In particular, the neural systems that contribute to the expression of social behavior are altered by early life social environments. However, paradigms that have been used to alter the social environment during development have typically focused on exposure to stress, adversity, and deprivation of species-typical social stimulation. Here, we explore whether complex social environments can shape the development of complex social behavior. We describe lab-based paradigms for studying early life social complexity in rodents that are generally focused on enriching the social and sensory experiences of the neonatal and juvenile periods of development. The impact of these experiences on social behavior and neuroplasticity is highlighted. Finally, we discuss the degree to which our current approaches for studying social behavior outcomes give insight into "complex" social behavior and how social complexity can be better integrated into lab-based methodologies.
Collapse
Affiliation(s)
- James P Curley
- Department of Psychology, The University of Texas at Austin, Austin, Texas, USA
| | - Frances A Champagne
- Department of Psychology, The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
44
|
Myers AM, Bowen SE, Brummelte S. Maternal care behavior and physiology moderate offspring outcomes following gestational exposure to opioids. Dev Psychobiol 2023; 65:e22433. [PMID: 38010303 DOI: 10.1002/dev.22433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 08/31/2023] [Accepted: 09/29/2023] [Indexed: 11/29/2023]
Abstract
The opioid epidemic has resulted in a drastic increase in gestational exposure to opioids. Opioid-dependent pregnant women are typically prescribed medications for opioid use disorders ("MOUD"; e.g., buprenorphine [BUP]) to mitigate the harmful effects of abused opioids. However, the consequences of exposure to synthetic opioids, particularly BUP, during gestation on fetal neurodevelopment and long-term outcomes are poorly understood. Further, despite the known adverse effects of opioids on maternal care, many preclinical and clinical studies investigating the effects of gestational opioid exposure on offspring outcomes fail to report on maternal care behaviors. Considering that offspring outcomes are heavily dependent upon the quality of maternal care, it is important to evaluate the effects of gestational opioid exposure in the context of the mother-infant dyad. This review compares offspring outcomes after prenatal opioid exposure and after reduced maternal care and integrates this information to potentially identify common underlying mechanisms. We explore whether adverse outcomes after gestational BUP exposure are due to direct effects of opioids in utero, deficits in maternal care, or a combination of both factors. Finally, suggestions for improving preclinical models of prenatal opioid exposure are provided to promote more translational studies that can help to improve clinical outcomes for opioid-dependent mothers.
Collapse
Affiliation(s)
- Abigail M Myers
- Department of Psychology, Wayne State University, Detroit, Michigan, USA
| | - Scott E Bowen
- Department of Psychology, Wayne State University, Detroit, Michigan, USA
- Translational Neuroscience Program, Wayne State University, Detroit, Michigan, USA
| | - Susanne Brummelte
- Department of Psychology, Wayne State University, Detroit, Michigan, USA
- Translational Neuroscience Program, Wayne State University, Detroit, Michigan, USA
| |
Collapse
|
45
|
Conde SV, Polotsky VY, Joseph V, Kinkead R. On the origins of sleep disordered breathing, cardiorespiratory and metabolic dysfunction: which came first, the chicken or the egg? J Physiol 2023; 601:5509-5525. [PMID: 36988138 PMCID: PMC10539476 DOI: 10.1113/jp284113] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 03/08/2023] [Indexed: 03/30/2023] Open
Abstract
Sleep disordered breathing (SDB) is a complex, sex specific and highly heterogeneous group of respiratory disorders. Nevertheless, sleep fragmentation and repeated fluctuations of arterial blood gases for several hours per night are at the core of the problem; together, they impose significant stress to the organism with deleterious consequences on physical and mental health. SDB increases the risk of obesity, diabetes, depression and anxiety disorders; however, the same health issues are risk factors for SDB. So, which came first, the chicken or the egg? What causes the appearance of the first significant apnoeic events during sleep? These are important questions because although moderate to severe SDB affects ∼500 million adults globally, we still have a poor understanding of the origins of the disease, and the main treatments (and animal models) focus on the symptoms rather than the cause. Because obesity, metabolic dysfunction and stress-related neurological disorders generally appear progressively, we discuss how the development of these diseases can lead to specific anatomical and non-anatomical traits of SDB in males and females while considering the impacts of sex steroids. In light of the growing evidence indicating that the carotid bodies are important sensors of key metabolic and endocrine signals associated with stress and dysmetabolism, we propose that these organs play a key role in the process.
Collapse
Affiliation(s)
- Silvia V. Conde
- NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Vsevolod Y Polotsky
- Department of Anesthesiology and Critical Care Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Vincent Joseph
- Département de Pédiatrie, Université Laval & Research Center of the Québec Heart and Lung Institute, Québec, QC. Canada
| | - Richard Kinkead
- Département de Pédiatrie, Université Laval & Research Center of the Québec Heart and Lung Institute, Québec, QC. Canada
| |
Collapse
|
46
|
Jimeno B, Gerritsma Y, Mulder E, Verhulst S. Glucocorticoid receptor expression in blood, but not across brain regions, reveals long-term effects of early life adversity in zebra finches. Physiol Behav 2023; 271:114310. [PMID: 37543106 DOI: 10.1016/j.physbeh.2023.114310] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 05/12/2023] [Accepted: 07/28/2023] [Indexed: 08/07/2023]
Abstract
Early-life environment can affect organisms for life on many levels. The glucocorticoid receptor (GR) gene has a pivotal role mediating organismal physiological and behavioral responses to environmental change, and is sensitive to early-life environmental conditions and epigenetic programming. Longitudinal studies require non-lethal sampling of peripheral tissues (e.g. blood), but this approach is dependent on the extent to which GR expression in peripheral tissues covaries with GR expression in central tissues. To test for the long-term effects of early life adversity on GR expression across brain and peripheral tissues, we manipulated developmental conditions of captive zebra finches (n = 45), rearing them in either benign or harsh conditions through manipulation of parental foraging costs. We measured relative GR mRNA expression in blood and five brain regions in adulthood: hippocampus, hypothalamus, amygdala, ventral striatum, and the nidopallium caudolaterale (analogous to the mammalian prefrontal cortex), using qPCR. We further tested whether GR expression was modulated by natal brood size (which affected growth), age at sampling, and sex. GR expression correlations among tissues varied widely in magnitude and direction, ranging from -0.27 to +0.80, indicating that our understanding of developmental effects on GR expression and associated phenotypes needs to be region specific rather than organism wide. A more consistent pattern was that GR expression increased with age in blood, ventral striatum and hippocampus; GR expression was independent of age in other tissues. Developmental treatment did not affect GR expression in any of the tissues measured directly, but in blood and ventral striatum of adult females we found a positive correlation between nestling mass and GR expression. Thus, GR expression in blood was affected by early life conditions as reflected in growth in adult females, a pattern also found in one brain tissue, but not ubiquitous across brain regions. These results point at sex-dependent physiological constraints during development, shaping early life effects on GR expression in females only. Further study is required to investigate whether these tissue-dependent effects more generally reflect tissue-dependent long-term effects of early life adversity. This, together with investigating the physiological consequences of GR expression levels on individual performance and coping abilities, will be fundamental towards understanding the mechanisms mediating long-term impacts of early life, and the extent to which these can be quantified through non-lethal sampling.
Collapse
Affiliation(s)
- Blanca Jimeno
- Instituto de Investigación en Recursos Cinegéticos (IREC), CSIC-UCLM-JCCM, Ronda de Toledo 12, 13005 Ciudad Real; Instituto Pirenaico de Ecologia (IPE), CSIC, Avda. Nuestra Señora de la Victoria, 16, Jaca, Spain.
| | - Yoran Gerritsma
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, the Netherlands
| | - Ellis Mulder
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, the Netherlands
| | - Simon Verhulst
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, the Netherlands
| |
Collapse
|
47
|
Jimeno B, Verhulst S. Meta-analysis reveals glucocorticoid levels reflect variation in metabolic rate, not 'stress'. eLife 2023; 12:RP88205. [PMID: 37889839 PMCID: PMC10611431 DOI: 10.7554/elife.88205] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2023] Open
Abstract
Glucocorticoid (GC) variation has long been thought to reflect variation in organismal 'stress,' but associations between GCs and Darwinian fitness components are diverse in magnitude, direction, and highly context-dependent. This paradox reveals our poor understanding of the causes of GC variation, contrasting with the detailed knowledge of the functional consequences of GC variation. Amongst an array of effects in many physiological systems, GCs orchestrate energy availability to anticipate and recover from predictable and unpredictable environmental fluctuations and challenges. Although this is mechanistically well-known, the extent to which GC levels are quantitatively explained by energy metabolism is unresolved. We investigated this association through meta-analysis, selecting studies of endotherms in which (1) an experiment was performed that affected metabolic rate and (2) metabolic rate and GC levels were measured simultaneously. We found that an increase in metabolic rate was associated with an increase in GC levels in 20 out of 21 studies (32 out of 35 effect sizes). More importantly, there was a strong positive correlation between the increases in metabolic rate and GCs (p=0.003). This pattern was similar in birds and mammals, and independent of the nature of the experimental treatment. We conclude that metabolic rate is a major driver of GC variation within individuals. Stressors often affect metabolic rate, leading us to question whether GC levels provide information on 'stress' beyond the stressor's effect on metabolic rate.
Collapse
Affiliation(s)
- Blanca Jimeno
- Instituto de Investigación en Recursos Cinegéticos (IREC), CSIC-UCLM-JCC, Ciudad Real, Spain
- Instituto Pirenaico de Ecologia (IPE), CSIC, Avda. Nuestra Señora de la Victoria, Jaca, Spain
| | | |
Collapse
|
48
|
Lapp HE, Salazar MG, Champagne FA. Automated maternal behavior during early life in rodents (AMBER) pipeline. Sci Rep 2023; 13:18277. [PMID: 37880307 PMCID: PMC10600172 DOI: 10.1038/s41598-023-45495-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 10/20/2023] [Indexed: 10/27/2023] Open
Abstract
Mother-infant interactions during the early postnatal period are critical for infant survival and the scaffolding of infant development. Rodent models are used extensively to understand how these early social experiences influence neurobiology across the lifespan. However, methods for measuring postnatal dam-pup interactions typically involve time-consuming manual scoring, vary widely between research groups, and produce low density data that limits downstream analytical applications. To address these methodological issues, we developed the Automated Maternal Behavior during Early life in Rodents (AMBER) pipeline for quantifying home-cage maternal and mother-pup interactions using open-source machine learning tools. DeepLabCut was used to track key points on rat dams (32 points) and individual pups (9 points per pup) in postnatal day 1-10 video recordings. Pose estimation models reached key point test errors of approximately 4.1-10 mm (14.39 pixels) and 3.44-7.87 mm (11.81 pixels) depending on depth of animal in the frame averaged across all key points for dam and pups respectively. Pose estimation data and human-annotated behavior labels from 38 videos were used with Simple Behavioral Analysis (SimBA) to generate behavior classifiers for dam active nursing, passive nursing, nest attendance, licking and grooming, self-directed grooming, eating, and drinking using random forest algorithms. All classifiers had excellent performance on test frames, with F1 scores above 0.886. Performance on hold-out videos remained high for nest attendance (F1 = 0.990), active nursing (F1 = 0.828), and licking and grooming (F1 = 0.766) but was lower for eating, drinking, and self-directed grooming (F1 = 0.534-0.554). A set of 242 videos was used with AMBER and produced behavior measures in the expected range from postnatal 1-10 home-cage videos. This pipeline is a major advancement in assessing home-cage dam-pup interactions in a way that reduces experimenter burden while increasing reproducibility, reliability, and detail of data for use in developmental studies without the need for special housing systems or proprietary software.
Collapse
Affiliation(s)
- Hannah E Lapp
- Department of Psychology, University of Texas at Austin, 108 E. Dean Keaton St, Austin, TX, 78712, USA.
| | - Melissa G Salazar
- Department of Psychology, University of Texas at Austin, 108 E. Dean Keaton St, Austin, TX, 78712, USA
| | - Frances A Champagne
- Department of Psychology, University of Texas at Austin, 108 E. Dean Keaton St, Austin, TX, 78712, USA
| |
Collapse
|
49
|
Alberry B, Silveira PP. Brain insulin signaling as a potential mediator of early life adversity effects on physical and mental health. Neurosci Biobehav Rev 2023; 153:105350. [PMID: 37544390 DOI: 10.1016/j.neubiorev.2023.105350] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 07/31/2023] [Accepted: 08/03/2023] [Indexed: 08/08/2023]
Abstract
In numerous brain structures, insulin signaling modulates the homeostatic processes, sensitivity to reward pathways, executive function, memory, and cognition. Through human studies and animal models, mounting evidence implicates central insulin signaling in the metabolic, physiological, and psychological consequences of early life adversity. In this review, we describe the consequences of early life adversity in the brain where insulin signaling is a key factor and how insulin may moderate the effects of adversity on psychiatric and cardio-metabolic health outcomes. Further understanding of how early life adversity and insulin signaling impact specific brain regions and mental and physical health outcomes will assist in prevention, diagnosis, and potential intervention following early life adversity.
Collapse
Affiliation(s)
- Bonnie Alberry
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Patricia Pelufo Silveira
- Department of Psychiatry, McGill University, Montreal, QC, Canada; Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada.
| |
Collapse
|
50
|
Yuan M, Yang B, Rothschild G, Mann JJ, Sanford LD, Tang X, Huang C, Wang C, Zhang W. Epigenetic regulation in major depression and other stress-related disorders: molecular mechanisms, clinical relevance and therapeutic potential. Signal Transduct Target Ther 2023; 8:309. [PMID: 37644009 PMCID: PMC10465587 DOI: 10.1038/s41392-023-01519-z] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 05/14/2023] [Accepted: 05/31/2023] [Indexed: 08/31/2023] Open
Abstract
Major depressive disorder (MDD) is a chronic, generally episodic and debilitating disease that affects an estimated 300 million people worldwide, but its pathogenesis is poorly understood. The heritability estimate of MDD is 30-40%, suggesting that genetics alone do not account for most of the risk of major depression. Another factor known to associate with MDD involves environmental stressors such as childhood adversity and recent life stress. Recent studies have emerged to show that the biological impact of environmental factors in MDD and other stress-related disorders is mediated by a variety of epigenetic modifications. These epigenetic modification alterations contribute to abnormal neuroendocrine responses, neuroplasticity impairment, neurotransmission and neuroglia dysfunction, which are involved in the pathophysiology of MDD. Furthermore, epigenetic marks have been associated with the diagnosis and treatment of MDD. The evaluation of epigenetic modifications holds promise for further understanding of the heterogeneous etiology and complex phenotypes of MDD, and may identify new therapeutic targets. Here, we review preclinical and clinical epigenetic findings, including DNA methylation, histone modification, noncoding RNA, RNA modification, and chromatin remodeling factor in MDD. In addition, we elaborate on the contribution of these epigenetic mechanisms to the pathological trait variability in depression and discuss how such mechanisms can be exploited for therapeutic purposes.
Collapse
Affiliation(s)
- Minlan Yuan
- Mental Health Center and Psychiatric Laboratory, the State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Biao Yang
- Department of Abdominal Oncology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Gerson Rothschild
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - J John Mann
- Department of Psychiatry, Columbia University, New York, NY, 10032, USA
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY, 10032, USA
- Department of Radiology, Columbia University, New York, NY, 10032, USA
| | - Larry D Sanford
- Sleep Research Laboratory, Center for Integrative Neuroscience and Inflammatory Diseases, Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Xiangdong Tang
- Sleep Medicine Center, Department of Respiratory and Critical Care Medicine, Mental Health Center, Translational Neuroscience Center, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Canhua Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chuang Wang
- Department of Pharmacology, and Provincial Key Laboratory of Pathophysiology in School of Medicine, Ningbo University, Ningbo, Zhejiang, 315211, China.
| | - Wei Zhang
- Mental Health Center and Psychiatric Laboratory, the State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Medical Big Data Center, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|