1
|
Bao M, Wu R, Li J, Tang R, Song C. Research summary, possible mechanisms and perspectives of gut microbiota changes causing precocious puberty. Front Nutr 2025; 12:1596654. [PMID: 40352262 PMCID: PMC12061974 DOI: 10.3389/fnut.2025.1596654] [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: 03/20/2025] [Accepted: 04/08/2025] [Indexed: 05/14/2025] Open
Abstract
The increasing global incidence of precocious puberty, linked to environmental, metabolic, and genetic factors, necessitates innovative therapies beyond gonadotropin-releasing hormone (GnRH) analogs. Accumulating evidence implicates gut microbiota dysbiosis as a pivotal regulator of pubertal timing via interactions with hormone metabolism (e.g., estrogen reactivation via β-glucuronidase), neuroendocrine pathways (nitric oxide signaling), and immune-inflammatory responses. This review delineates taxonomic alterations in central precocious puberty (CPP) and obesity-related subtypes, including Streptococcus enrichment and Alistipes depletion, alongside functional shifts in microbial metabolite production. Mechanistic insights highlight microbiota-driven modulation of the hypothalamic-pituitary-gonadal (HPG) axis, leptin/insulin dynamics, and epigenetic regulation. Emerging interventions-probiotics, fecal microbiota transplantation (FMT), and dietary modifications-demonstrate efficacy in preclinical models and early clinical studies for delaying puberty onset and restoring hormonal balance. Translational efforts to validate these strategies are critical for addressing the clinical and psychosocial challenges posed by precocious puberty, positioning gut microbiota modulation as a novel therapeutic frontier in pediatric endocrinology.
Collapse
Affiliation(s)
- Maorong Bao
- Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Rui Wu
- Ba’nan Hospital Affiliated to Chongqing Medical University, Chongqing, China
| | - Jingwei Li
- Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Runan Tang
- Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Cui Song
- Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
2
|
Ramadan YN, Alqifari SF, Alshehri K, Alhowiti A, Mirghani H, Alrasheed T, Aljohani F, Alghamdi A, Hetta HF. Microbiome Gut-Brain-Axis: Impact on Brain Development and Mental Health. Mol Neurobiol 2025:10.1007/s12035-025-04846-0. [PMID: 40234288 DOI: 10.1007/s12035-025-04846-0] [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: 07/12/2024] [Accepted: 03/12/2025] [Indexed: 04/17/2025]
Abstract
The current discovery that the gut microbiome, which contains roughly 100 trillion microbes, affects health and disease has catalyzed a boom in multidisciplinary research efforts focused on understanding this relationship. Also, it is commonly demonstrated that the gut and the CNS are closely related in a bidirectional pathway. A balanced gut microbiome is essential for regular brain activities and emotional responses. On the other hand, the CNS regulates the majority of GI physiology. Any disruption in this bidirectional pathway led to a progression of health problems in both directions, neurological and gastrointestinal diseases. In this review, we hope to shed light on the complicated connections of the microbiome-gut-brain axis and the critical roles of gut microbiome in the early development of the brain in order to get a deeper knowledge of microbiome-mediated pathological conditions and management options through rebalancing of gut microbiome.
Collapse
Affiliation(s)
- Yasmin N Ramadan
- Department of Microbiology and Immunology, Faculty of Pharmacy, Assiut University, Assiut, 71515, Egypt.
| | - Saleh F Alqifari
- Department of Pharmacy Practice, Faculty of Pharmacy, University of Tabuk, 71491, Tabuk, Saudi Arabia
| | - Khaled Alshehri
- Department of Internal Medicine (Neurology), Faculty of Medicine, University of Tabuk, Tabuk, Saudi Arabia
| | - Amirah Alhowiti
- Department of Family and Community Medicine, Faculty of Medicine, University of Tabuk, Tabuk, Saudi Arabia
| | - Hyder Mirghani
- Department of Internal Medicine, Faculty of Medicine, University of Tabuk, Tabuk, Saudi Arabia
| | - Tariq Alrasheed
- Department of Internal Medicine, Faculty of Medicine, University of Tabuk, Tabuk, Saudi Arabia
| | - Faisal Aljohani
- Division of Medicine and Gastroenterology, Department of Medicine, Faculty of Medicine, University of Tabuk, Tabuk, Saudi Arabia
| | - Abdulaziz Alghamdi
- Department of Medicine, Division of Psychiatry, Faculty of Medicine in Rabigh, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Helal F Hetta
- Division of Microbiology, Immunology and Biotechnology, Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, 71491, Tabuk, Saudi Arabia
| |
Collapse
|
3
|
Borzadaran FM, Rezakhani S, Kamali R, Esmaeilpour K. Maternal Separation Exhibits a Sex Dimorphism in Memory Impairments in Adolescent Rats: Acute Methylphenidate Administration as a Treatment. Birth Defects Res 2025; 117:e2441. [PMID: 39916605 PMCID: PMC11803433 DOI: 10.1002/bdr2.2441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 12/30/2024] [Accepted: 01/14/2025] [Indexed: 02/11/2025]
Abstract
INTRODUCTION Rodents are highly dependent on maternal care after birth. Disturbing mother and pup interactions leads to detrimental alternations for the rat and the mother. Maternal separation (MS) is an accepted model for investigating disruption of mother and pup relationship. In addition to other detrimental effects, MS is a model known to induce permanent changes in learning and memory. Methylphenidate has been effective in memory enhancement in individuals suffering from memory deficits, attention-deficit hyperactive disorder (ADHD), as well as healthy subjects for better performance in exams. MATERIAL AND METHODS In this research, a 21-day separation for 3 h was implemented, and the effects of MS on spatial and passive avoidance learning, and memory were evaluated in the mid-adolescence period of rats, in both males and females. Also, a drug intervention of a high therapeutic dose of 5 mg per kg was used in a five-day period in different control and MS groups. Morris water maze was utilized for spatial learning and memory analysis, and a shuttle box paradigm was used for passive avoidance learning and memory. RESULTS Through our behavioral tests, we have shown that MS can alter spatial learning and memory in males. On the other hand, females are protected from the detrimental effects of MS on spatial learning and memory. Furthermore, passive avoidance learning was not different among groups, be it male or female. However, in the case of memory evaluation in the passive avoidance test, the male did not exhibit a significant difference in step-through latency. However, maternally separated females had poor performance in the memory phase with shorter step-through latencies. CONCLUSION Methylphenidate compensated for the deleterious effects of MS on learning and spatial memory for the male group and passive avoidance memory in the female group at the behavioral level.
Collapse
Affiliation(s)
| | - Soheila Rezakhani
- Neuroscience Research Center, Institute of NeuropharmacologyKerman University of Medical SciencesKermanIran
| | | | - Khadijeh Esmaeilpour
- Neuroscience Research Center, Institute of NeuropharmacologyKerman University of Medical SciencesKermanIran
- School of Public Health SciencesUniversity of WaterlooWaterlooOntarioCanada
| |
Collapse
|
4
|
Esfand SM, Querdasi FR, Gancz NN, Savoca PW, Nussbaum S, Somers JA, Ditzer J, Figueroa MB, Chu K, Towner E, Callaghan BL. The mind, brain, and body study: A protocol for examining the effects of the gut-brain-immune axis on internalizing symptoms in youth exposed to caregiving-related early adversity. Brain Behav Immun Health 2024; 42:100880. [PMID: 39881818 PMCID: PMC11776082 DOI: 10.1016/j.bbih.2024.100880] [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: 08/30/2024] [Revised: 09/25/2024] [Accepted: 09/29/2024] [Indexed: 01/31/2025] Open
Abstract
Experiences of caregiving-related adversity are common and one of the strongest predictors of internalizing psychopathology (i.e., anxiety and depression). Specifically, individuals who have been exposed to such early adversities have altered affective neurodevelopment, impaired memory systems, increased risk of developing internalizing disorders, greater inflammation, and differences in gastrointestinal (gut) microbiome composition. Crucially, the gut microbiome undergoes a sensitive period of development that precedes neural and immune sensitive periods, thus making it a potentially fruitful target for intervention. Though previous work has assessed neural, immune, and gut microbiome systems in individuals exposed to early adversity, studies have primarily looked at these biological systems independently. The Mind, Brain, and Body study (MBB) implements multimodal and longitudinal design to assess how changes in the gut microbiome following caregiving-related adversity may underlie altered affective neurodevelopment, memory, and immune functioning in youth and contribute to internalizing symptoms. Across three waves, spread approximately 12-18 months apart, youth with and without previous experiences of caregiving-related adversity completed self-report measures of mental and physical health, provided stool, saliva, hair, and blood samples, and completed an MRI scan. Results of this study will expand our knowledge on how the gut microbiome shapes several biological and cognitive systems and motivate future work investigating the gut microbiome as potential target for intervention.
Collapse
Affiliation(s)
- Shiba M. Esfand
- Department of Psychology, University of California, Los Angeles, 502 Portola Plaza, Los Angeles, CA, 90095, USA
| | - Francesca R. Querdasi
- Department of Psychology, University of California, Los Angeles, 502 Portola Plaza, Los Angeles, CA, 90095, USA
| | - Naomi N. Gancz
- Department of Psychology, University of California, Los Angeles, 502 Portola Plaza, Los Angeles, CA, 90095, USA
| | - Paul W. Savoca
- Department of Psychology, University of California, Los Angeles, 502 Portola Plaza, Los Angeles, CA, 90095, USA
| | - Siyan Nussbaum
- Department of Psychology, University of California, Los Angeles, 502 Portola Plaza, Los Angeles, CA, 90095, USA
| | - Jennifer A. Somers
- Department of Psychology, University of California, Los Angeles, 502 Portola Plaza, Los Angeles, CA, 90095, USA
- Department of Psychological Sciences, Auburn University, 111 Thach Hall, Auburn, AL, 36849, USA
| | - Julia Ditzer
- Department of Psychology, Clinical Child and Adolescent Psychology, Technische Universität Dresden, Dresden, Germany
| | - Matthew B. Figueroa
- Department of Psychology, University of California, Los Angeles, 502 Portola Plaza, Los Angeles, CA, 90095, USA
| | - Kristen Chu
- Department of Psychology, University of California, Los Angeles, 502 Portola Plaza, Los Angeles, CA, 90095, USA
| | - Emily Towner
- Department of Psychology, University of California, Los Angeles, 502 Portola Plaza, Los Angeles, CA, 90095, USA
- Department of Psychology, University of Cambridge, Cambridge, United Kingdom
| | - Bridget L. Callaghan
- Department of Psychology, University of California, Los Angeles, 502 Portola Plaza, Los Angeles, CA, 90095, USA
| |
Collapse
|
5
|
Granata L, Fanikos M, Brenhouse HC. Early life adversity accelerates hypothalamic drive of pubertal timing in female rats with associated enhanced acoustic startle. Horm Behav 2024; 159:105478. [PMID: 38241961 PMCID: PMC10926229 DOI: 10.1016/j.yhbeh.2024.105478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 01/04/2024] [Accepted: 01/09/2024] [Indexed: 01/21/2024]
Abstract
Early life adversity in the form of childhood maltreatment in humans or as modeled by maternal separation (MS) in rodents is often associated with an earlier emergence of puberty in females. Earlier pubertal initiation is an example of accelerated biological aging and predicts later risk for anxiety in women, especially in populations exposed to early life trauma. Here we investigated external pubertal markers as well as hypothalamic gene expression of pubertal regulators kisspeptin and gonadotropin-releasing hormone, to determine a biological substrate for MS-induced accelerated puberty. We further investigated a mechanism by which developmental stress might regulate pubertal timing. As kisspeptin and gonadotropin-releasing hormone secretion are typically inhibited by corticotropin releasing hormone at its receptor CRH-R1, we hypothesized that MS induces a downregulation of Crhr1 gene transcription in a cell-specific manner. Finally, we explored the association between pubertal timing and anxiety-like behavior in an acoustic startle paradigm, to drive future preclinical research linking accelerated puberty and anxiety. We replicated previous findings that MS leads to earlier puberty in females but not males, and found expression of kisspeptin and gonadotropin-releasing hormone mRNA to be prematurely increased in MS females. RNAscope confirmed increased expression of these genes, and further revealed that kisspeptin-expressing neurons in females were less likely to express Crhr1 after MS. Early puberty was associated with higher acoustic startle magnitude in females. Taken together, these findings indicate precocial maturation of central pubertal timing mechanisms after MS, as well as a potential role of CRH-R1 in these effects and an association with a translational measure of anxiety.
Collapse
Affiliation(s)
- Lauren Granata
- Psychology Department, Northeastern University, Boston, MA, United States of America
| | - Michaela Fanikos
- Psychology Department, Northeastern University, Boston, MA, United States of America
| | - Heather C Brenhouse
- Psychology Department, Northeastern University, Boston, MA, United States of America.
| |
Collapse
|
6
|
Kigar SL, Cuarenta A, Zuniga CL, Chang L, Auger AP, Bakshi VP. Brain, behavior, and physiological changes associated with predator stress-An animal model for trauma exposure in adult and neonatal rats. Front Mol Neurosci 2024; 17:1322273. [PMID: 38486962 PMCID: PMC10938396 DOI: 10.3389/fnmol.2024.1322273] [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: 10/16/2023] [Accepted: 02/07/2024] [Indexed: 03/17/2024] Open
Abstract
The use of predators and predator odor as stressors is an important and ecologically relevant model for studying the impact of behavioral responses to threat. Here we summarize neural substrates and behavioral changes in rats resulting from predator exposure. We briefly define the impact predator exposure has on neural targets throughout development (neonatal, juvenile, and adulthood). These findings allow us to conceptualize the impact of predator exposure in the brain, which in turn may have broader implications for human disorders such as PTSD. Importantly, inclusion of sex as a biological variable yields distinct results that may indicate neural substrates impacted by predator exposure differ based on sex.
Collapse
Affiliation(s)
- Stacey L. Kigar
- Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Amelia Cuarenta
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| | - Carla L. Zuniga
- Department of Psychiatry, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Liza Chang
- College of Agricultural and Life Sciences Academic Affairs, University of Wisconsin-Madison, Madison, WI, United States
| | - Anthony P. Auger
- Department of Psychology, University of Wisconsin-Madison, Madison, WI, United States
| | - Vaishali P. Bakshi
- Department of Psychiatry, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| |
Collapse
|
7
|
Gunawan SP, Huang SY, Wang CC, Huynh LBP, Nguyen NN, Hsu SY, Chen YC. Sleep deprivation alters pubertal timing in humans and rats: the role of the gut microbiome. Sleep 2024; 47:zsad308. [PMID: 38065690 DOI: 10.1093/sleep/zsad308] [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: 07/24/2023] [Revised: 11/27/2023] [Indexed: 02/09/2024] Open
Abstract
STUDY OBJECTIVES Evidence implied that sleeping duration is associated with the timing of puberty and that sleep deprivation triggers early pubertal onset in adolescents. Sleep deprivation can affect metabolic changes and gut microbiota composition. This study investigated the effects of sleep deprivation on pubertal onset and gut microbiota composition in animal models and a human cohort. METHODS This study comprised 459 boys and 959 girls from the Taiwan Pubertal Longitudinal Study. Sleep duration was evaluated using the self-report Pittsburgh Sleep Quality Index questionnaire. Early sexual maturation was defined by pediatric endocrinologist assessments. Mediation analyses were done to examine the association between sleep parameters, obesity, and early sexual maturation. Besides, Sprague Dawley juvenile rats were exposed to 4 weeks of chronic sleep deprivation. Vaginal opening (VO) and preputial separation (PS) were observed every morning to determine pubertal onset in female and male rats. RESULTS The sleep-deprived juvenile rats in the sleep-deprived-female (SDF) and sleep-deprived-male (SDM) groups experienced delayed VO (mean VO days: 33 days in control; 35 days in SDF; p-value < 0.05) and PS (mean PS days: 42 days in control; 45 days in SDM; p-value < 0.05), respectively. Relative to their non-sleep-deprived counterparts, the sleep-deprived juvenile rats exhibited lower body weight and body fat percentage. Significant differences in relative bacterial abundance at genus levels and decreased fecal short-chain-fatty-acid levels were identified in both the SDF and SDM groups. In the human cohort, insufficient sleep increased the risk of early sexual maturation, particularly in girls (OR, 1.44; 95% CI: 1.09 to 1.89; p-value < 0.01). Insufficient sleep also indirectly affected early sexual maturation in girls, with obesity serving as the mediator. CONCLUSIONS Overall, sleep deprivation altered the timing of puberty in both animal and human models but in different directions. In the rat model, sleep deprivation delayed the pubertal onset in juvenile rats through gut dysbiosis and metabolic changes, leading to a low body weight and body fat percentage. In the human model, sleep deprivation led to fat accumulation, causing obesity in girls, which increased the risk of early puberty.
Collapse
Affiliation(s)
| | - Shih-Yi Huang
- Graduate Institute of Metabolism and Obesity Sciences Taipei Medical University, Taipei, Taiwan
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei, Taiwan
| | - Chun-Chi Wang
- Department of Family Medicine, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | - Linh Ba Phuong Huynh
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei, Taiwan
| | - Nam Nhat Nguyen
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shih-Yuan Hsu
- Department of Family Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yang-Ching Chen
- Graduate Institute of Metabolism and Obesity Sciences Taipei Medical University, Taipei, Taiwan
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei, Taiwan
- Department of Family Medicine, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
- Department of Family Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
8
|
Yue M, Zhang L. Exploring the Mechanistic Interplay between Gut Microbiota and Precocious Puberty: A Narrative Review. Microorganisms 2024; 12:323. [PMID: 38399733 PMCID: PMC10892899 DOI: 10.3390/microorganisms12020323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
The gut microbiota has been implicated in the context of sexual maturation during puberty, with discernible differences in its composition before and after this critical developmental stage. Notably, there has been a global rise in the prevalence of precocious puberty in recent years, particularly among girls, where approximately 90% of central precocious puberty cases lack a clearly identifiable cause. While a link between precocious puberty and the gut microbiota has been observed, the precise causality and underlying mechanisms remain elusive. This narrative review aims to systematically elucidate the potential mechanisms that underlie the intricate relationship between the gut microbiota and precocious puberty. Potential avenues of exploration include investigating the impact of the gut microbiota on endocrine function, particularly in the regulation of hormones, such as gonadotropin-releasing hormone (GnRH), luteinizing hormone (LH), and follicle-stimulating hormone (FSH). Additionally, this review will delve into the intricate interplay between the gut microbiome, metabolism, and obesity, considering the known association between obesity and precocious puberty. This review will also explore how the microbiome's involvement in nutrient metabolism could impact precocious puberty. Finally, attention is given to the microbiota's ability to produce neurotransmitters and neuroactive compounds, potentially influencing the central nervous system components involved in regulating puberty. By exploring these mechanisms, this narrative review seeks to identify unexplored targets and emerging directions in understanding the role of the gut microbiome in relation to precocious puberty. The ultimate goal is to provide valuable insights for the development of non-invasive diagnostic methods and innovative therapeutic strategies for precocious puberty in the future, such as specific probiotic therapy.
Collapse
Affiliation(s)
- Min Yue
- Microbiome-X, National Institute of Health Data Science of China & Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Lei Zhang
- Microbiome-X, National Institute of Health Data Science of China & Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| |
Collapse
|
9
|
Wall EK, Teo JN, Roth A, Chan ME, Brandt J, Hibri M, Richardson R, Baker KD. Effects of social buffering on fear extinction in adolescent rats. Behav Res Ther 2024; 173:104457. [PMID: 38134498 DOI: 10.1016/j.brat.2023.104457] [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: 05/11/2023] [Revised: 10/12/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023]
Abstract
Across social species, the presence of another individual can reduce stress reactions to adverse stimuli, a phenomenon known as social buffering. The present study investigated whether social buffering influences the expression and extinction of learned fear in adolescence, a developmental period of diminished fear inhibition and increased social interaction. Quality of maternal care and degree of social investigation were examined as factors that may influence social buffering. In adolescence, male rats were fear conditioned and then given extinction training either in the presence of a same-age rat or alone. Animals were then tested alone for extinction retention. In two experiments, the presence of a conspecific robustly reduced conditioned fear responses during extinction training. Interestingly, a persistent social buffering effect was observed when the extinction and conditioning contexts had prominent differences in features (Experiment 1), but not when these contexts were relatively similar (Experiment 2). Neither quality of maternal care nor degree of social investigation predicted the effects of social buffering. These findings suggest that social buffering robustly dampens fear responses during adolescence when a peer is present and this suppression can persist, in some instances, even when the peer is absent.
Collapse
Affiliation(s)
- Emily K Wall
- School of Psychology, UNSW Sydney, NSW, 2052, Australia
| | - Jia Ni Teo
- School of Psychology, UNSW Sydney, NSW, 2052, Australia
| | | | - Mei E Chan
- School of Psychology, UNSW Sydney, NSW, 2052, Australia
| | | | - Maya Hibri
- School of Psychology, UNSW Sydney, NSW, 2052, Australia
| | | | | |
Collapse
|
10
|
Dalla C, Jaric I, Pavlidi P, Hodes GE, Kokras N, Bespalov A, Kas MJ, Steckler T, Kabbaj M, Würbel H, Marrocco J, Tollkuhn J, Shansky R, Bangasser D, Becker JB, McCarthy M, Ferland-Beckham C. Practical solutions for including sex as a biological variable (SABV) in preclinical neuropsychopharmacological research. J Neurosci Methods 2024; 401:110003. [PMID: 37918446 PMCID: PMC10842858 DOI: 10.1016/j.jneumeth.2023.110003] [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: 08/29/2023] [Revised: 10/13/2023] [Accepted: 10/27/2023] [Indexed: 11/04/2023]
Abstract
Recently, many funding agencies have released guidelines on the importance of considering sex as a biological variable (SABV) as an experimental factor, aiming to address sex differences and avoid possible sex biases to enhance the reproducibility and translational relevance of preclinical research. In neuroscience and pharmacology, the female sex is often omitted from experimental designs, with researchers generalizing male-driven outcomes to both sexes, risking a biased or limited understanding of disease mechanisms and thus potentially ineffective therapeutics. Herein, we describe key methodological aspects that should be considered when sex is factored into in vitro and in vivo experiments and provide practical knowledge for researchers to incorporate SABV into preclinical research. Both age and sex significantly influence biological and behavioral processes due to critical changes at different timepoints of development for males and females and due to hormonal fluctuations across the rodent lifespan. We show that including both sexes does not require larger sample sizes, and even if sex is included as an independent variable in the study design, a moderate increase in sample size is sufficient. Moreover, the importance of tracking hormone levels in both sexes and the differentiation between sex differences and sex-related strategy in behaviors are explained. Finally, the lack of robust data on how biological sex influences the pharmacokinetic (PK), pharmacodynamic (PD), or toxicological effects of various preclinically administered drugs to animals due to the exclusion of female animals is discussed, and methodological strategies to enhance the rigor and translational relevance of preclinical research are proposed.
Collapse
Affiliation(s)
- Christina Dalla
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Greece.
| | - Ivana Jaric
- Animal Welfare Division, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Pavlina Pavlidi
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Greece
| | - Georgia E Hodes
- School of Neuroscience, Virginia Tech, Blacksburg, VA 24060, USA
| | - Nikolaos Kokras
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Greece; First Department of Psychiatry, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Greece
| | - Anton Bespalov
- Partnership for Assessment and Accreditation of Scientific Practice (PAASP GmbH), Heidelberg, Germany
| | - Martien J Kas
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, the Netherlands
| | | | - Mohamed Kabbaj
- Department of Biomedical Sciences & Neurosciences, College of Medicine, Florida State University, USA
| | - Hanno Würbel
- Animal Welfare Division, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Jordan Marrocco
- Department of Biology, Touro University, New York, NY 10027, USA
| | | | - Rebecca Shansky
- Department of Psychology, Northeastern University, Boston, MA 02128, USA
| | - Debra Bangasser
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA; Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA 30303, USA
| | - Jill B Becker
- Department of Psychology and Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Margaret McCarthy
- University of Maryland School of Medicine, Department of Pharmacology, Baltimore MD, USA
| | | |
Collapse
|
11
|
Power SD, Stewart E, Zielke LG, Byrne EP, Douglas A, Ortega-de San Luis C, Lynch L, Ryan TJ. Immune activation state modulates infant engram expression across development. SCIENCE ADVANCES 2023; 9:eadg9921. [PMID: 37939176 PMCID: PMC10631722 DOI: 10.1126/sciadv.adg9921] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 10/06/2023] [Indexed: 11/10/2023]
Abstract
Infantile amnesia is possibly the most ubiquitous form of memory loss in mammals. We investigated how memories are stored in the brain throughout development by integrating engram labeling technology with mouse models of infantile amnesia. Here, we found a phenomenon in which male offspring in maternal immune activation models of autism spectrum disorder do not experience infantile amnesia. Maternal immune activation altered engram ensemble size and dendritic spine plasticity. We rescued the same apparently forgotten infantile memories in neurotypical mice by optogenetically reactivating dentate gyrus engram cells labeled during complex experiences in infancy. Furthermore, we permanently reinstated lost infantile memories by artificially updating the memory engram, demonstrating that infantile amnesia is a reversible process. Our findings suggest not only that infantile amnesia is due to a reversible retrieval deficit in engram expression but also that immune activation during development modulates innate, and reversible, forgetting switches that determine whether infantile amnesia will occur.
Collapse
Affiliation(s)
- Sarah D. Power
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, Ireland
- Center for Lifespan Psychology, Max Planck Institute for Human Development, Berlin, Germany
| | - Erika Stewart
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Louisa G. Zielke
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, Ireland
- Faculty of Psychology and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Eric P. Byrne
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Aaron Douglas
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Clara Ortega-de San Luis
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, Ireland
| | - Lydia Lynch
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Tomás J. Ryan
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
- Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, Ireland
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Melbourne, VIC, Australia
- Child & Brain Development Program, Canadian Institute for Advanced Research (CIFAR), Toronto, ON, Canada
| |
Collapse
|
12
|
Yuan X, Shangguan H, Zhang Y, Lin X, Chen R. Intervention Effect of Probiotics on the Early Onset of Puberty Induced by Daidzein in Female Mice. Mol Nutr Food Res 2023; 67:e2200501. [PMID: 36528780 DOI: 10.1002/mnfr.202200501] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 11/28/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND The relationship between soy isoflavones (SI)-induced gut dysbiosis and puberty onset has not been explored, and the protective effect of probiotic is still controversial. This study investigates the action of daidzein (the main components of SI) and probiotic on the age of puberty onset in female mice. METHODS AND RESULTS Changes in the gut microbiota and production of short chain fatty acids (SCFAs) metabolism are highlighted to analyze a possible causative relationship to puberty onset in female c57/bl mice. The results demonstrate that daidzein promotes earlier onset of puberty, and can significantly alter the composition of the fecal bacterial community. Furthermore, daidzein alters the gut microbiota such that levels of butyrate, isovalerate, and hexanoate are reduced. Moreover, a probiotic treatment normalizes the timing of puberty onset, likely due to alteration in the gut microbiota to enhance SCFAs production. CONCLUSION These findings provide evidence that 95% daidzein has the potential to advance the timing of puberty onset in female mice, and the gut microbiome can be a therapeutic target to regulate the timing of puberty onset.
Collapse
Affiliation(s)
- Xin Yuan
- Department of Endocrinology, Genetics and Metabolism, Fuzhou Children's Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Huakun Shangguan
- Department of Endocrinology, Genetics and Metabolism, Fuzhou Children's Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Ying Zhang
- Department of Endocrinology, Genetics and Metabolism, Fuzhou Children's Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Xiangquan Lin
- Department of Endocrinology, Genetics and Metabolism, Fuzhou Children's Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Ruimin Chen
- Department of Endocrinology, Genetics and Metabolism, Fuzhou Children's Hospital of Fujian Medical University, Fuzhou, 350005, China
| |
Collapse
|
13
|
Points of divergence on a bumpy road: early development of brain and immune threat processing systems following postnatal adversity. Mol Psychiatry 2023; 28:269-283. [PMID: 35705633 DOI: 10.1038/s41380-022-01658-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 05/16/2022] [Accepted: 06/01/2022] [Indexed: 01/11/2023]
Abstract
Lifelong indices of maladaptive behavior or illness often stem from early physiological aberrations during periods of dynamic development. This is especially true when dysfunction is attributable to early life adversity (ELA), when the environment itself is unsuitable to support development of healthy behavior. Exposure to ELA is strongly associated with atypical sensitivity and responsivity to potential threats-a characteristic that could be adaptive in situations where early adversity prepares individuals for lifelong danger, but which often manifests in difficulties with emotion regulation and social relationships. By synthesizing findings from animal research, this review will consider threat sensitivity through the lenses of associated corticolimbic brain circuitry and immune mechanisms, both of which are immature early in life to maximize adaptation for protection against environmental challenges to an individual's well-being. The forces that drive differential development of corticolimbic circuits include caretaking stimuli, physiological and psychological stressors, and sex, which influences developmental trajectories. These same forces direct developmental processes of the immune system, which bidirectionally communicates with sensory systems and emotion regulation circuits within the brain. Inflammatory signals offer a further force influencing the timing and nature of corticolimbic plasticity, while also regulating sensitivity to future threats from the environment (i.e., injury or pathogens). The early development of these systems programs threat sensitivity through juvenility and adolescence, carving paths for probable function throughout adulthood. To strategize prevention or management of maladaptive threat sensitivity in ELA-exposed populations, it is necessary to fully understand these early points of divergence.
Collapse
|
14
|
Manohar K, Mesfin FM, Liu J, Shelley WC, Brokaw JP, Markel TA. Gut-Brain cross talk: The pathogenesis of neurodevelopmental impairment in necrotizing enterocolitis. Front Pediatr 2023; 11:1104682. [PMID: 36873645 PMCID: PMC9975605 DOI: 10.3389/fped.2023.1104682] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/23/2023] [Indexed: 02/17/2023] Open
Abstract
Necrotizing enterocolitis (NEC) is a devastating condition of multi-factorial origin that affects the intestine of premature infants and results in high morbidity and mortality. Infants that survive contend with several long-term sequelae including neurodevelopmental impairment (NDI)-which encompasses cognitive and psychosocial deficits as well as motor, vision, and hearing impairment. Alterations in the gut-brain axis (GBA) homeostasis have been implicated in the pathogenesis of NEC and the development of NDI. The crosstalk along the GBA suggests that microbial dysbiosis and subsequent bowel injury can initiate systemic inflammation which is followed by pathogenic signaling cascades with multiple pathways that ultimately lead to the brain. These signals reach the brain and activate an inflammatory cascade in the brain resulting in white matter injury, impaired myelination, delayed head growth, and eventual downstream NDI. The purpose of this review is to summarize the NDI seen in NEC, discuss what is known about the GBA, explore the relationship between the GBA and perinatal brain injury in the setting of NEC, and finally, highlight the existing research into possible therapies to help prevent these deleterious outcomes.
Collapse
Affiliation(s)
- Krishna Manohar
- Department of Surgery, Indiana University School of Medicine (IUSM), Indianapolis, IN, United States
| | - Fikir M Mesfin
- Department of Surgery, Indiana University School of Medicine (IUSM), Indianapolis, IN, United States
| | - Jianyun Liu
- Department of Surgery, Indiana University School of Medicine (IUSM), Indianapolis, IN, United States
| | - W Christopher Shelley
- Department of Surgery, Indiana University School of Medicine (IUSM), Indianapolis, IN, United States
| | - John P Brokaw
- Department of Surgery, Indiana University School of Medicine (IUSM), Indianapolis, IN, United States
| | - Troy A Markel
- Department of Surgery, Indiana University School of Medicine (IUSM), Indianapolis, IN, United States.,Riley Hospital for Children, Indiana University Health, Indianapolis, IN, United States
| |
Collapse
|
15
|
Tsai MC, Lee YL, Chen YC. Association of the consumption of common drinks with early puberty in both sexes. Front Public Health 2022; 10:854477. [PMID: 36536777 PMCID: PMC9758723 DOI: 10.3389/fpubh.2022.854477] [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: 01/14/2022] [Accepted: 11/15/2022] [Indexed: 12/05/2022] Open
Abstract
Background We examined the effect of sugar-sweetened beverages (SSB) and common drink intake on pubertal development in both sexes. Methods Data were retrieved from Taiwan Children Health Study, which involved detailed pubertal stage assessments of 2,819 schoolchildren aged 11 years in 2011-2012. Drawings of secondary sexual characteristics and self-reported age at menarche or voice breaking were used to assess pubertal stages. Dietary intake was assessed using a detailed semi-quantitative food frequency questionnaire. Generalized estimating equation modeling was applied to obtain odds ratios (ORs) and 95% confidence intervals (CIs) to represent the effects of each drink on early pubertal development outcomes. Results In boys, an one cup/day increment of a SSB was associated with earlier voice breaking (β = -0.12; 95% CI = -0.20, -0.04), whereas consuming yogurt (≥2 cups/day) was a protective factor against early puberty (OR = 0.78; 95% CI = 0.73, 0.83). In girls, SSB consumption was associated with increased risk of early puberty in a dose-response manner, and a similar protective effect of yogurt consumption and fermented probiotic drink (≥2 cups/day) against early puberty was observed (OR = 0.96; 95% CI = 0.94, 0.99). Furthermore, the intake of both total sugar and added sugar within SSBs increased risk of early puberty in girls but not in boys. Conclusions Sugar-sweetened beverages were associated with early puberty, and probiotic drinks appeared to mitigate this link. These findings indicate that the gut-brain axis could play a crucial role in sexual maturation.
Collapse
Affiliation(s)
- Meng-Che Tsai
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yungling Leo Lee
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan,College of Public Health, China Medical University, Taichung, Taiwan,Graduate Institute of Clinical Medicine, Taipei Medical University, Taipei, Taiwan,Yungling Leo Lee
| | - Yang Ching Chen
- Department of Family Medicine, Taipei Medical University Hospital, Taipei, Taiwan,Department of Family Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan,School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei, Taiwan,Graduate Institute of Metabolism and Obesity Sciences, Taipei Medical University, Taipei, Taiwan,*Correspondence: Yang Ching Chen
| |
Collapse
|
16
|
Freimer D, Yang TT, Ho TC, Tymofiyeva O, Leung C. The gut microbiota, HPA axis, and brain in adolescent-onset depression: Probiotics as a novel treatment. Brain Behav Immun Health 2022; 26:100541. [PMID: 36536630 PMCID: PMC9758412 DOI: 10.1016/j.bbih.2022.100541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 10/22/2022] [Indexed: 11/09/2022] Open
Abstract
Stress-associated disruptions in the development of frontolimbic regions may play a critical role in the emergence of adolescent-onset depression. These regions are particularly sensitive to Hypothalamic-Pituitary-Adrenal (HPA) axis signaling. The HPA axis is hyperactive in adolescent depression, and interventions that attenuate such hyperactivity hold promise as potential treatments. The Microbiome-Gut-Brain (MGB) axis is an important pathway through which stress dysregulates HPA-axis activity and thus exerts deleterious effects on the adolescent brain. Probiotic agents, which alter the gut microbiota composition by introducing bacterial strains with beneficial physiological effects, normalize aberrant HPA-axis activity and reduce depressive symptoms in both animal studies and adult clinical trials. While the potential utility of such agents in treating or preventing adolescent depression remains largely unexplored, recent data suggest the existence of an adolescent sensitive window during which probiotics may be especially efficacious in reducing depressive symptoms compared to effects observed in adult populations. In this review, we outline evidence that probiotic use may attenuate stress effects on frontolimbic development, providing a novel means of improving depressive symptoms among adolescent populations.
Collapse
Affiliation(s)
- Daniel Freimer
- University of California, San Francisco (UCSF), School of Medicine, USA
| | - Tony T. Yang
- University of California, San Francisco (UCSF), School of Medicine, USA
- Department of Psychiatry and Behavioral Sciences, Weill Institute for Neurosciences, USA
- Division of Child and Adolescent Psychiatry, USA
- The Langley Porter Psychiatric Institute, USA
| | - Tiffany C. Ho
- University of California, San Francisco (UCSF), School of Medicine, USA
- Department of Psychiatry and Behavioral Sciences, Weill Institute for Neurosciences, USA
| | - Olga Tymofiyeva
- University of California, San Francisco (UCSF), School of Medicine, USA
- Department of Radiology and Biomedical Imaging, USA
| | - Cherry Leung
- University of California, San Francisco (UCSF), School of Nursing, Department of Community Health Systems, USA
| |
Collapse
|
17
|
Esposito P, Kearns MM, Smith KB, Chandrasegaram R, Kadamani AK, Gandelman M, Liang J, Nikpoor N, Tompkins TA, Ismail N. The effects of antimicrobials and lipopolysaccharide on acute immune responsivity in pubertal male and female CD1 mice. COMPREHENSIVE PSYCHONEUROENDOCRINOLOGY 2022; 11:100147. [PMID: 35967925 PMCID: PMC9363646 DOI: 10.1016/j.cpnec.2022.100147] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 05/26/2022] [Accepted: 05/30/2022] [Indexed: 11/18/2022] Open
Abstract
Exposure to stress during critical periods of development-such as puberty-is associated with long-term disruptions in brain function and neuro-immune responsivity. However, the mechanisms underlying the effect of stress on the pubertal neuro-immune response has yet to be elucidated. Therefore, the objective of the current study was to investigate the effect antimicrobial and lipopolysaccharide (LPS) treatments on acute immune responsivity in pubertal male and female mice. Moreover, the potential for probiotic supplementation to mitigate these effects was also examined. 240 male and female CD1 mice were treated with one week of antimicrobial treatment (mixed antimicrobials or water) and probiotic treatment (L. rhamnosis R0011 and L. helveticus R0052 or L. helveticus R0052 and B. longum R0175) or placebo at five weeks of age. At six weeks of age (pubertal stress-sensitive period), the mice received a single injection of LPS or saline. Sickness behaviours were assessed, and mice were euthanized 8 h post-injection. Brain, blood, and intestinal samples were collected. The results indicated that the antimicrobial treatment reduced sickness behaviours, and potentiated LPS-induced plasma cytokine concentrations and pro-inflammatory markers in the pre-frontal cortex (PFC) and hippocampus, in a sex-dependent manner. However, probiotics reduced LPS-induced plasma cytokine concentrations along with hippocampal and PFC pro-inflammatory markers in a sex-dependent manner. L. rhamnosis R0011 and L. helveticus R0052 treatment also mitigated antimicrobial-induced plasma cytokine concentrations and sickness behaviours. These findings suggest that the microbiome is an important modulator of the pro-inflammatory immune response during puberty.
Collapse
Affiliation(s)
- Pasquale Esposito
- NISE Laboratory, School of Psychology, Faculty of Social Sciences, University of Ottawa, Ontario, K1N 6N5, Canada
| | - Madeleine M. Kearns
- NISE Laboratory, School of Psychology, Faculty of Social Sciences, University of Ottawa, Ontario, K1N 6N5, Canada
| | - Kevin B. Smith
- NISE Laboratory, School of Psychology, Faculty of Social Sciences, University of Ottawa, Ontario, K1N 6N5, Canada
| | - Rajini Chandrasegaram
- Department of Neuroscience, Faculty of Health Sciences, University of Cardiff, Cardiff, CF24 2FN, United Kingdom
| | - Anthony K. Kadamani
- NISE Laboratory, School of Psychology, Faculty of Social Sciences, University of Ottawa, Ontario, K1N 6N5, Canada
| | - Michelle Gandelman
- NISE Laboratory, School of Psychology, Faculty of Social Sciences, University of Ottawa, Ontario, K1N 6N5, Canada
| | - Jacky Liang
- NISE Laboratory, School of Psychology, Faculty of Social Sciences, University of Ottawa, Ontario, K1N 6N5, Canada
| | - Naghmeh Nikpoor
- Lallemand Health Solutions Inc, Montreal, Quebec, H1W 2N8, Canada
| | | | - Nafissa Ismail
- NISE Laboratory, School of Psychology, Faculty of Social Sciences, University of Ottawa, Ontario, K1N 6N5, Canada
- Brain and Mind Research Institute, University of Ottawa, Ottawa, Ontario, K1N 6N5, Canada
| |
Collapse
|
18
|
Sánchez-Garrido MA, García-Galiano D, Tena-Sempere M. Early programming of reproductive health and fertility: novel neuroendocrine mechanisms and implications in reproductive medicine. Hum Reprod Update 2022; 28:346-375. [PMID: 35187579 PMCID: PMC9071071 DOI: 10.1093/humupd/dmac005] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/29/2021] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND According to the Developmental Origins of Health and Disease (DOHaD) hypothesis, environmental changes taking place during early maturational periods may alter normal development and predispose to the occurrence of diverse pathologies later in life. Indeed, adverse conditions during these critical developmental windows of high plasticity have been reported to alter the offspring developmental trajectory, causing permanent functional and structural perturbations that in the long term may enhance disease susceptibility. However, while solid evidence has documented that fluctuations in environmental factors, ranging from nutrient availability to chemicals, in early developmental stages (including the peri-conceptional period) have discernible programming effects that increase vulnerability to develop metabolic perturbations, the impact and eventual mechanisms involved, of such developmental alterations on the reproductive phenotype of offspring have received less attention. OBJECTIVE AND RATIONALE This review will summarize recent advances in basic and clinical research that support the concept of DOHaD in the context of the impact of nutritional and hormonal perturbations, occurring during the periconceptional, fetal and early postnatal stages, on different aspects of reproductive function in both sexes. Special emphasis will be given to the effects of early nutritional stress on the timing of puberty and adult gonadotropic function, and to address the underlying neuroendocrine pathways, with particular attention to involvement of the Kiss1 system in these reproductive perturbations. The implications of such phenomena in terms of reproductive medicine will also be considered. SEARCH METHODS A comprehensive MEDLINE search, using PubMed as main interface, of research articles and reviews, published mainly between 2006 and 2021, has been carried out. Search was implemented using multiple terms, focusing on clinical and preclinical data from DOHaD studies, addressing periconceptional, gestational and perinatal programming of reproduction. Selected studies addressing early programming of metabolic function have also been considered, when relevant. OUTCOMES A solid body of evidence, from clinical and preclinical studies, has documented the impact of nutritional and hormonal fluctuations during the periconceptional, prenatal and early postnatal periods on pubertal maturation, as well as adult gonadotropic function and fertility. Furthermore, exposure to environmental chemicals, such as bisphenol A, and maternal stress has been shown to negatively influence pubertal development and gonadotropic function in adulthood. The underlying neuroendocrine pathways and mechanisms involved have been also addressed, mainly by preclinical studies, which have identified an, as yet incomplete, array of molecular and neurohormonal effectors. These include, prominently, epigenetic regulatory mechanisms and the hypothalamic Kiss1 system, which likely contribute to the generation of reproductive alterations in conditions of early nutritional and/or metabolic stress. In addition to the Kiss1 system, other major hypothalamic regulators of GnRH neurosecretion, such as γ-aminobutyric acid and glutamate, may be targets of developmental programming. WIDER IMPLICATIONS This review addresses an underdeveloped area of reproductive biology and medicine that may help to improve our understanding of human reproductive disorders and stresses the importance, and eventual pathogenic impact, of early determinants of puberty, adult reproductive function and fertility.
Collapse
Affiliation(s)
- Miguel Angel Sánchez-Garrido
- Instituto Maimónides de Investigación Biomédica de Cordoba (IMIBIC), Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
- Hospital Universitario Reina Sofia, Cordoba, Spain
| | - David García-Galiano
- Instituto Maimónides de Investigación Biomédica de Cordoba (IMIBIC), Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
- Hospital Universitario Reina Sofia, Cordoba, Spain
| | - Manuel Tena-Sempere
- Instituto Maimónides de Investigación Biomédica de Cordoba (IMIBIC), Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
- Hospital Universitario Reina Sofia, Cordoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Cordoba, Spain
- Institute of Biomedicine, University of Turku, Turku, Finland
| |
Collapse
|
19
|
Prenatal Exposure to an EDC Mixture, NeuroMix: Effects on Brain, Behavior, and Stress Responsiveness in Rats. TOXICS 2022; 10:toxics10030122. [PMID: 35324748 PMCID: PMC8954446 DOI: 10.3390/toxics10030122] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/22/2022] [Accepted: 02/26/2022] [Indexed: 12/11/2022]
Abstract
Humans and wildlife are exposed to endocrine-disrupting chemicals (EDCs) throughout their lives. Environmental EDCs are implicated in a range of diseases/disorders with developmental origins, including neurodevelopment and behavior. EDCs are most often studied one by one; here, we assessed outcomes induced by a mixture designed to represent the real-world situation of multiple simultaneous exposures. The choice of EDCs, which we refer to as “NeuroMix,” was informed by evidence for neurobiological effects in single-compound studies and included bisphenols, phthalates, vinclozolin, and perfluorinated, polybrominated, and polychlorinated compounds. Pregnant Sprague Dawley rats were fed the NeuroMix or vehicle, and then offspring of both sexes were assessed for effects on postnatal development and behaviors and gene expression in the brain in adulthood. In order to determine whether early-life EDCs predisposed to subsequent vulnerability to postnatal life challenges, a subset of rats were also given a stress challenge in adolescence. Prenatal NeuroMix exposure decreased body weight and delayed puberty in males but not females. In adulthood, NeuroMix caused changes in anxiety-like, social, and mate preference behaviors only in females. Effects of stress were predominantly observed in males. Several interactions of NeuroMix and stress were found, especially for the mate preference behavior and gene expression in the brain. These findings provide novel insights into how two realistic environmental challenges lead to developmental and neurobehavioral deficits, both alone and in combination, in a sex-specific manner.
Collapse
|
20
|
Sonuga-Barke EJS, Hanć T, Stehli A, Trampush JW, Kennedy M, Kreppner J, Rutter M, Swanson JM. Severe deprivation in early childhood leads to permanent growth stunting: Longitudinal analysis of height trajectories from childhood-to-adulthood. CHILD ABUSE & NEGLECT 2022; 123:105427. [PMID: 34896879 DOI: 10.1016/j.chiabu.2021.105427] [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: 04/15/2021] [Revised: 11/11/2021] [Accepted: 12/01/2021] [Indexed: 06/14/2023]
Abstract
BACKGROUND Childhood institutional deprivation is associated with growth stunting in childhood but long-term effects in adulthood remain uncertain. OBJECTIVE To examine the impact of global institutional deprivation experienced in early childhood on subsequent growth with a special focus on final adult height and puberty timing. PARTICIPANTS & SETTING The study was originally set in the UK, though some adoptive families lived abroad by the time of the adult follow up. 165 individuals adopted by UK families before 43 months of age from Romanian orphanages after the fall of the Ceaușescu regime in the early 1990's were compared to 51 non-deprived UK adoptees, adopted before the age of 6 months. METHODS The English and Romanian Adoptees (ERA) study is a 20-year longitudinal natural experiment on the effects of institutional deprivation on development. Key growth milestones were extracted from growth curve modelling of height data collected at ages 4, 6, 11, 15 and 23 years using a Bayesian approach to fit the JPA2 model. RESULTS Deprivation effects on height were present at the take-off point of accelerating adolescent growth and persisted into adulthood - the largest effects being for individuals who experienced over six months of deprivation. Deprivation was associated with earlier take-off and achievement of peak height velocity of adolescent growth acceleration - an effect driven largely by females' data and correlated with parent ratings of pubertal development. CONCLUSIONS Early deprivation appears to reset tempo of growth early in development leading to permanent growth stunting in adulthood and accelerated onset of puberty, specifically in females.
Collapse
Affiliation(s)
- Edmund J S Sonuga-Barke
- Department of Child & Adolescent Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, Kings College London, DeCrespigny Park, London SE5 8AF, UK; Department of Child & Adolescent Psychiatry, Aarhus University, Denmark.
| | - Tomasz Hanć
- Institute of Human Biology and Evolution, Faculty of Biology, Adam Mickiewicz University, ul. Uniwersytetu Poznanskiego 6, 61-614 Poznan, Poland
| | - Annamarie Stehli
- Department of Pediatrics, University of California, Irvine, 333 The City Blvd. West, Suite 800, Orange, CA 92868-4482, USA
| | - Joey W Trampush
- Department of Psychiatry and the Behavioural Sciences, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, Suite 2200, Los Angeles, CA 90033, USA
| | - Mark Kennedy
- Department of Child & Adolescent Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, Kings College London, DeCrespigny Park, London SE5 8AF, UK
| | - Jana Kreppner
- Centre for Innovation in Mental Health, School of Psychology, University of Southampton, University Road, Southampton SO17 1BJ, UK
| | - Michael Rutter
- Social, Developmental, Genetic Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, Kings College London, DeCrespigny Park, London, SE5 8AF, UK
| | - James M Swanson
- Department of Pediatrics, University of California, Irvine, 333 The City Blvd. West, Suite 800, Orange, CA 92868-4482, USA
| |
Collapse
|
21
|
Natale NR, Kent M, Fox N, Vavra D, Lambert K. Neurobiological effects of a probiotic-supplemented diet in chronically stressed male Long-Evans rats: Evidence of enhanced resilience. IBRO Neurosci Rep 2021; 11:207-215. [PMID: 34849506 PMCID: PMC8607205 DOI: 10.1016/j.ibneur.2021.10.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 10/25/2021] [Accepted: 10/28/2021] [Indexed: 12/15/2022] Open
Abstract
Probiotics that regulate the microbiome-gut-brain axis and provide mental health benefits to the host are referred to as psychobiotics. Preclinical studies have demonstrated psychobiotic effects on early life stress-induced anxiety- and depression-related behavior in rodents; however, the specific mechanisms remain ill-defined. In the current study, we investigated the effects of probiotic supplementation on neurobiological responses to chronic stress in adult male Long-Evans rats. Twenty-four rats were randomly assigned to probiotic (PB) or vehicle control (VEH) groups, then to either chronic unpredictable stress (CUS) or no-stress control (CON) conditions within each group (n = 6/subgroup). We hypothesized that PB supplementation would reduce markers of anxiety and enhance emotional resilience, especially in the CUS animals. In the cognitive uncertainty task, a nonsignificant trend was observed indicating that the PB-supplemented animals spent more time oriented toward the food reward than VEH animals. In the open-field task, CUS-PB animals spent more time in the center of the arena than CUS-VEH animals, an effect not observed between the two CON groups. In the swim task, the PB animals, regardless of stress assignment, exhibited increased floating, suggesting a conserved response in a challenging context. Focusing on the endocrine measures, higher dehydroepiandrosterone (DHEA)-to-corticosterone fecal metabolite ratios, a correlate of emotional resilience, were observed in PB animals. Further, PB animals exhibited reduced microglia immunoreactivity in the basolateral amygdala, possibly indicating a neuroprotective effect of PB supplements in this rodent model. These results provide evidence that PB supplementation interacts with stress exposure to influence adaptive responses associated with endocrine, neural, and behavioral indices of anxiety.
Collapse
Affiliation(s)
- Nick R. Natale
- Dept of Psychology, University of Richmond, VA 23173, USA
| | - Molly Kent
- Dept of Biology, Virginia Military Institute, Lexington, VA 24450, USA
| | - Nathan Fox
- Dept of Psychology, University of Richmond, VA 23173, USA
| | - Dylan Vavra
- Dept of Psychology, University of Richmond, VA 23173, USA
| | - Kelly Lambert
- Dept of Psychology, University of Richmond, VA 23173, USA
| |
Collapse
|
22
|
Ellis SN, Honeycutt JA. Sex Differences in Affective Dysfunction and Alterations in Parvalbumin in Rodent Models of Early Life Adversity. Front Behav Neurosci 2021; 15:741454. [PMID: 34803622 PMCID: PMC8600234 DOI: 10.3389/fnbeh.2021.741454] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 10/13/2021] [Indexed: 01/08/2023] Open
Abstract
The early life environment markedly influences brain and behavioral development, with adverse experiences associated with increased risk of anxiety and depressive phenotypes, particularly in females. Indeed, early life adversity (ELA) in humans (i.e., caregiver deprivation, maltreatment) and rodents (i.e., maternal separation, resource scarcity) is associated with sex-specific emergence of anxious and depressive behaviors. Although these disorders show clear sex differences in humans, little attention has been paid toward evaluating sex as a biological variable in models of affective dysfunction; however, recent rodent work suggests sex-specific effects. Two widely used rodent models of ELA approximate caregiver deprivation (i.e., maternal separation) and resource scarcity (i.e., limited bedding). While these approaches model aspects of ELA experienced in humans, they span different portions of the pre-weaning developmental period and may therefore differentially contribute to underlying mechanistic risk. This is borne out in the literature, where evidence suggests differences in trajectories of behavior depending on the type of ELA and/or sex; however, the neural underpinning of these differences is not well understood. Because anxiety and depression are thought to involve dysregulation in the balance of excitatory and inhibitory signaling in ELA-vulnerable brain regions (e.g., prefrontal cortex, amygdala, hippocampus), outcomes are likely driven by alterations in local and/or circuit-specific inhibitory activity. The most abundant GABAergic subtypes in the brain, accounting for approximately 40% of inhibitory neurons, contain the calcium-binding protein Parvalbumin (PV). As PV-expressing neurons have perisomatic and proximal dendritic targets on pyramidal neurons, they are well-positioned to regulate excitatory/inhibitory balance. Recent evidence suggests that PV outcomes following ELA are sex, age, and region-specific and may be influenced by the type and timing of ELA. Here, we suggest the possibility of a combined role of PV and sex hormones driving differences in behavioral outcomes associated with affective dysfunction following ELA. This review evaluates the literature across models of ELA to characterize neural (PV) and behavioral (anxiety- and depressive-like) outcomes as a function of sex and age. Additionally, we detail a putative mechanistic role of PV on ELA-related outcomes and discuss evidence suggesting hormone influences on PV expression/function which may help to explain sex differences in ELA outcomes.
Collapse
Affiliation(s)
- Seneca N Ellis
- Program in Neuroscience, Bowdoin College, Brunswick, ME, United States
| | - Jennifer A Honeycutt
- Program in Neuroscience, Bowdoin College, Brunswick, ME, United States.,Department of Psychology, Bowdoin College, Brunswick, ME, United States
| |
Collapse
|
23
|
Gee DG. Early Adversity and Development: Parsing Heterogeneity and Identifying Pathways of Risk and Resilience. Am J Psychiatry 2021; 178:998-1013. [PMID: 34734741 DOI: 10.1176/appi.ajp.2021.21090944] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Adversity early in life is common and is a major risk factor for the onset of psychopathology. Delineating the neurodevelopmental pathways by which early adversity affects mental health is critical for early risk identification and targeted treatment approaches. A rapidly growing cross-species literature has facilitated advances in identifying the mechanisms linking adversity with psychopathology, specific dimensions of adversity and timing-related factors that differentially relate to outcomes, and protective factors that buffer against the effects of adversity. Yet, vast complexity and heterogeneity in early environments and neurodevelopmental trajectories contribute to the challenges of understanding risk and resilience in the context of early adversity. In this overview, the author highlights progress in four major areas-mechanisms, heterogeneity, developmental timing, and protective factors; synthesizes key challenges; and provides recommendations for future research that can facilitate progress in the field. Translation across species and ongoing refinement of conceptual models have strong potential to inform prevention and intervention strategies that can reduce the immense burden of psychopathology associated with early adversity.
Collapse
Affiliation(s)
- Dylan G Gee
- Department of Psychology, Yale University, New Haven, Conn
| |
Collapse
|
24
|
Phumsatitpong C, Wagenmaker ER, Moenter SM. Neuroendocrine interactions of the stress and reproductive axes. Front Neuroendocrinol 2021; 63:100928. [PMID: 34171353 PMCID: PMC8605987 DOI: 10.1016/j.yfrne.2021.100928] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 06/17/2021] [Accepted: 06/19/2021] [Indexed: 01/27/2023]
Abstract
Reproduction is controlled by a sequential regulation of the hypothalamo-pituitary-gonadal (HPG) axis. The HPG axis integrates multiple inputs to maintain proper reproductive functions. It has long been demonstrated that stress alters fertility. Nonetheless, the central mechanisms of how stress interacts with the reproductive system are not fully understood. One of the major pathways that is activated during the stress response is the hypothalamo-pituitary-adrenal (HPA) axis. In this review, we discuss several aspects of the interactions between these two neuroendocrine systems to offer insights to mechanisms of how the HPA and HPG axes interact. We have also included discussions of other systems, for example GABA-producing neurons, where they are informative to the overall picture of stress effects on reproduction.
Collapse
Affiliation(s)
- Chayarndorn Phumsatitpong
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Elizabeth R Wagenmaker
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Suzanne M Moenter
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States; Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States; Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, United States.
| |
Collapse
|
25
|
Methods and Challenges in Investigating Sex-Specific Consequences of Social Stressors in Adolescence in Rats: Is It the Stress or the Social or the Stage of Development? Curr Top Behav Neurosci 2021; 54:23-58. [PMID: 34455576 DOI: 10.1007/7854_2021_245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Adolescence is a time of social learning and social restructuring that is accompanied by changes in both the hypothalamic-pituitary-gonadal axis and the hypothalamic-pituitary-adrenal (HPA) axis. The activation of these axes by puberty and stressors, respectively, shapes adolescent development. Models of social stress in rats are used to understand the consequences of perturbations of the social environment for ongoing brain development. This paper reviews the challenges in investigating the sex-specific consequences of social stressors, sex differences in the models of social stress used in rats and the sex-specific effects on behaviour and provides an overview of sex differences in HPA responding to stressors, the variability in pubertal development and in strains of rats that require consideration in conducting such research, and directions for future research.
Collapse
|
26
|
Psychobiotic interventions for anxiety in young people: a systematic review and meta-analysis, with youth consultation. Transl Psychiatry 2021; 11:352. [PMID: 34131108 PMCID: PMC8206413 DOI: 10.1038/s41398-021-01422-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 04/08/2021] [Accepted: 04/19/2021] [Indexed: 01/10/2023] Open
Abstract
The human gut microbiome influence on brain function and mental health is an emerging area of intensive research. Animal and human research indicates adolescence as a sensitive period when the gut-brain axis is fine-tuned, where dietary interventions to change the microbiome may have long-lasting consequences for mental health. This study reports a systematic review and meta-analysis of microbiota-targeted (psychobiotics) interventions on anxiety in youth, with discussion of a consultation on the acceptability of psychobiotic interventions for mental health management amongst youth with lived experience. Six databases were searched for controlled trials in human samples (age range: 10-24 years) seeking to reduce anxiety. Post intervention outcomes were extracted as standard mean differences (SMDs) and pooled based on a random-effects model. 5416 studies were identified: 14 eligible for systematic review and 10 eligible for meta-analysis (total of 324 experimental and 293 control subjects). The meta-analysis found heterogeneity I2 was 12% and the pooled SMD was -0.03 (95% CI: -0.21, 0.14), indicating an absence of effect. One study presented with low bias risk, 5 with high, and 4 with uncertain risk. Accounting for risk, sensitivities analysis revealed a SMD of -0.16 (95% CI: -0.38, 0.07), indicative of minimal efficacy of psychobiotics for anxiety treatment in humans. There is currently limited evidence for use of psychobiotics to treat anxiety in youth. However, future progress will require a multidisciplinary research approach, which gives priority to specifying mechanisms in the human models, providing causal understanding, and addressing the wider context, and would be welcomed by anxious youths.
Collapse
|
27
|
Pincus M, Godfrey JR, Feczko E, Earl E, Miranda-Dominguez O, Fair D, Wilson ME, Sanchez MM, Kelly C. Chronic psychosocial stress and experimental pubertal delay affect socioemotional behavior and amygdala functional connectivity in adolescent female rhesus macaques. Psychoneuroendocrinology 2021; 127:105154. [PMID: 33647571 PMCID: PMC11578542 DOI: 10.1016/j.psyneuen.2021.105154] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 12/24/2020] [Accepted: 01/28/2021] [Indexed: 12/20/2022]
Abstract
In females, pubertal onset appears to signal the opening of a window of increased vulnerability to the effects of stress on neurobehavioral development. What is the impact of pubertal timing on this process? We assessed the effects of pubertal timing and stress on behavior and amygdala functional connectivity (FC) in adolescent female macaques, whose social hierarchy provides an ethologically valid model of chronic psychosocial stress. Monkeys experienced puberty spontaneously (n = 34) or pubertal delay via Lupron treatment from age 16-33 months (n = 36). We examined the effects of stress (continuous dimension spanning dominant/low-stress to subordinate/high-stress) and experimental pubertal delay (Lupron-treated vs. Control) on socioemotional behavior and FC at 43-46 months, after all animals had begun puberty. Regardless of treatment, subordinate monkeys were more submissive and less affiliative, and exhibited weaker FC between amygdala and dorsolateral prefrontal cortex and stronger FC between amygdala and temporal pole. Regardless of social rank, Lupron-treated monkeys were also more submissive and less affiliative but were less anxious and exhibited less displacement behavior in a "Human Intruder" task than untreated monkeys; they exhibited stronger FC between amygdala and orbitofrontal cortex. No interactions between rank and Lupron treatment were observed. These similar behavioral outcomes may reflect the common factor of delayed puberty - whether this is stress-related (untreated subordinate animals) or pharmacologically-induced (treated animals). In the brain, however, delayed puberty and subordination stress had separable effects, suggesting that the overlapping socioemotional outcomes may be mediated by distinct neuroplastic mechanisms. To gain further insights, additional longitudinal studies are required.
Collapse
Affiliation(s)
- Melanie Pincus
- Division of Developmental and Cognitive Neuroscience, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Jodi R Godfrey
- Division of Developmental and Cognitive Neuroscience, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Eric Feczko
- Masonic Institute for the Developing Brain (MIDB), University of Minnesota, Minneapolis, MN, USA; Institute of Child Development, College of Education and Human Development, University of Minnesota, Minneapolis, MN, USA; Department of Pediatrics, University of Minnesota Medical School, University of Minnesota, Minneapolis, MN, USA
| | - Eric Earl
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA
| | - Oscar Miranda-Dominguez
- Masonic Institute for the Developing Brain (MIDB), University of Minnesota, Minneapolis, MN, USA; Institute of Child Development, College of Education and Human Development, University of Minnesota, Minneapolis, MN, USA; Department of Pediatrics, University of Minnesota Medical School, University of Minnesota, Minneapolis, MN, USA
| | - Damien Fair
- Masonic Institute for the Developing Brain (MIDB), University of Minnesota, Minneapolis, MN, USA; Institute of Child Development, College of Education and Human Development, University of Minnesota, Minneapolis, MN, USA; Department of Pediatrics, University of Minnesota Medical School, University of Minnesota, Minneapolis, MN, USA
| | - Mark E Wilson
- Division of Developmental and Cognitive Neuroscience, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA; Department of Psychiatry & Behavioral Sciences, Emory University, USA
| | - Mar M Sanchez
- Division of Developmental and Cognitive Neuroscience, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Clare Kelly
- School of Psychology, Trinity College Dublin, Dublin, Ireland; Department of Psychiatry at the School of Medicine, Trinity College Dublin, Dublin, Ireland; Trinity College Institute of Neuroscience, Trinity College, Dublin, Ireland
| |
Collapse
|
28
|
Tremblay A, Lingrand L, Maillard M, Feuz B, Tompkins TA. The effects of psychobiotics on the microbiota-gut-brain axis in early-life stress and neuropsychiatric disorders. Prog Neuropsychopharmacol Biol Psychiatry 2021; 105:110142. [PMID: 33069817 DOI: 10.1016/j.pnpbp.2020.110142] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/28/2020] [Accepted: 10/12/2020] [Indexed: 02/07/2023]
Abstract
Psychobiotics are considered among potential avenues for modulating the bidirectional communication between the gastrointestinal tract and central nervous system, defined as the microbiota-gut-brain axis (MGBA). Even though causality has not yet been established, intestinal dysbiosis has emerged as a hallmark of several diseases, including neuropsychiatric disorders (NPDs). The fact that the microbiota and central nervous system are co-developing during the first years of life has provided a paradigm suggesting a potential role of psychobiotics for earlier interventions. Studies in animal models of early-life stress (ELS) have shown that they can counteract the pervasive effects of stress during this crucial developmental period, and rescue behavioral symptoms related to anxiety and depression later in life. In humans, evidence from clinical studies on the efficacy of psychobiotics at improving mental outcomes in most NPDs remain limited, except for major depressive disorder for which more studies are available. Consequently, the beneficial effect of psychobiotics on depression-related outcomes in adults are becoming clearer. While the specific mechanisms at play remain elusive, the effect of psychobiotics are generally considered to involve the hypothalamic-pituitary-adrenal axis, intestinal permeability, and inflammation. It is anticipated that future clinical studies will explore the potential role of psychobiotics at mitigating the risk developing NPDs in vulnerable individuals or in the context of childhood adversity. However, such studies remain challenging at present in terms of design and target populations; the profound impact of stress on the proper development of the MGBA during the first year of life is becoming increasingly recognized, but the trajectories post-ELS in humans and the mechanisms by which stress affects the susceptibility to various NPDs are still ill-defined. As psychobiotics are likely to exert both shared and specific mechanisms, a better definition of target subpopulations would allow to tailor psychobiotics selection by aligning mechanistic properties with known pathophysiological mechanisms or risk factors. Here we review the available evidence from clinical and preclinical studies supporting a role for psychobiotics at ameliorating depression-related outcomes, highlighting the knowledge gaps and challenges associated with conducting longitudinal studies to address outstanding key questions in the field.
Collapse
Affiliation(s)
- Annie Tremblay
- Rosell® Institute for Microbiome and Probiotics, 6100 Royalmount Avenue, Montreal, Quebec H4P 2R2, Canada
| | - Lucie Lingrand
- Lallemand Health Solutions, 19 Rue des Briquetiers, 31702 Blagnac, France
| | - Morgane Maillard
- Lallemand Health Solutions, 19 Rue des Briquetiers, 31702 Blagnac, France
| | - Berengere Feuz
- Lallemand Health Solutions, 19 Rue des Briquetiers, 31702 Blagnac, France
| | - Thomas A Tompkins
- Rosell® Institute for Microbiome and Probiotics, 6100 Royalmount Avenue, Montreal, Quebec H4P 2R2, Canada.
| |
Collapse
|
29
|
Lopizzo N, Mazzelli M, Zonca V, Begni V, D'Aprile I, Cattane N, Pariante CM, Riva MA, Cattaneo A. Alterations in 'inflammatory' pathways in the rat prefrontal cortex as early biological predictors of the long-term negative consequences of exposure to stress early in life. Psychoneuroendocrinology 2021; 124:104794. [PMID: 33429258 DOI: 10.1016/j.psyneuen.2020.104794] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 06/24/2020] [Accepted: 06/25/2020] [Indexed: 12/17/2022]
Abstract
Early life stress, especially when experienced during the first period of life, affects the brain developmental trajectories leading to an enhanced vulnerability for stress-related psychiatric disorders later in life. Although both clinical and preclinical studies clearly support this association, the biological pathways deregulated by such exposure, and the effects in shaping the neurodevelopmental trajectories, have so far been poorly investigated. By using the prenatal stress (PNS) model, a well-established rat model of early life stress, we performed transcriptomic analyses in the prefrontal cortex of rats exposed or not to PNS and sacrificed at different postnatal days (PNDs 21, 40, 62). We first investigated the long-lasting mechanisms and pathways affected in the PFC. We have decided to focus on the prefrontal cortex because we have previously shown that this brain region is highly sensitive to PNS exposure. We found that adult animals exposed to PNS show alterations in 389 genes, mainly involved in stress and inflammatory signalling. We then wanted to establish whether PNS exposure could also affect the neurodevelopmental trajectories in order to identify the most critical temporal window. We found that PNS rats show the most significant changes during adolescence (between PND 40 versus PND 21), with alterations of several pathways related to stress, inflammation and metabolism, which were maintained until adulthood.
Collapse
Affiliation(s)
- Nicola Lopizzo
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy; Department of Pharmacological and Biomolecular Sciences, University of Milan, Italy
| | - Monica Mazzelli
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy; Department of Pharmacological and Biomolecular Sciences, University of Milan, Italy
| | - Valentina Zonca
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy; Stress, Psychiatry and Immunology Laboratory, Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, UK
| | - Veronica Begni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Italy
| | - Ilari D'Aprile
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Nadia Cattane
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Carmine M Pariante
- Stress, Psychiatry and Immunology Laboratory, Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, UK
| | - Marco A Riva
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Italy
| | - Annamaria Cattaneo
- Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy; Department of Pharmacological and Biomolecular Sciences, University of Milan, Italy.
| |
Collapse
|
30
|
Holingue C, Budavari AC, Rodriguez KM, Zisman CR, Windheim G, Fallin MD. Sex Differences in the Gut-Brain Axis: Implications for Mental Health. Curr Psychiatry Rep 2020; 22:83. [PMID: 33216233 PMCID: PMC7717677 DOI: 10.1007/s11920-020-01202-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/20/2020] [Indexed: 12/17/2022]
Abstract
PURPOSE OF REVIEW The purpose of this article is to highlight how sex differences in the gut-brain axis may contribute to the discrepancies in incidence of neurodevelopmental, psychiatric, and neurodegenerative disorders between females and males. We focus on autism spectrum disorder, psychotic disorders, stress and anxiety disorders, depression, Alzheimer's disease, and Parkinson's disease and additionally discuss the comorbidity between inflammatory bowel disorder and mental health disorders. RECENT FINDINGS Human and animal studies show that sex may modify the relationship between the gut or immune system and brain and behavior. Sex also appears to modify the effect of microbial treatments such as probiotics and antibiotics on brain and behavior. There is emerging evidence that assessing the role of sex in the gut-brain axis may help elucidate the etiology of and identify effective treatments for neurodevelopmental, psychiatric, and neurodegenerative disorders.
Collapse
Affiliation(s)
- Calliope Holingue
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA.
- Department of Neuropsychology, Kennedy Krieger Institute, Baltimore, MD, USA.
- , Baltimore, USA.
| | - Alexa Curhan Budavari
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Katrina M Rodriguez
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Corina R Zisman
- Section on the Neurobiology of Fear and Anxiety, National Institute of Mental Health, Bethesda, MD, USA
| | - Grace Windheim
- Public Health Studies, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - M Daniele Fallin
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| |
Collapse
|
31
|
Lach G, Fülling C, Bastiaanssen TFS, Fouhy F, Donovan ANO, Ventura-Silva AP, Stanton C, Dinan TG, Cryan JF. Enduring neurobehavioral effects induced by microbiota depletion during the adolescent period. Transl Psychiatry 2020; 10:382. [PMID: 33159036 PMCID: PMC7648059 DOI: 10.1038/s41398-020-01073-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/15/2020] [Accepted: 10/05/2020] [Indexed: 12/20/2022] Open
Abstract
The gut microbiota is an essential regulator of many aspects of host physiology. Disruption of gut microbial communities affects gut-brain communication which ultimately can manifest as changes in brain function and behaviour. Transient changes in gut microbial composition can be induced by various intrinsic and extrinsic factors, however, it is possible that enduring shifts in the microbiota composition can be achieved by perturbation at a timepoint when the gut microbiota has not fully matured or is generally unstable, such as during early life or ageing. In this study, we investigated the effects of 3-week microbiota depletion with antibiotic treatment during the adolescent period and in adulthood. Following a washout period to restore the gut microbiota, behavioural and molecular hallmarks of gut-brain communication were investigated. Our data revealed that transient microbiota depletion had long-lasting effects on microbiota composition and increased anxiety-like behaviour in mice exposed to antibiotic treatment during adolescence but not in adulthood. Similarly, gene expression in the amygdala was more severely affected in mice treated during adolescence. Taken together these data highlight the vulnerability of the gut microbiota during the critical adolescent period and the long-lasting impact manipulations of the microbiota can have on gene expression and behaviour in adulthood.
Collapse
Affiliation(s)
- Gilliard Lach
- grid.7872.a0000000123318773APC Microbiome Ireland, University College Cork, Cork, Ireland ,grid.4305.20000 0004 1936 7988Present Address: University of Edinburgh, Edinburgh, Scotland UK
| | - Christine Fülling
- grid.7872.a0000000123318773APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Thomaz F. S. Bastiaanssen
- grid.7872.a0000000123318773APC Microbiome Ireland, University College Cork, Cork, Ireland ,grid.7872.a0000000123318773Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Fiona Fouhy
- grid.7872.a0000000123318773APC Microbiome Ireland, University College Cork, Cork, Ireland ,grid.6435.40000 0001 1512 9569Teagasc Food Research Centre, Food Biosciences Department, Moorepark, Fermoy, Ireland
| | - Aoife N. O’ Donovan
- grid.7872.a0000000123318773APC Microbiome Ireland, University College Cork, Cork, Ireland ,grid.6435.40000 0001 1512 9569Teagasc Food Research Centre, Food Biosciences Department, Moorepark, Fermoy, Ireland ,grid.7872.a0000000123318773School of Microbiology, University College Cork, Cork, Ireland
| | | | - Catherine Stanton
- grid.7872.a0000000123318773APC Microbiome Ireland, University College Cork, Cork, Ireland ,grid.6435.40000 0001 1512 9569Teagasc Food Research Centre, Food Biosciences Department, Moorepark, Fermoy, Ireland
| | - Timothy G. Dinan
- grid.7872.a0000000123318773APC Microbiome Ireland, University College Cork, Cork, Ireland ,grid.7872.a0000000123318773Department of Psychiatry and Neurobehavioural Sciences, University College Cork, Cork, Ireland
| | - John F. Cryan
- grid.7872.a0000000123318773APC Microbiome Ireland, University College Cork, Cork, Ireland ,grid.7872.a0000000123318773Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| |
Collapse
|
32
|
King RB, Paukner A. Translating Socioeconomic Status Between Nonhuman Primate and Human Studies in Health Research. CHILD DEVELOPMENT PERSPECTIVES 2020. [DOI: 10.1111/cdep.12386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
| | - Annika Paukner
- National Institutes of Health
- Nottingham Trent University
| |
Collapse
|
33
|
van der Veen R, Bonapersona V, Joëls M. The relevance of a rodent cohort in the Consortium on Individual Development. Dev Cogn Neurosci 2020; 45:100846. [PMID: 32957026 PMCID: PMC7509002 DOI: 10.1016/j.dcn.2020.100846] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 07/29/2020] [Accepted: 08/23/2020] [Indexed: 12/31/2022] Open
Abstract
One of the features of the Consortium on Individual Development is the existence of a rodent cohort, in parallel with the human cohorts. Here we give an overview of the current status. We first elaborate on the choice of rat and mouse models mimicking early life adverse or beneficial conditions during development. We performed a systematic literature search on early life adversity and adult social behavior to address the status quo. Next, we describe the behavioral tasks we used and designed to examine behavioral control and social competence in rodents. The results so far indicate that manipulation of the environment in the first postnatal week only subtly affects social behavior. Stronger effects were seen in the model that targeted early adolescence; once adult, these rats are characterized by increased attention, a higher degree of impulsiveness and reduced social interest in peers. Many experiments in our rodent models with tightly controlled conditions were inspired by findings in human cohorts, and now allow in-depth mechanistic investigations. Vice versa, some of the findings in rodents are currently followed up by dedicated investigations in the human cohorts. This exemplifies the added value of animal investigations in a consortium encompassing primarily human developmental cohorts.
Collapse
Affiliation(s)
- Rixt van der Veen
- Dept. Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; Faculty of Social and Behavioral Sciences, Leiden University, Leiden, the Netherlands.
| | - Valeria Bonapersona
- Dept. Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Marian Joëls
- Dept. Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
34
|
Noschang C, Krolow R, Arcego DM, Marcolin M, Ferreira AG, da Cunha AA, Wyse ATS, Dalmaz C. Early-life stress affects behavioral and neurochemical parameters differently in male and female juvenile Wistar rats. Int J Dev Neurosci 2020; 80:547-557. [PMID: 32683715 DOI: 10.1002/jdn.10050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 06/14/2020] [Accepted: 07/10/2020] [Indexed: 12/15/2022] Open
Abstract
Neonatal handling is an early life stressor that leads to behavioral and neurochemical changes in adult rats in a sex-specific manner and possibly affects earlier stages of development. Here, we investigated the effects of neonatal handling (days 1-10 after birth) on juvenile rats focusing on biochemical parameters and olfactory memory after weaning. Male neonatal handled rats performed more crossings on the hole-board task, increased Na+ /K+ -ATPase activity in the olfactory bulb, and decreased acetylcholinesterase activity in the hippocampus versus non-handled males. Female neonatal handled animals increased the number of rearing and nose-pokes on the hole-board task, decreased glutathione peroxidase activity, and total thiol content in the hippocampus versus non-handled females. This study reinforces that early life stress affects behavioral and neurochemical parameters in a sex-specific manner even before the puberty onset.
Collapse
Affiliation(s)
- C Noschang
- Departamento de Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - R Krolow
- Departamento de Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - D M Arcego
- Departamento de Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - M Marcolin
- Departamento de Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - A G Ferreira
- Departamento de Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - A A da Cunha
- Departamento de Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - A T S Wyse
- Departamento de Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - C Dalmaz
- Departamento de Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| |
Collapse
|
35
|
Eck SR, Ardekani CS, Salvatore M, Luz S, Kim ED, Rogers CM, Hall A, Lee DE, Famularo ST, Bhatnagar S, Bangasser DA. The effects of early life adversity on growth, maturation, and steroid hormones in male and female rats. Eur J Neurosci 2020; 52:2664-2680. [PMID: 31660665 PMCID: PMC8027906 DOI: 10.1111/ejn.14609] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 10/18/2019] [Accepted: 10/23/2019] [Indexed: 02/06/2023]
Abstract
Early life adversity is a risk factor for psychiatric disorders, yet the mechanisms by which adversity increases this risk are still being delineated. Here, we used a limited bedding and nesting (LBN) manipulation in rats that models a low resource environment to examine effects on growth, developmental milestones, and endocrine endpoints. In LBN, dams and pups, from pups' postnatal days 2-9, are exposed to an environment where dams lack proper materials to build a nest. This manipulation is compared to control housing conditions, where rat dams have access to ample nesting materials and enrichment throughout pups' development. We found that the LBN condition altered maternal care, increasing pup-directed behaviors while reducing self-care. This, perhaps compensatory, increase in nursing and attention to pups did not mitigate against changes in metabolism, as LBN reduced weight gain in both sexes and this effect persisted into adulthood. Although adult stress hormone levels in both sexes and vaginal opening and estrous cycle length in females were not disrupted, there was other evidence of endocrine dysregulation. Compared to controls, LBN rats of both sexes had shortened anogenital distances, indicating reduced androgen exposure. LBN males also had higher plasma estradiol levels in adulthood. This combination of results suggests that LBN causes a demasculinizing effect in males that could contribute to lasting changes in the brain and behavior. Importantly, alterations in metabolic and endocrine systems due to early life adversity could be one mechanism by which stress early in life increases risk for later disease.
Collapse
Affiliation(s)
- Samantha R. Eck
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, USA
| | - Cory S. Ardekani
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, USA
| | - Madeleine Salvatore
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, USA
| | - Sandra Luz
- Department of Anesthesiology and Critical Care, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Eric D. Kim
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, USA
| | - Charleanne M. Rogers
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, USA
| | - Arron Hall
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, USA
| | - Demetrius E. Lee
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, USA
| | - Sydney T. Famularo
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, USA
| | - Seema Bhatnagar
- Department of Anesthesiology and Critical Care, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA,University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Debra A. Bangasser
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, USA
| |
Collapse
|
36
|
Knop J, van IJzendoorn MH, Bakermans-Kranenburg MJ, Joëls M, van der Veen R. Maternal care of heterozygous dopamine receptor D4 knockout mice: Differential susceptibility to early-life rearing conditions. GENES BRAIN AND BEHAVIOR 2020; 19:e12655. [PMID: 32306548 PMCID: PMC7540036 DOI: 10.1111/gbb.12655] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 03/30/2020] [Accepted: 04/13/2020] [Indexed: 12/27/2022]
Abstract
The differential susceptibility hypothesis proposes that individuals who are more susceptible to the negative effects of adverse rearing conditions may also benefit more from enriched environments. Evidence derived from human experiments suggests the lower efficacy dopamine receptor D4 (DRD4) 7‐repeat as a main factor in exhibiting these for better and for worse characteristics. However, human studies lack the genetic and environmental control offered by animal experiments, complicating assessment of causal relations. To study differential susceptibility in an animal model, we exposed Drd4+/− mice and control litter mates to a limited nesting/bedding (LN), standard nesting (SN) or communal nesting (CN) rearing environment from postnatal day (P) 2‐14. Puberty onset was examined from P24 to P36 and adult females were assessed on maternal care towards their own offspring. In both males and females, LN reared mice showed a delay in puberty onset that was partly mediated by a reduction in body weight at weaning, irrespective of Drd4 genotype. During adulthood, LN reared females exhibited characteristics of poor maternal care, whereas dams reared in CN environments showed lower rates of unpredictability towards their own offspring. Differential susceptibility was observed only for licking/grooming levels of female offspring towards their litter; LN reared Drd4+/− mice exhibited the lowest and CN reared Drd4+/− mice the highest levels of licking/grooming. These results indicate that both genetic and early‐environmental factors play an important role in shaping maternal care of the offspring for better and for worse.
Collapse
Affiliation(s)
- Jelle Knop
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.,Faculty of Social and Behavioural Sciences, Leiden University, Leiden, The Netherlands
| | - Marinus H van IJzendoorn
- Department of Psychology, Education and Child Studies, Erasmus University Rotterdam, Rotterdam, The Netherlands.,Primary Care Unit, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | | | - Marian Joëls
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.,University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Rixt van der Veen
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.,Faculty of Social and Behavioural Sciences, Leiden University, Leiden, The Netherlands
| |
Collapse
|
37
|
White JD, Kaffman A. The Moderating Effects of Sex on Consequences of Childhood Maltreatment: From Clinical Studies to Animal Models. Front Neurosci 2019; 13:1082. [PMID: 31680821 PMCID: PMC6797834 DOI: 10.3389/fnins.2019.01082] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 09/25/2019] [Indexed: 12/12/2022] Open
Abstract
Stress has pronounced effects on the brain, and thus behavioral outputs. This is particularly true when the stress occurs during vulnerable points in development. A review of the clinical literature regarding the moderating effects of sex on psychopathology in individuals exposed to childhood maltreatment (CM) is complicated by a host of variables that are difficult to quantify and control in clinical settings. As a result, the precise role of sex in moderating the consequences of CM remains elusive. In this review, we explore the rationale for studying this important question and their implications for treatment. We examine this issue using the threat/deprivation conceptual framework and highlight a growing body of work demonstrating important sex differences in human studies and in animal models of early life stress (ELS). The challenges and obstacles for effectively studying this question are reviewed and are followed by recommendations on how to move forward at the clinical and preclinical settings. We hope that this review will help inspire additional studies on this important topic.
Collapse
Affiliation(s)
- Jordon D White
- Department of Psychiatry, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - Arie Kaffman
- Department of Psychiatry, Yale School of Medicine, Yale University, New Haven, CT, United States
| |
Collapse
|
38
|
Cryan JF, O'Riordan KJ, Cowan CSM, Sandhu KV, Bastiaanssen TFS, Boehme M, Codagnone MG, Cussotto S, Fulling C, Golubeva AV, Guzzetta KE, Jaggar M, Long-Smith CM, Lyte JM, Martin JA, Molinero-Perez A, Moloney G, Morelli E, Morillas E, O'Connor R, Cruz-Pereira JS, Peterson VL, Rea K, Ritz NL, Sherwin E, Spichak S, Teichman EM, van de Wouw M, Ventura-Silva AP, Wallace-Fitzsimons SE, Hyland N, Clarke G, Dinan TG. The Microbiota-Gut-Brain Axis. Physiol Rev 2019; 99:1877-2013. [PMID: 31460832 DOI: 10.1152/physrev.00018.2018] [Citation(s) in RCA: 2665] [Impact Index Per Article: 444.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The importance of the gut-brain axis in maintaining homeostasis has long been appreciated. However, the past 15 yr have seen the emergence of the microbiota (the trillions of microorganisms within and on our bodies) as one of the key regulators of gut-brain function and has led to the appreciation of the importance of a distinct microbiota-gut-brain axis. This axis is gaining ever more traction in fields investigating the biological and physiological basis of psychiatric, neurodevelopmental, age-related, and neurodegenerative disorders. The microbiota and the brain communicate with each other via various routes including the immune system, tryptophan metabolism, the vagus nerve and the enteric nervous system, involving microbial metabolites such as short-chain fatty acids, branched chain amino acids, and peptidoglycans. Many factors can influence microbiota composition in early life, including infection, mode of birth delivery, use of antibiotic medications, the nature of nutritional provision, environmental stressors, and host genetics. At the other extreme of life, microbial diversity diminishes with aging. Stress, in particular, can significantly impact the microbiota-gut-brain axis at all stages of life. Much recent work has implicated the gut microbiota in many conditions including autism, anxiety, obesity, schizophrenia, Parkinson’s disease, and Alzheimer’s disease. Animal models have been paramount in linking the regulation of fundamental neural processes, such as neurogenesis and myelination, to microbiome activation of microglia. Moreover, translational human studies are ongoing and will greatly enhance the field. Future studies will focus on understanding the mechanisms underlying the microbiota-gut-brain axis and attempt to elucidate microbial-based intervention and therapeutic strategies for neuropsychiatric disorders.
Collapse
Affiliation(s)
- John F. Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Kenneth J. O'Riordan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Caitlin S. M. Cowan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Kiran V. Sandhu
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Thomaz F. S. Bastiaanssen
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Marcus Boehme
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Martin G. Codagnone
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Sofia Cussotto
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Christine Fulling
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Anna V. Golubeva
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Katherine E. Guzzetta
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Minal Jaggar
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Caitriona M. Long-Smith
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Joshua M. Lyte
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Jason A. Martin
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Alicia Molinero-Perez
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Gerard Moloney
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Emanuela Morelli
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Enrique Morillas
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Rory O'Connor
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Joana S. Cruz-Pereira
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Veronica L. Peterson
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Kieran Rea
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Nathaniel L. Ritz
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Eoin Sherwin
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Simon Spichak
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Emily M. Teichman
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Marcel van de Wouw
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Ana Paula Ventura-Silva
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Shauna E. Wallace-Fitzsimons
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Niall Hyland
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Timothy G. Dinan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| |
Collapse
|
39
|
Knop J, van IJzendoorn MH, Bakermans-Kranenburg MJ, Joëls M, van der Veen R. The effects of different rearing conditions on sexual maturation and maternal care in heterozygous mineralocorticoid receptor knockout mice. Horm Behav 2019; 112:54-64. [PMID: 30953639 DOI: 10.1016/j.yhbeh.2019.04.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 03/14/2019] [Accepted: 04/01/2019] [Indexed: 12/21/2022]
Abstract
Sexual and social development is affected by a complex interplay between genetic makeup and the early-life rearing environment. While many rodent studies focused primarily on the detrimental effects of early-life stress, human literature suggests that genetic susceptibility may not be restricted to negative environments; it may also enhance the beneficial effects of positive rearing conditions. To examine this interaction in a controlled setting, heterozygous mineralocorticoid receptor knockout (MR+/-) mice and control litter mates were exposed to a limited nesting/bedding (LN, impoverished), standard nesting (SN, control) or communal nesting (CN, enriched) paradigm from postnatal day 2-9 (P2-P9). Offspring was monitored for puberty onset between P24-P36 and, in females, maternal care-giving (i.e. as F1) during adulthood, after which basal corticosterone was measured. Different home-cage environments resulted in profound differences in received maternal care and offspring body weight. In male offspring, LN resulted in delayed puberty onset that was mediated by body weight and unpredictability of maternal care received during early development. In female offspring, rearing condition did not significantly alter sexual maturation and had little effect on their own maternal care-giving behavior. Genotype did affect maternal care: female MR+/- offspring exhibited a less active nursing style and upregulated fragmentation during adulthood, irrespective of early life conditions. Basal corticosterone levels were highest in MR+/- mice with a background of LN. Overall, we found a gene-by-environment interaction with respect to basal corticosterone levels, but not for sexual maturation or maternal behavior.
Collapse
Affiliation(s)
- Jelle Knop
- Dept. Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; Faculty of Social and Behavioural Sciences, Leiden University, Leiden, the Netherlands
| | - Marinus H van IJzendoorn
- Dept. of Psychology, Education and Child Studies, Erasmus University Rotterdam, Rotterdam, the Netherlands; Primary Care Unit, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | | | - Marian Joëls
- Dept. Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Rixt van der Veen
- Dept. Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands; Faculty of Social and Behavioural Sciences, Leiden University, Leiden, the Netherlands.
| |
Collapse
|
40
|
Strzelewicz AR, Ordoñes Sanchez E, Rondón-Ortiz AN, Raneri A, Famularo ST, Bangasser DA, Kentner AC. Access to a high resource environment protects against accelerated maturation following early life stress: A translational animal model of high, medium and low security settings. Horm Behav 2019; 111:46-59. [PMID: 30708031 PMCID: PMC6527488 DOI: 10.1016/j.yhbeh.2019.01.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 12/18/2018] [Accepted: 01/10/2019] [Indexed: 12/21/2022]
Abstract
Early life exposure to a low security setting, characterized by a scarcity of resources and limited food access, increases the risk for psychiatric illness and metabolic dysfunction. We utilized a translational rat model to mimic a low security environment and determined how this manipulation affected offspring behavior, metabolism, and puberty. Because food insecurity in humans is associated with reduced access to healthy food options the "low security" rat manipulation combined a Western diet with exposure to a limited bedding and nesting manipulation (WD-LB). In this setting, dams were provided with limited nesting materials during the pups' early life (P2-P10). This manipulation was contrasted with standard rodent caging (SD) and environmental enrichment (EE), to model "medium security" and "high security" environments, respectively. To determine if transitioning from a low to high security environment improved outcomes, some juvenile WD-LB offspring were exposed to EE. Maternal care was impacted by these environments such that EE dams engaged in high quality care when on the nest, but spent less time on the nest than SD dams. Although WD-LB dams excessively chased their tails, they were very attentive to their pups, perhaps to compensate for limited resources. Offspring exposed to WD-LB only displayed subtle changes in behavior. However, WD-LB exposure resulted in significant metabolic dysfunction characterized by increased body weight, precocious puberty and alterations in the hypothalamic kisspeptin system. These negative effects of WD-LB on puberty and weight regulation were mitigated by EE exposure. Collectively, these studies suggest that both compensatory maternal care and juvenile enrichment can reduce the impact of a low security environment. Moreover, they highlight how utilizing diverse models of resource (in)stability can reveal mechanisms that confer vulnerability and resilience to early life stress.
Collapse
Affiliation(s)
- Arielle R Strzelewicz
- School of Pharmacy, Massachusetts College of Pharmacy and Health Sciences, Boston,MA 02115, United States
| | | | - Alejandro N Rondón-Ortiz
- School of Pharmacy, Massachusetts College of Pharmacy and Health Sciences, Boston,MA 02115, United States
| | - Anthony Raneri
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115, United States
| | - Sydney T Famularo
- Department of Psychology, Temple University, Philadelphia, PA 19122, United States
| | - Debra A Bangasser
- Department of Psychology, Temple University, Philadelphia, PA 19122, United States
| | - Amanda C Kentner
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115, United States.
| |
Collapse
|
41
|
Sear R, Sheppard P, Coall DA. Cross-cultural evidence does not support universal acceleration of puberty in father-absent households. Philos Trans R Soc Lond B Biol Sci 2019; 374:20180124. [PMID: 30966893 PMCID: PMC6460089 DOI: 10.1098/rstb.2018.0124] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/17/2019] [Indexed: 12/20/2022] Open
Abstract
Father absence in early life has been shown to be associated with accelerated reproductive development in girls. Evolutionary social scientists have proposed several adaptive hypotheses for this finding. Though there is variation in the detail of these hypotheses, they all assume that family environment in early life influences the development of life-history strategy, and, broadly, that early reproductive development is an adaptive response to father absence. Empirical evidence to support these hypotheses, however, has been derived from WEIRD (Western, Educated, Industrialized, Rich and Democratic) populations. Data from a much broader range of human societies are necessary in order to properly test adaptive hypotheses. Here, we review the empirical literature on father absence and puberty in both sexes, focusing on recent studies that have tested this association beyond the WEIRD world. We find that relationships between father absence and age at puberty are more varied in contexts beyond WEIRD societies, and when relationships beyond the father-daughter dyad are considered. This has implications for our understanding of how early-life environment is linked to life-history strategies, and for our understanding of pathways to adult health outcomes, given that early reproductive development may be linked to negative health outcomes in later life This article is part of the theme issue 'Developing differences: early-life effects and evolutionary medicine'.
Collapse
Affiliation(s)
- Rebecca Sear
- Population Health, London School of Hygiene and Tropical Medicine, London, UK
| | - Paula Sheppard
- Department of Sociology, University of Oxford, Oxford UK
| | - David A. Coall
- Medical and Health Sciences, Edith Cowan University, Joondalup, Australia
| |
Collapse
|
42
|
Cowan CSM, Stylianakis AA, Richardson R. Early-life stress, microbiota, and brain development: probiotics reverse the effects of maternal separation on neural circuits underpinning fear expression and extinction in infant rats. Dev Cogn Neurosci 2019; 37:100627. [PMID: 30981894 PMCID: PMC6969299 DOI: 10.1016/j.dcn.2019.100627] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 01/17/2019] [Accepted: 02/19/2019] [Indexed: 12/21/2022] Open
Abstract
Early-life stress has pervasive, typically detrimental, effects on physical and mental health across the lifespan. In rats, maternal-separation stress results in premature expression of an adult-like profile of fear regulation that predisposes stressed rats to persistent fear, one of the hallmarks of clinical anxiety. Probiotic treatment attenuates the effects of maternal separation on fear regulation. However, the neural pathways underlying these behavioral changes are unknown. Here, we examined the neural correlates of stress-induced alterations in fear behavior and their reversal by probiotic treatment. Male Sprague-Dawley rats were exposed to either standard rearing conditions or maternal-separation stress (postnatal days [P] 2–14). Some maternally-separated (MS) animals were also exposed to probiotics (Lactobacillus rhamnosus and L. helveticus) via the maternal drinking water during the period of stress. Using immunohistochemistry, we demonstrated that stressed rat pups prematurely exhibit adult-like engagement of the medial prefrontal cortex during fear regulation, an effect that can be prevented using a probiotic treatment. The present results add to the cross-species evidence that early adversity hastens maturation in emotion-related brain circuits. Importantly, our results also demonstrate that the precocious neural maturation in stressed infants is prevented by a non-invasive probiotic treatment.
Collapse
Affiliation(s)
- Caitlin S M Cowan
- School of Psychology, The University of New South Wales, Sydney, Australia.
| | | | - Rick Richardson
- School of Psychology, The University of New South Wales, Sydney, Australia
| |
Collapse
|
43
|
Childhood urbanicity and hair steroid hormone levels in ten-year-old children. Psychoneuroendocrinology 2019; 102:53-57. [PMID: 30513500 DOI: 10.1016/j.psyneuen.2018.11.039] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 11/27/2018] [Accepted: 11/28/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND Research suggests that it may be more stressful for children to grow up in an urban area than in a rural area. Urbanicity may affect physiological stress system functioning as well as the timing of sexual maturation. The purpose of the current study was to investigate whether moderate urbanicity (current and childhood, ranging from rural areas to small cities) was associated with indices of long-term hypothalamic-pituitary-adrenal and hypothalamic-pituitary-gonadal axis functioning (cortisol, cortisone, dehydroepiandrosterone and progesterone levels) and whether sex moderated any associations. METHOD Children (N = 92) were all 10 years old and from the Dutch general population. Hair samples were collected and single segments (the three cm most proximal to the scalp) were assayed for concentrations of steroid hormones (LCMS/MS method). Neighborhood-level urbanicity and socioeconomic status were measured from birth through age ten years. Analyses were controlled for neighborhood- and family socioeconomic status, body mass index and season of sampling. RESULTS The results from multivariate analyses of variance showed no associations between current or childhood moderate urbanicity and hair steroid hormone concentrations. Interaction terms between moderate urbanicity and sex were not statistically significant. CONCLUSIONS Associations between urbanicity and steroid hormone levels may only be detectable in highly urban areas and/or during later stages of adolescence. Alternatively, our findings may have been due to most children being from families with a higher socioeconomic status.
Collapse
|
44
|
Kentner AC, Cryan JF, Brummelte S. Resilience priming: Translational models for understanding resiliency and adaptation to early life adversity. Dev Psychobiol 2019; 61:350-375. [PMID: 30311210 PMCID: PMC6447439 DOI: 10.1002/dev.21775] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 06/22/2018] [Accepted: 07/10/2018] [Indexed: 12/20/2022]
Abstract
Despite the increasing attention to early life adversity and its long-term consequences on health, behavior, and the etiology of neurodevelopmental disorders, our understanding of the adaptations and interventions that promote resiliency and rescue against such insults are underexplored. Specifically, investigations of the perinatal period often focus on negative events/outcomes. In contrast, positive experiences (i.e. enrichment/parental care//healthy nutrition) favorably influence development of the nervous and endocrine systems. Moreover, some stressors result in adaptations and demonstrations of later-life resiliency. This review explores the underlying mechanisms of neuroplasticity that follow some of these early life experiences and translates them into ideas for interventions in pediatric settings. The emerging role of the gut microbiome in mediating stress susceptibility is also discussed. Since many negative outcomes of early experiences are known, it is time to identify mechanisms and mediators that promote resiliency against them. These range from enrichment, quality parental care, dietary interventions and those that target the gut microbiota.
Collapse
Affiliation(s)
- Amanda C. Kentner
- School of Arts & Sciences, Massachusetts College of Pharmacy and Health Sciences, 179 Longwood Ave, Boston, MA 02115,
| | - John F. Cryan
- Dept. Anatomy & Neuroscience & APC Microbiome Institute, University College Cork, College Rd., Cork, Ireland,
| | - Susanne Brummelte
- Department of Psychology, Wayne State University, 5057 Woodward Ave, Detroit, MI 48202,
| |
Collapse
|