1
|
Qing L, Qian X, Zhu H, Wang J, Sun J, Jin Z, Tang X, Zhao Y, Wang G, Zhao J, Chen W, Tian P. Maternal-infant probiotic transmission mitigates early-life stress-induced autism in mice. Gut Microbes 2025; 17:2456584. [PMID: 39931863 PMCID: PMC11817528 DOI: 10.1080/19490976.2025.2456584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 12/14/2024] [Accepted: 01/13/2025] [Indexed: 02/14/2025] Open
Abstract
Autism, a disorder influenced by both genetic and environmental factors, presents significant challenges for prevention and treatment. While maternal-infant gut microbiota has been a focus in autism research, preventive strategies targeting maternal gut microbiota remain underexplored. This study demonstrates that prenatal probiotic intake can effectively prevent maternal separation-induced autistic-like behaviors in offspring without altering the embryonic neurodevelopment in mice. Using specific PCR primers and cross-fostering experiments, we traced the vertical transmission of probiotics, primarily via fecal/vaginal contamination. Early probiotic colonization conferred resilience against stress-induced gut pathogenic microbes and Th17-mediated peripheral inflammation while significantly inhibiting hypermyelination and neuroinflammation linked to systemic inflammation. Microbial metabolites like tyrosol and xanthurenic acid alleviated neuroinflammation and hypermyelination in vitro, though the causal relationship among neuroinflammation, hypermyelination, and autism in vivo requires further validation. These findings underscore the importance of the maternal-infant microbiota transmission window in autism prevention and highlight the clinical potential of prenatal probiotic interventions.
Collapse
Affiliation(s)
- Li Qing
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Xin Qian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Huiyue Zhu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Jingyu Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Jingge Sun
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Zhiying Jin
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Xinyu Tang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Yingqi Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Gang Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, P. R. China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou, P. R. China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Peijun Tian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| |
Collapse
|
2
|
Teng T, Wu Q, Yin B, Zhang J, Li X, Zhang L, Zhou X, Xie P. Single-Nucleus Transcriptomics of the Nucleus Accumbens Reveals Cell-Type-Specific Dysregulation in Adolescent Macaques with Depressive-Like Behaviors. Neurosci Bull 2025:10.1007/s12264-025-01412-5. [PMID: 40399551 DOI: 10.1007/s12264-025-01412-5] [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: 09/04/2024] [Accepted: 01/08/2025] [Indexed: 05/23/2025] Open
Abstract
Adolescent depression is increasingly recognized as a serious mental health disorder with distinct clinical and molecular features. Using single-nucleus RNA sequencing, we identified cell-specific transcriptomic changes in the nucleus accumbens (NAc), particularly in astrocytes, of adolescent macaques exhibiting depressive-like behaviors. The level of diacylglycerol kinase beta was significantly reduced in neurons and glial cells of depressed macaques, while FKBP5 levels increased in glial cells. Disruption of GABAergic synapses and disruption of D-glutamine and D-glutamate metabolism were linked to depressive phenotypes in medium spiny neurons (MSNs) and subtypes of astrocytes. Communication pathways between astrocytes and D1/D2-MSNs were also disrupted, involving factors like bone morphogenetic protein-6 and Erb-B2 receptor tyrosine kinase-4. Bulk transcriptomic and proteomic analyses corroborated these findings, and FKBP5 upregulation was confirmed by qRT-PCR, western blotting, and immunofluorescence in the NAc of rats and macaques with chronic unpredictable mild stress. Our results highlight the specific roles of different cell types in adolescent depression in the NAc, offering potential targets for new antidepressant therapies.
Collapse
Affiliation(s)
- Teng Teng
- Department of Neurology, NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400014, China
- Department of Psychiatry, Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing, 400016, China
| | - Qingyuan Wu
- Department of Neurology, NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400014, China
- Department of Neurology, Chongqing University Three Gorges Hospital, Chongqing University, Wanzhou, 404000, China
| | - Bangmin Yin
- Department of Neurology, NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400014, China
- Department of Psychiatry, Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing, 400016, China
| | - Jushuang Zhang
- Department of Psychiatry, Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing, 400016, China
| | - Xuemei Li
- Department of Neurology, NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400014, China
- Department of Psychiatry, Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing, 400016, China
| | - Lige Zhang
- Department of Psychiatry, Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing, 400016, China
| | - Xinyu Zhou
- Department of Neurology, NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400014, China.
- Department of Psychiatry, Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing, 400016, China.
| | - Peng Xie
- Department of Neurology, NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400014, China.
| |
Collapse
|
3
|
Birnie MT, Baram TZ. The evolving neurobiology of early-life stress. Neuron 2025; 113:1474-1490. [PMID: 40101719 PMCID: PMC12097948 DOI: 10.1016/j.neuron.2025.02.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/31/2025] [Accepted: 02/14/2025] [Indexed: 03/20/2025]
Abstract
Because early-life stress is common and constitutes a strong risk factor for cognitive and mental health disorders, it has been the focus of a multitude of studies in humans and experimental models. Yet, we have an incomplete understanding of what is perceived as stressful by the developing brain, what aspects of stress influence brain maturation, what developmental ages are particularly vulnerable to stress, which molecules mediate the effects of stress on brain operations, and how transient stressful experiences can lead to enduring emotional and cognitive dysfunctions. Here, we discuss these themes, highlight the challenges and progress in resolving them, and propose new concepts and avenues for future research.
Collapse
Affiliation(s)
- Matthew T Birnie
- Department of Pediatrics, University of California, Irvine, Irvine, CA, USA
| | - Tallie Z Baram
- Department of Pediatrics, University of California, Irvine, Irvine, CA, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA, USA; Department of Neurology, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
4
|
Gómez-Ilescas A, Silveira PP. Early adversity and the comorbidity between metabolic disease and psychopathology. J Physiol 2025. [PMID: 40349327 DOI: 10.1113/jp285927] [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: 10/20/2024] [Accepted: 04/01/2025] [Indexed: 05/14/2025] Open
Abstract
Although the co-existence of metabolic and psychiatric disorders in the same individual (comorbidity) is very prevalent, the mechanisms by which these disorders co-occur are poorly understood, but a history of early-life adversity is a common developmental risk factor. Exposure to adverse environments during critical periods of development (e.g. fetal life and infancy) modifies the metabolism and the function of the brain persistently, influencing behaviours that contribute to both metabolic and mental health disarrangements over the life course. We will review molecular and clinical evidence supporting the notion that early adversity is an important risk factor for the comorbidity between metabolic and psychiatric conditions. We will also discuss the possible mechanisms involved: neurometabolic programming, epigenetic alterations and the cumulative effects of altered inflammatory and oxidative pathways linked to early adversity.
Collapse
Affiliation(s)
| | - Patricia Pelufo Silveira
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Research Centre, McGill University, Montreal, QC, Canada
- Department of Psychiatry, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| |
Collapse
|
5
|
Geiger LT, Balouek JAR, Barrett MR, Thompson JM, Fang LZ, Farrelly LA, Chen AS, Tang M, Bennett SN, Garcia BA, Maze I, Creed MC, Peña CJ. Early-life stress alters chromatin modifications in VTA to prime stress sensitivity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.03.14.584631. [PMID: 38559030 PMCID: PMC10980038 DOI: 10.1101/2024.03.14.584631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Early-life stress increases sensitivity to subsequent stress, which has been observed at behavioral, neural activity, and gene expression levels. However, the molecular mechanisms underlying such long-lasting sensitivity are poorly understood. We tested the hypothesis that persistent changes in transcription and transcriptional potential were maintained at the level of the epigenome, through changes in chromatin. We used a combination of bottom-up mass spectrometry, viral-mediated epigenome-editing, RNA-sequencing, patch clamp electrophysiology of dopamine neurons, and behavioral quantification in a mouse model of early-life stress, focusing on the ventral tegmental area (VTA), a dopaminergic brain region critically implicated in motivation, reward learning, stress response, and mood and drug disorders. We found that early-life stress alters histone dynamics in VTA, including enrichment of histone-3 lysine-4 monomethylation - associated with open chromatin and primed or active enhancers - and the H3K4 monomethylase Setd7. Mimicking early-life stress through postnatal overexpression of Setd7 and enrichment of H3K4me1 in VTA sensitizes transcriptional, physiological, and behavioral response to adult stress. These findings link early-life stress experience to long-term stress hypersensitivity within the brain's dopaminergic circuitry, providing a mechanism by which early-life stress increases risk for mood and anxiety disorders later in life.
Collapse
|
6
|
Chen X, Kim Y, Kawaguchi D. Development of the rodent prefrontal cortex: circuit formation, plasticity, and impacts of early life stress. Front Neural Circuits 2025; 19:1568610. [PMID: 40206866 PMCID: PMC11979153 DOI: 10.3389/fncir.2025.1568610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 03/11/2025] [Indexed: 04/11/2025] Open
Abstract
The prefrontal cortex (PFC), located at the anterior region of the cerebral cortex, is a multimodal association cortex essential for higher-order brain functions, including decision-making, attentional control, memory processing, and regulation of social behavior. Structural, circuit-level, and functional abnormalities in the PFC are often associated with neurodevelopmental disorders. Here, we review recent findings on the postnatal development of the PFC, with a particular emphasis on rodent studies, to elucidate how its structural and circuit properties are established during critical developmental windows and how these processes influence adult behaviors. Recent evidence also highlights the lasting effects of early life stress on the PFC structure, connectivity, and function. We explore potential mechanisms underlying these stress-induced alterations, with a focus on epigenetic regulation and its implications for PFC maturation and neurodevelopmental disorders. By integrating these insights, this review provides an overview of the developmental processes shaping the PFC and their implications for brain health and disease.
Collapse
Affiliation(s)
| | | | - Daichi Kawaguchi
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
7
|
Vasquez LS, Stack S, Taylor WW, Dias BG. Intergenerational Effects of Stress - A Focus on Learning and Memory. Curr Top Behav Neurosci 2025. [PMID: 40119217 DOI: 10.1007/7854_2025_578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2025]
Abstract
Stress is a ubiquitous facet of life. Ranging in form (e.g., psychosocial, physical, nutritional, economic) and longevity (e.g., acute, chronic), stressors affect the biology of those directly in their line of attack. As is becoming increasingly appreciated, the pernicious effects of stress echo across generations (Dias et al. 2015; Yehuda and Lehrner 2018; Jawaid et al. 2021; Dion et al. 2022; Zhou and Ryan 2023; Dias 2024). With a focus on learning and memory, this chapter addresses how stressors derail learning and memory in the generation directly exposed to them andin future generations. To do so, with a specific emphasis on associative fear conditioning in humans and rodents, we touch upon the relevance of extinction training in the aftermath of such conditioning and the recall of such extinction training as windows into normative and disrupted learning. Next, we briefly discuss underlying neuroanatomical substrates mediating these processes. We then draw attention to influences of postnatal, in utero, and pre-conceptional stress on learning and memory across generations. Finally, we briefly outline biological factors that underlie how learning and memory is derailed by these stressors.
Collapse
Affiliation(s)
- L S Vasquez
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
- Developmental Neuroscience and Neurogenetics Program, The Saban Research Institute, Los Angeles, CA, USA
| | - S Stack
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
- Developmental Neuroscience and Neurogenetics Program, The Saban Research Institute, Los Angeles, CA, USA
| | - W W Taylor
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
- Developmental Neuroscience and Neurogenetics Program, The Saban Research Institute, Los Angeles, CA, USA
| | - B G Dias
- Developmental Neuroscience and Neurogenetics Program, The Saban Research Institute, Los Angeles, CA, USA.
- Division of Endocrinology, Diabetes and Metabolism, Children's Hospital Los Angeles, Los Angeles, CA, USA.
- Department of Pediatrics, Keck School of Medicine of USC, Los Angeles, CA, USA.
| |
Collapse
|
8
|
Saad HA, Marzouk M, Abdelrahman H, Moradikor N. Mechanisms underlying stress effects on the brain: Basic concepts and clinical implications. PROGRESS IN BRAIN RESEARCH 2025; 291:21-47. [PMID: 40222781 DOI: 10.1016/bs.pbr.2025.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
Chronic stress impacts the brain through complex physiological, neurological, and immunological responses. The stress response involves the activation of the sympathetic-adrenal-medullary (SAM) system and the hypothalamic-pituitary-adrenal (HPA) axis, releasing stress hormones like norepinephrine and cortisol. While these responses are adaptive short-term, chronic stress disrupts homeostasis, increasing the risk of cardiovascular diseases, neurodegenerative disorders, and psychiatric conditions such as depression. This dysregulation is linked to persistent neuroinflammation, oxidative stress, and neurotransmitter imbalances involving dopamine and serotonin, impairing neuroplasticity and leading to structural changes in critical brain areas, such as the hippocampus and prefrontal cortex. Moreover, stress affects gene expression, particularly neuroinflammatory pathways, contributing to long-term cognitive function and emotional regulation alterations. Advancements in neuroimaging and molecular techniques, including MRI, PET, and SPECT, hold promise for identifying biomarkers and better understanding stress-induced brain changes. These insights are critical for developing targeted interventions to mitigate the adverse effects of chronic stress on brain health.
Collapse
Affiliation(s)
- Hager Adel Saad
- Faculty of Pharmacy and Biotechnology, German University in Cairo, (GUC), New Cairo, Cairo, Egypt.
| | - Mahmoud Marzouk
- Faculty of Pharmacy and Biotechnology, German University in Cairo, (GUC), New Cairo, Cairo, Egypt
| | - Hla Abdelrahman
- Faculty of Pharmacy and Biotechnology, German University in Cairo, (GUC), New Cairo, Cairo, Egypt
| | - Nasrollah Moradikor
- International Center for Neuroscience Research, Institute for Intelligent Research, Tbilisi, Georgia
| |
Collapse
|
9
|
Orso R, Viola TW, Heberle BA, Creutzberg KC, Lumertz FS, Grassi-Oliveira R. Sex-Specific Effects of Early-Life Stress Exposure on Memory Performance and the Medial Prefrontal Cortex Transcriptomic Pattern in Adolescent Mice. Mol Neurobiol 2025:10.1007/s12035-025-04803-x. [PMID: 40038196 DOI: 10.1007/s12035-025-04803-x] [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: 08/03/2024] [Accepted: 02/22/2025] [Indexed: 03/06/2025]
Abstract
Early life stress (ELS) is considered a risk factor for the development of cognitive and executive dysfunctions throughout development. The medial portion of the prefrontal cortex (mPFC) is directly implicated in short-term working memory. Furthermore, due to its late development compared to other brain regions, the mPFC is considered a vulnerable brain region to ELS exposure. Here, we investigated the effects of the ELS on PFC-dependent memory and mPFC transcriptomic profiles. From postnatal day (PND) 2 to PND 15, BALB/cJ mice were exposed to maternal separation (MS) for 3 h per day combined with limited bedding (ELS group) or left undisturbed (CT group). During the period of stress, maternal behavior was recorded pre-MS and post-MS. From PND 45 to PND 47, males and females were tested for working memory performance in the Y-maze and short-term recognition memory in the object in place task (OIP). Later, we assessed mRNA level alterations in the mPFC by RNA-seq. Here, we showed that ELS increases maternal care post-MS and the number of nest exits pre-MS and post-MS. Furthermore, males and females exposed to ELS exhibited impairments in the OIP, while only females performed worse in the Y-maze. With respect to the mPFC transcriptome, we identified 13 DEGs in the females, which were significantly influenced by chaperone-mediated protein folding processes, while 4 genes were altered in males. In conclusion, we showed that, compared with male sex, ELS alters maternal behavior and leads to more extensive impairments in memory function and transcriptomic alterations in females.
Collapse
Affiliation(s)
- Rodrigo Orso
- Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, USA
| | - Thiago Wendt Viola
- School of Medicine, Pontifical Catholic University of Rio Grande Do Sul, Porto Alegre, Brazil
| | | | | | | | - Rodrigo Grassi-Oliveira
- School of Medicine, Pontifical Catholic University of Rio Grande Do Sul, Porto Alegre, Brazil.
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 11, A701-129, 8200, Aarhus, Denmark.
| |
Collapse
|
10
|
Peña CJ. Early-life stress sensitizes response to future stress: Evidence and mechanisms. Neurobiol Stress 2025; 35:100716. [PMID: 40134543 PMCID: PMC11932861 DOI: 10.1016/j.ynstr.2025.100716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/12/2025] [Accepted: 03/07/2025] [Indexed: 03/27/2025] Open
Abstract
Early-life stress sensitizes individuals to additional stressors and increases lifetime risk for mood and anxiety disorders. Research in both human populations and rodent models of early-life stress have sought to determine how different types of stressors contribute to vulnerability, and whether there are developmental sensitive periods for such effects. Although differences in the type and timing of rodent early-life stress paradigms have led to differences in specific behavioral outcomes, this complexity is present among humans as well. Robust rodent research now shows how early-life stress increases sensitivity to future stressors at behavioral, neural circuit, and molecular levels. These recent discoveries are laying the foundation for translation to more effective interventions relevant for those who experienced childhood stress and trauma.
Collapse
Affiliation(s)
- Catherine Jensen Peña
- Princeton Neuroscience Institute, Princeton University, 40 Woodlands Way, Princeton, NJ, 08544, USA
| |
Collapse
|
11
|
Price KM, Polter AM. Interactions of sex and stress in modulation of ventral tegmental area dopaminergic activity. Curr Opin Behav Sci 2025; 61:101477. [PMID: 40364819 PMCID: PMC12068853 DOI: 10.1016/j.cobeha.2024.101477] [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] [Indexed: 05/15/2025]
Abstract
Dopaminergic (DA) neurons of the ventral tegmental area (VTA) have long been studied for their role in reward prediction and goal-directed behaviors. However, appreciation is growing for a complementary role of VTA DA neurons in responding to aversive stimuli and as critical substrates for behavioral sequelae of stressful experiences. As is the case across neuroscience, the majority of our knowledge about VTA DA neurons comes from studies in male subjects. Recent years have seen an increase in inclusion of female subjects and exploration of sex differences. There is now an emerging body of literature showing that although there are minimal basal structural and functional sex differences in VTA DA neurons, experience-dependent changes in these neurons can differ significantly between males and females. Here, we discuss potential implications of sex differences in VTA function and review recent data on sex differences and similarities of DA neurons at baseline and following stress.
Collapse
Affiliation(s)
- Kailyn M. Price
- Department of Pharmacology and Physiology, George Washington University School of Medicine and Health Sciences, Washington, DC 20037
| | - Abigail M. Polter
- Department of Pharmacology and Physiology, George Washington University School of Medicine and Health Sciences, Washington, DC 20037
| |
Collapse
|
12
|
Parise LF, Joseph Burnett C, Russo SJ. Early life stress and altered social behaviors: A perspective across species. Neurosci Res 2025; 211:65-74. [PMID: 37992997 PMCID: PMC11102940 DOI: 10.1016/j.neures.2023.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 07/21/2023] [Accepted: 11/04/2023] [Indexed: 11/24/2023]
Abstract
Childhood and adolescent affiliations guide how individuals engage in social relationships throughout their lifetime and adverse experiences can promote biological alterations that facilitate behavioral maladaptation. Indeed, childhood victims of abuse are more likely to be diagnosed with conduct or mood disorders which are both characterized by altered social engagement. A key domain particularly deserving of attention is aggressive behavior, a hallmark of many disorders characterized by deficits in reward processing. Animal models have been integral in identifying both the short- and long-term consequences of stress exposure and suggest that whether it is disruption to parental care or social isolation, chronic exposure to early life stress increases corticosterone, changes the expression of neurotransmitters and neuromodulators, and facilitates structural alterations to the hypothalamus, hippocampus, and amygdala, influencing how these brain regions communicate with other reward-related substrates. Herein, we describe how adverse early life experiences influence social behavioral outcomes across a wide range of species and highlight the long-term biological mechanisms that are most relevant to maladaptive aggressive behavior.
Collapse
Affiliation(s)
- Lyonna F Parise
- Icahn School of Medicine, Nash Family Department of Neuroscience and Friedman Brain Institute, New York, NY, USA.
| | - C Joseph Burnett
- Icahn School of Medicine, Nash Family Department of Neuroscience and Friedman Brain Institute, New York, NY, USA
| | - Scott J Russo
- Icahn School of Medicine, Nash Family Department of Neuroscience and Friedman Brain Institute, New York, NY, USA.
| |
Collapse
|
13
|
Liao C, O’Farrell E, Qalieh Y, Savalia NK, Girgenti MJ, Kwan KY, Kwan AC. Single-nucleus transcriptomics reveals time-dependent and cell-type-specific effects of psilocybin on gene expression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.04.631335. [PMID: 39803502 PMCID: PMC11722411 DOI: 10.1101/2025.01.04.631335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
There is growing interest to investigate classic psychedelics as potential therapeutics for mental illnesses. Previous studies have demonstrated that one dose of psilocybin leads to persisting neural and behavioral changes. The durability of psilocybin's effects suggests that there are likely alterations of gene expression at the transcriptional level. In this study, we performed single-nucleus RNA sequencing of the dorsal medial frontal cortex of male and female mice. Samples were collected at 1, 2, 4, 24, or 72 hours after psilocybin or ketamine administration and from control animals. At baseline, major excitatory and GABAergic cell types selectively express particular serotonin receptor transcripts. The psilocybin-evoked differentially expressed genes in excitatory neurons were involved in synaptic plasticity, which were distinct from genes enriched in GABAergic neurons that contribute to mitochondrial function and cellular metabolism. The effect of psilocybin on gene expression was time-dependent, including an early phase at 1-2 hours followed by a late phase at 72 hours of transcriptional response after administration. Ketamine administration produced transcriptional changes that show a high degree of correlation to those induced by psilocybin. Collectively, the results reveal that psilocybin produces time-dependent and cell-type specific changes in gene expression in the medial frontal cortex, which may underpin the drug's long-term effects on neural circuits and behavior.
Collapse
Affiliation(s)
- Clara Liao
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
- Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Ethan O’Farrell
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yaman Qalieh
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Neil K. Savalia
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
- Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT 06511, USA
- Medical Scientist Training Program, Yale University School of Medicine, New Haven, Connecticut, 06511, USA
| | - Matthew J. Girgenti
- Department of Psychiatry, Yale School of Medicine, New Haven, CT 06511, USA
- Clinical Neuroscience Division, National Center for Posttraumatic Stress Disorder, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Kenneth Y. Kwan
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Alex C. Kwan
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
14
|
Cuarenta A. Retrotransposons and the brain: Exploring a complex relationship between mobile elements, stress, and neurological health. Neurobiol Stress 2025; 34:100709. [PMID: 39927173 PMCID: PMC11803260 DOI: 10.1016/j.ynstr.2025.100709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/14/2024] [Accepted: 01/13/2025] [Indexed: 02/11/2025] Open
Abstract
Environmental experiences during early life, including stress, can significantly impact brain development and behavior. Early life stress (ELS) is linked to an increased risk for various psychiatric disorders including anxiety, depression, and substance use disorders. Epigenetic mechanisms have increasingly been of interest to understand how environmental factors contribute to reprogramming the brain and alter risk and resilience to developing psychiatric disorders. However, we know very little about mobile elements or the regulation of mobile elements and their contribution to psychiatric disorders. Recently, advances in genomics have contributed to our understanding of mobile elements, including the retrotransposon LINE-1 (L1) and their potential role in mediating environmental experiences. Yet we still do not understand how these elements may contribute to psychiatric disorders. Future research leveraging cutting-edge technologies will deepen our understanding of these mobile elements. By elucidating their role in development and how stress may impact them, we may unlock new avenues for therapeutic and diagnostic innovations.
Collapse
Affiliation(s)
- Amelia Cuarenta
- Neuroscience Institute and the Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA, USA
| |
Collapse
|
15
|
Mancini C, Babicola L, Chila G, Di Segni M, Municchi D, D’Addario SL, Spoleti E, Passeri A, Cifani C, Andolina D, Cabib S, Ferlazzo F, Iosa M, Rossi R, Di Lorenzo G, Renzi M, Ventura R. Secure attachment to caregiver prevents adult depressive symptoms in a sex-dependent manner: A translational study. iScience 2024; 27:111328. [PMID: 39758994 PMCID: PMC11700650 DOI: 10.1016/j.isci.2024.111328] [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: 07/29/2024] [Revised: 09/13/2024] [Accepted: 11/04/2024] [Indexed: 01/07/2025] Open
Abstract
Although clinically relevant, evidence for a protective effect of early secure attachment against the development of depressive symptoms in adulthood is still inconsistent. This study used a translational approach to overcome this limitation. The analysis of a non-clinical adult population revealed a moderating effect of secure attachment on depressive symptoms in women only. Thus, we tested the causal link between early attachment with caregiver and adult depressive-like phenotypes in a mouse model of early adversities that is especially effective in females. Repeated cross fostering (RCF) in the first postnatal days prevented the development of pups' secure attachment with the caregiver as tested in a rodent version of the "strange situation"-the standard human test-induced depressive-like behaviors and altered activity of the ventral tegmental area dopamine neurons in adulthood. Finally, a stable alternative caregiver during the RCF experience prevented all these effects, modeling human "earned attachment."
Collapse
Affiliation(s)
- Camilla Mancini
- University of Camerino, School of Pharmacy, Pharmacology Unit, Camerino, Italy
| | | | - Gilda Chila
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Matteo Di Segni
- Child Psychopathology Unit, Scientific Institute IRCCS Eugenio Medea, Bosisio Parini, Italy
| | - Diana Municchi
- Department of Psychology, Sapienza University, Rome, Italy
| | | | - Elena Spoleti
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Alice Passeri
- Department of Psychology, Sapienza University, Rome, Italy
| | - Carlo Cifani
- University of Camerino, School of Pharmacy, Pharmacology Unit, Camerino, Italy
| | - Diego Andolina
- IRCCS Santa Lucia Foundation, Rome, Italy
- Department of Psychology, Sapienza University, Rome, Italy
| | - Simona Cabib
- IRCCS Santa Lucia Foundation, Rome, Italy
- Department of Psychology, Sapienza University, Rome, Italy
| | - Fabio Ferlazzo
- Department of Psychology, Sapienza University, Rome, Italy
| | - Marco Iosa
- IRCCS Santa Lucia Foundation, Rome, Italy
- Department of Psychology, Sapienza University, Rome, Italy
| | - Rodolfo Rossi
- Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Giorgio Di Lorenzo
- Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Massimiliano Renzi
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Rossella Ventura
- Department of Psychology, Sapienza University, Rome, Italy
- IRCCS San Raffaele, Rome, Italy
| |
Collapse
|
16
|
Hagenauer MH, Sannah Y, Hebda-Bauer EK, Rhoads C, O'Connor AM, Flandreau E, Watson SJ, Akil H. Resource: A curated database of brain-related functional gene sets (Brain.GMT). MethodsX 2024; 13:102788. [PMID: 39049932 PMCID: PMC11267058 DOI: 10.1016/j.mex.2024.102788] [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: 04/17/2024] [Accepted: 05/31/2024] [Indexed: 07/27/2024] Open
Abstract
Transcriptional profiling has become a common tool for investigating the nervous system. During analysis, differential expression results are often compared to functional ontology databases, which contain curated gene sets representing well-studied pathways. This dependence can cause neuroscience studies to be interpreted in terms of functional pathways documented in better studied tissues (e.g., liver) and topics (e.g., cancer), and systematically emphasizes well-studied genes, leaving other findings in the obscurity of the brain "ignorome". To address this issue, we compiled a curated database of 918 gene sets related to nervous system function, tissue, and cell types ("Brain.GMT") that can be used within common analysis pipelines (GSEA, limma, edgeR) to interpret results from three species (rat, mouse, human). Brain.GMT includes brain-related gene sets curated from the Molecular Signatures Database (MSigDB) and extracted from public databases (GeneWeaver, Gemma, DropViz, BrainInABlender, HippoSeq) and published studies containing differential expression results. Although Brain.GMT is still undergoing development and currently only represents a fraction of available brain gene sets, "brain ignorome" genes are already better represented than in traditional Gene Ontology databases. Moreover, Brain.GMT substantially improves the quantity and quality of gene sets identified as enriched with differential expression in neuroscience studies, enhancing interpretation. •We compiled a curated database of 918 gene sets related to nervous system function, tissue, and cell types ("Brain.GMT").•Brain.GMT can be used within common analysis pipelines (GSEA, limma, edgeR) to interpret neuroscience transcriptional profiling results from three species (rat, mouse, human).•Although Brain.GMT is still undergoing development, it substantially improved the interpretation of differential expression results within our initial use cases.
Collapse
Affiliation(s)
- Megan H. Hagenauer
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yusra Sannah
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Cosette Rhoads
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, USA
- National Institutes of Health, Bethesda, MD 20892, USA
| | - Angela M. O'Connor
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Stanley J. Watson
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Huda Akil
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
17
|
Duan TQ, Hagenauer MH, Flandreau EI, Bader A, Nguyen DM, Maras PM, Merscher S De Lima R, Gyles T, Mclain C, Meaney MJ, Nestler EJ, Watson SJ, Akil H. A meta-analysis of the effects of early life stress on the prefrontal cortex transcriptome suggests long-term effects on myelin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.22.624315. [PMID: 39605735 PMCID: PMC11601536 DOI: 10.1101/2024.11.22.624315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Background Early life stress (ELS) refers to exposure to negative childhood experiences, such as neglect, disaster, and physical, mental, or emotional abuse. ELS can permanently alter the brain, leading to cognitive impairment, increased sensitivity to future stressors, and mental health risks. The prefrontal cortex (PFC) is a key brain region implicated in the effects of ELS. Methods To better understand the effects of ELS on the PFC, we ran a meta-analysis of publicly available transcriptional profiling datasets. We identified five datasets (GSE89692, GSE116416, GSE14720, GSE153043, GSE124387) that characterized the long-term effects of multi-day postnatal ELS paradigms (maternal separation, limited nesting/bedding) in male and female laboratory rodents (rats, mice). The outcome variable was gene expression in the PFC later in adulthood as measured by microarray or RNA-Seq. To conduct the meta-analysis, preprocessed gene expression data were extracted from the Gemma database. Following quality control, the final sample size was n=89: n=42 controls & n=47 ELS: GSE116416 n=23 (no outliers); GSE116416 n=44 (2 outliers); GSE14720 n=7 (no outliers); GSE153043 n=9 (1 outlier), and GSE124387 n=6 (no outliers). Differential expression was calculated using the limma pipeline followed by an empirical Bayes correction. For each gene, a random effects meta-analysis model was then fit to the ELS vs. Control effect sizes (Log2 Fold Changes) from each study. Results Our meta-analysis yielded stable estimates for 11,885 genes, identifying five genes with differential expression following ELS (false discovery rate< 0.05): transforming growth factor alpha ( Tgfa ), IQ motif containing GTPase activating protein 3 ( Iqgap3 ), collagen, type XI, alpha 1 ( Col11a1 ), claudin 11 ( Cldn11 ) and myelin associated glycoprotein ( Mag ), all of which were downregulated. Broadly, gene sets associated with oligodendrocyte differentiation, myelination, and brain development were downregulated following ELS. In contrast, genes previously shown to be upregulated in Major Depressive Disorder patients were upregulated following ELS. Conclusion These findings suggest that ELS during critical periods of development may produce long-term effects on the efficiency of transmission in the PFC and drive changes in gene expression similar to those underlying depression.
Collapse
|
18
|
Ding R, Tang Y, Cao G, Mai Y, Fu Y, Ren Z, Li W, Hou J, Sun S, Chen B, Han X, He Z, Ye JH, Zhou L, Fu R. Lateral habenula IL-10 controls GABA A receptor trafficking and modulates depression susceptibility after maternal separation. Brain Behav Immun 2024; 122:122-136. [PMID: 39128573 DOI: 10.1016/j.bbi.2024.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 07/26/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024] Open
Abstract
Maternal separation (MS), a form of early life adversity, increases the risk of psychiatric disorders in adulthood by intricately linking cytokines and mood-regulating brain circuits. The Lateral Habenula (LHb) encodes aversive experiences, contributes to negative moods, and is pivotal in depression development. However, the precise impact of MS on LHb cytokine signaling and synaptic plasticity remains unclear. We reported that adolescent MS offspring mice displayed susceptibility to depression behavioral phylotypes, with neuronal hyperactivity and an imbalance in pro-inflammatory and anti-inflammatory cytokines in the LHb. Moreover, the decreased IL-10 level negatively correlated with depressive-like behaviors in susceptible mice. Functionally, LHb IL-10 overexpression restored decreased levels of PI3K, phosphorylated AKT (pAKT), gephyrin, and membrane GABAA receptor proteins while reducing abnormally elevated GSK3β and Fos expression, rescuing the MS-induced depression. Conversely, LHb neuronal IL-10 receptor knockdown in naive mice increased Fos expression and elicited depression-like symptoms, potentially through impaired membrane GABAA receptor trafficking by suppressing the PI3K/pAKT/gephyrin cascades. Hence, this work establishes a mechanism by which MS promotes susceptibility to adolescent depression by impeding the critical role of IL-10 signaling on neuronal GABAA receptor function.
Collapse
Affiliation(s)
- Ruxuan Ding
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong 518106, PR China
| | - Ying Tang
- Basic and Clinical Medicine Teaching Laboratory, School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong 518100, PR China
| | - Guoxin Cao
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong 518106, PR China
| | - Yunlin Mai
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong 518106, PR China
| | - Yixin Fu
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong 518106, PR China
| | - Zhiheng Ren
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong 518106, PR China
| | - Wenfu Li
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong 518106, PR China
| | - Jiawei Hou
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong 518106, PR China
| | - Shizhu Sun
- Basic and Clinical Medicine Teaching Laboratory, School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong 518100, PR China
| | - Bingqing Chen
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong 518106, PR China
| | - Xiaojiao Han
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong 518106, PR China
| | - Zelei He
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong 518106, PR China
| | - Jiang-Hong Ye
- Department of Anesthesiology, Pharmacology, Physiology & Neuroscience, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, NJ 07103, USA.
| | - Lihua Zhou
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong 518106, PR China.
| | - Rao Fu
- Department of Anatomy, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, Guangdong 518106, PR China; Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen 518106, PR China.
| |
Collapse
|
19
|
Baram TZ, Birnie MT. Enduring memory consequences of early-life stress / adversity: Structural, synaptic, molecular and epigenetic mechanisms. Neurobiol Stress 2024; 33:100669. [PMID: 39309367 PMCID: PMC11415888 DOI: 10.1016/j.ynstr.2024.100669] [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: 03/19/2024] [Revised: 08/13/2024] [Accepted: 08/28/2024] [Indexed: 09/25/2024] Open
Abstract
Adverse early life experiences are strongly associated with reduced cognitive function throughout life. The link is strong in many human studies, but these do not enable assigning causality, and the limited access to the live human brain can impede establishing the mechanisms by which early-life adversity (ELA) may induce cognitive problems. In experimental models, artificially imposed chronic ELA/stress results in deficits in hippocampus dependent memory as well as increased vulnerability to the deleterious effects of adult stress on memory. This causal relation of ELA and life-long memory impairments provides a framework to probe the mechanisms by which ELA may lead to human cognitive problems. Here we focus on the consequences of a one-week exposure to adversity during early postnatal life in the rodent, the spectrum of the ensuing memory deficits, and the mechanisms responsible. We highlight molecular, cellular and circuit mechanisms using convergent trans-disciplinary approaches aiming to enable translation of the discoveries in experimental models to the clinic.
Collapse
Affiliation(s)
- Tallie Z. Baram
- Department of Pediatrics, University of California-Irvine, Irvine, CA, USA
- Department of Anatomy/Neurobiology, University of California-Irvine, Irvine, CA, USA
- Department of Neurology, University of California-Irvine, Irvine, CA, USA
| | - Matthew T. Birnie
- Department of Pediatrics, University of California-Irvine, Irvine, CA, USA
| |
Collapse
|
20
|
Rayan NA, Aow J, Lim MGL, Arcego DM, Ryan R, Nourbakhsh N, de Lima RMS, Craig K, Zhang TY, Goh YT, Sun AX, Tompkins T, Bronner S, Binda S, Diorio J, Parent C, Meaney MJ, Prabhakar S. Shared and unique transcriptomic signatures of antidepressant and probiotics action in the mammalian brain. Mol Psychiatry 2024; 29:3653-3668. [PMID: 38844534 DOI: 10.1038/s41380-024-02619-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 05/13/2024] [Accepted: 05/16/2024] [Indexed: 11/08/2024]
Abstract
Understanding the shared and divergent mechanisms across antidepressant (AD) classes and probiotics is critical for improving treatment for mood disorders. Here we examine the transcriptomic effects of bupropion (NDRI), desipramine (SNRI), fluoxetine (SSRI) and a probiotic formulation (Lacidofil®) on 10 regions across the mammalian brain. These treatments massively alter gene expression (on average, 2211 differentially expressed genes (DEGs) per region-treatment combination), highlighting the biological complexity of AD and probiotic action. Intersection of DEG sets against neuropsychiatric GWAS loci, sex-specific transcriptomic portraits of major depressive disorder (MDD), and mouse models of stress and depression reveals significant similarities and differences across treatments. Interestingly, molecular responses in the infralimbic cortex, basolateral amygdala and locus coeruleus are region-specific and highly similar across treatments, whilst responses in the Raphe, medial preoptic area, cingulate cortex, prelimbic cortex and ventral dentate gyrus are predominantly treatment-specific. Mechanistically, ADs concordantly downregulate immune pathways in the amygdala and ventral dentate gyrus. In contrast, protein synthesis, metabolism and synaptic signaling pathways are axes of variability among treatments. We use spatial transcriptomics to further delineate layer-specific molecular pathways and DEGs within the prefrontal cortex. Our study reveals complex AD and probiotics action on the mammalian brain and identifies treatment-specific cellular processes and gene targets associated with mood disorders.
Collapse
Affiliation(s)
- Nirmala Arul Rayan
- Genome Institute of Singapore, Agency for Science Technology and Research (A*STAR), Singapore, 138672, Singapore
| | - Jonathan Aow
- Genome Institute of Singapore, Agency for Science Technology and Research (A*STAR), Singapore, 138672, Singapore
- NUS Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Michelle Gek Liang Lim
- Genome Institute of Singapore, Agency for Science Technology and Research (A*STAR), Singapore, 138672, Singapore
| | - Danusa Mar Arcego
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, QC, H4H 1R3, Canada
| | - Richard Ryan
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, QC, H4H 1R3, Canada
| | - Nooshin Nourbakhsh
- Genome Institute of Singapore, Agency for Science Technology and Research (A*STAR), Singapore, 138672, Singapore
| | | | - Kelly Craig
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, QC, H4H 1R3, Canada
| | - Tie Yuan Zhang
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, QC, H4H 1R3, Canada
| | - Yeek Teck Goh
- Genome Institute of Singapore, Agency for Science Technology and Research (A*STAR), Singapore, 138672, Singapore
| | - Alfred Xuyang Sun
- Duke-NUS Graduate Medical School, Signature Research Program in Neuroscience and Behavioural Disorders, 8 College Road, Singapore, 169857, Singapore
| | - Thomas Tompkins
- Lallemand Bio-Ingredients, 1620 Rue Prefontaine, Montréal, QC, H1W 2N8, Canada
| | - Stéphane Bronner
- Lallemand Health Solutions, Rosell Institute for Microbiome and Probiotics, 6100 Avenue Royalmount, Montréal, QC, H4P 2R2, Canada
| | - Sylvie Binda
- Lallemand Health Solutions, Rosell Institute for Microbiome and Probiotics, 6100 Avenue Royalmount, Montréal, QC, H4P 2R2, Canada
| | - Josie Diorio
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, QC, H4H 1R3, Canada
| | - Carine Parent
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, QC, H4H 1R3, Canada
| | - Michael J Meaney
- NUS Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, QC, H4H 1R3, Canada.
- Singapore Institute for Clinical Sciences, A*STAR, Singapore, 117609, Singapore.
- Brain-Body Initiative, Institute for Cell & Molecular Biology, A*STAR, Singapore, Singapore.
| | - Shyam Prabhakar
- Genome Institute of Singapore, Agency for Science Technology and Research (A*STAR), Singapore, 138672, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore.
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore.
| |
Collapse
|
21
|
Torres-Berrío A, Estill M, Patel V, Ramakrishnan A, Kronman H, Minier-Toribio A, Issler O, Browne CJ, Parise EM, van der Zee YY, Walker DM, Martínez-Rivera FJ, Lardner CK, Durand-de Cuttoli R, Russo SJ, Shen L, Sidoli S, Nestler EJ. Mono-methylation of lysine 27 at histone 3 confers lifelong susceptibility to stress. Neuron 2024; 112:2973-2989.e10. [PMID: 38959894 PMCID: PMC11377169 DOI: 10.1016/j.neuron.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 02/05/2024] [Accepted: 06/07/2024] [Indexed: 07/05/2024]
Abstract
Histone post-translational modifications are critical for mediating persistent alterations in gene expression. By combining unbiased proteomics profiling and genome-wide approaches, we uncovered a role for mono-methylation of lysine 27 at histone H3 (H3K27me1) in the enduring effects of stress. Specifically, mice susceptible to early life stress (ELS) or chronic social defeat stress (CSDS) displayed increased H3K27me1 enrichment in the nucleus accumbens (NAc), a key brain-reward region. Stress-induced H3K27me1 accumulation occurred at genes that control neuronal excitability and was mediated by the VEFS domain of SUZ12, a core subunit of the polycomb repressive complex-2, which controls H3K27 methylation patterns. Viral VEFS expression changed the transcriptional profile of the NAc, led to social, emotional, and cognitive abnormalities, and altered excitability and synaptic transmission of NAc D1-medium spiny neurons. Together, we describe a novel function of H3K27me1 in the brain and demonstrate its role as a "chromatin scar" that mediates lifelong stress susceptibility.
Collapse
Affiliation(s)
- Angélica Torres-Berrío
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Lurie Center for Autism, Massachusetts General Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
| | - Molly Estill
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Vishwendra Patel
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Aarthi Ramakrishnan
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hope Kronman
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Angélica Minier-Toribio
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Orna Issler
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Caleb J Browne
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eric M Parise
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yentl Y van der Zee
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Deena M Walker
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, USA
| | - Freddyson J Martínez-Rivera
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Casey K Lardner
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Romain Durand-de Cuttoli
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Scott J Russo
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Li Shen
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Simone Sidoli
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, NY, USA
| | - Eric J Nestler
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
22
|
Speranza L, Filiz KD, Lippiello P, Ferraro MG, Pascarella S, Miniaci MC, Volpicelli F. Enduring Neurobiological Consequences of Early-Life Stress: Insights from Rodent Behavioral Paradigms. Biomedicines 2024; 12:1978. [PMID: 39335492 PMCID: PMC11429222 DOI: 10.3390/biomedicines12091978] [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: 06/17/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024] Open
Abstract
Stress profoundly affects physical and mental health, particularly when experienced early in life. Early-life stress (ELS) encompasses adverse childhood experiences such as abuse, neglect, violence, or chronic poverty. These stressors can induce long-lasting changes in brain structure and function, impacting areas involved in emotion regulation, cognition, and stress response. Consequently, individuals exposed to high levels of ELS are at an increased risk for mental health disorders like depression, anxiety, and post-traumatic stress disorders, as well as physical health issues, including metabolic disorders, cardiovascular disease, and cancer. This review explores the biological and psychological consequences of early-life adversity paradigms in rodents, such as maternal separation or deprivation and limited bedding or nesting. The study of these experimental models have revealed that the organism's response to ELS is complex, involving genetic and epigenetic mechanisms, and is associated with the dysregulation of physiological systems like the nervous, neuroendocrine, and immune systems, in a sex-dependent fashion. Understanding the impact of ELS is crucial for developing effective interventions and preventive strategies in humans exposed to stressful or traumatic experiences in childhood.
Collapse
Affiliation(s)
- Luisa Speranza
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy; (L.S.); (K.D.F.); (P.L.); (S.P.)
| | - Kardelen Dalim Filiz
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy; (L.S.); (K.D.F.); (P.L.); (S.P.)
| | - Pellegrino Lippiello
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy; (L.S.); (K.D.F.); (P.L.); (S.P.)
| | - Maria Grazia Ferraro
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy;
| | - Silvia Pascarella
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy; (L.S.); (K.D.F.); (P.L.); (S.P.)
| | - Maria Concetta Miniaci
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy; (L.S.); (K.D.F.); (P.L.); (S.P.)
| | - Floriana Volpicelli
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy; (L.S.); (K.D.F.); (P.L.); (S.P.)
| |
Collapse
|
23
|
Cattaneo A, Begni V, Zonca V, Riva MA. Early life adversities, psychopathologies and novel pharmacological strategies. Pharmacol Ther 2024; 260:108686. [PMID: 38969307 DOI: 10.1016/j.pharmthera.2024.108686] [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: 02/23/2024] [Revised: 06/05/2024] [Accepted: 07/02/2024] [Indexed: 07/07/2024]
Abstract
Exposure to adversities during early life stages (early life adversities - ELA), ranging from pregnancy to adolescence, represents a major risk factor for the vulnerability to mental disorders. Hence, it is important to understand the molecular and functional underpinning of such relationship, in order to develop strategies aimed at reducing the psychopathologic burden associated with ELA, which may eventually lead to a significant improvement in clinical practice. In this review, we will initially recapitulate clinical and preclinical evidence supporting the link between ELA and psychopathology and we will primarily discuss the main biological mechanisms that have been described as potential mediators of the effects of ELA on the psychopathologic risk, including the role for genetic factors as well as sex differences. The knowledge emerging from these studies may be instrumental for the development of novel therapeutic strategies aimed not only at correcting the deficits that emerge from ELA exposure, but also in preventing the manifestation of a full-blown psychopathologic condition. With this respect, we will specifically focus on adolescence as a key time frame for disease onset as well as for early therapeutic intervention. We believe that incorporating clinical and preclinical research data in the context of early life adversities can be instrumental to elucidate the mechanisms contributing to the risk for psychopathology or that may promote resilience. This will ultimately allow the identification of 'at risk' individuals who may benefit from specific forms of interventions that, by interfering with disease trajectories, could result in more benign clinical outcomes.
Collapse
Affiliation(s)
- Annamaria Cattaneo
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milan, Italy; Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Veronica Begni
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milan, Italy
| | - Valentina Zonca
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milan, Italy
| | - Marco A Riva
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milan, Italy; Biological Psychiatry Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.
| |
Collapse
|
24
|
Choi MK, Cook A, Mungikar K, Eachus H, Tochwin A, Linke M, Gerber S, Ryu S. Exposure to elevated glucocorticoid during development primes altered transcriptional responses to acute stress in adulthood. iScience 2024; 27:110160. [PMID: 38989456 PMCID: PMC11233911 DOI: 10.1016/j.isci.2024.110160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 05/02/2024] [Accepted: 05/29/2024] [Indexed: 07/12/2024] Open
Abstract
Early life stress (ELS) is a major risk factor for developing psychiatric disorders, with glucocorticoids (GCs) implicated in mediating its effects in shaping adult phenotypes. In this process, exposure to high levels of developmental GC (hdGC) is thought to induce molecular changes that prime differential adult responses. However, identities of molecules targeted by hdGC exposure are not completely known. Here, we describe lifelong molecular consequences of hdGC exposure using a newly developed zebrafish double-hit stress model, which shows altered behaviors and stress hypersensitivity in adulthood. We identify a set of primed genes displaying altered expression only upon acute stress in hdGC-exposed adult fish brains. Interestingly, this gene set is enriched in risk factors for psychiatric disorders in humans. Lastly, we identify altered epigenetic regulatory elements following hdGC exposure. Thus, our study provides comprehensive datasets delineating potential molecular targets mediating the impact of hdGC exposure on adult responses.
Collapse
Affiliation(s)
- Min-Kyeung Choi
- Living Systems Institute & Department of Clinical and Biomedical Sciences, University of Exeter, Stocker Road, EX4 4QD Exeter, UK
| | - Alexander Cook
- Institute of Human Genetics, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Kanak Mungikar
- Institute of Human Genetics, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Helen Eachus
- Living Systems Institute & Department of Clinical and Biomedical Sciences, University of Exeter, Stocker Road, EX4 4QD Exeter, UK
| | - Anna Tochwin
- Living Systems Institute & Department of Clinical and Biomedical Sciences, University of Exeter, Stocker Road, EX4 4QD Exeter, UK
| | - Matthias Linke
- Institute of Human Genetics, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Susanne Gerber
- Institute of Human Genetics, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Soojin Ryu
- Living Systems Institute & Department of Clinical and Biomedical Sciences, University of Exeter, Stocker Road, EX4 4QD Exeter, UK
| |
Collapse
|
25
|
Nestler EJ, Russo SJ. Neurobiological basis of stress resilience. Neuron 2024; 112:1911-1929. [PMID: 38795707 PMCID: PMC11189737 DOI: 10.1016/j.neuron.2024.05.001] [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: 01/02/2024] [Revised: 03/21/2024] [Accepted: 05/01/2024] [Indexed: 05/28/2024]
Abstract
A majority of humans faced with severe stress maintain normal physiological and behavioral function, a process referred to as resilience. Such stress resilience has been modeled in laboratory animals and, over the past 15 years, has transformed our understanding of stress responses and how to approach the treatment of human stress disorders such as depression, post-traumatic stress disorder (PTSD), and anxiety disorders. Work in rodents has demonstrated that resilience to chronic stress is an active process that involves much more than simply avoiding the deleterious effects of the stress. Rather, resilience is mediated largely by the induction of adaptations that are associated uniquely with resilience. Such mechanisms of natural resilience in rodents are being characterized at the molecular, cellular, and circuit levels, with an increasing number being validated in human investigations. Such discoveries raise the novel possibility that treatments for human stress disorders, in addition to being geared toward reversing the damaging effects of stress, can also be based on inducing mechanisms of natural resilience in individuals who are inherently more susceptible. This review provides a progress report on this evolving field.
Collapse
Affiliation(s)
- Eric J Nestler
- Nash Family Department of Neuroscience and Department of Psychiatry, The Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Scott J Russo
- Nash Family Department of Neuroscience and Department of Psychiatry, The Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
26
|
Weng J, Zhu YY, Liao LY, Yang XT, Dong YH, Meng WD, Sun DJ, Liu Y, Peng WZ, Jiang Y. Distinct epigenetic modulation of differentially expressed genes in the adult mouse brain following prenatal exposure to low-dose bisphenol A. Cell Biol Toxicol 2024; 40:37. [PMID: 38777957 PMCID: PMC11111541 DOI: 10.1007/s10565-024-09875-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 05/15/2024] [Indexed: 05/25/2024]
Abstract
Bisphenol A (BPA) is a common component in the manufacture of daily plastic consumer goods. Recent studies have suggested that prenatal exposure to BPA can increase the susceptibility of offspring to mental illness, although the underlying mechanisms remain unclear. In this study, we performed transcriptomic and epigenomic profiling in the adult mouse brain following prenatal exposure to low-dose BPA. We observed a sex-specific transcriptional dysregulation in the cortex, with more significant differentially expressed genes was observed in adult cortex from male offspring. Moreover, the upregulated genes primarily influenced neuronal functions, while the downregulated genes were significantly associated with energy metabolism pathways. More evidence supporting impaired mitochondrial function included a decreased ATP level and a reduced number of mitochondria in the cortical neuron of the BPA group. We further investigated the higher-order chromatin regulatory patterns of DEGs by incorporating published Hi-C data. Interestingly, we found that upregulated genes exhibited more distal interactions with multiple enhancers, while downregulated genes displayed relatively short-range interactions among adjacent genes. Our data further revealed decreased H3K9me3 signal on the distal enhancers of upregulated genes, whereas increased DNA methylation and H3K27me3 signals on the promoters of downregulated genes. In summary, our study provides compelling evidence for the potential health risks associated with prenatal exposure to BPA, and uncovers sex-specific transcriptional changes with a complex interplay of multiple epigenetic mechanisms.
Collapse
Affiliation(s)
- Jie Weng
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Yue-Yan Zhu
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Li-Yong Liao
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Xin-Tong Yang
- Shanghai Medical college, Fudan University, Shanghai, 200032, China
| | - Yu-Hao Dong
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Wei-da Meng
- The MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Dai-Jing Sun
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Yun Liu
- The MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Wen-Zhu Peng
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Yan Jiang
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
27
|
Hagenauer MH, Sannah Y, Hebda-Bauer EK, Rhoads C, O'Connor AM, Watson SJ, Akil H. Resource: A Curated Database of Brain-Related Functional Gene Sets (Brain.GMT). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.05.588301. [PMID: 38645214 PMCID: PMC11030436 DOI: 10.1101/2024.04.05.588301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Transcriptional profiling has become a common tool for investigating the nervous system. During analysis, differential expression results are often compared to functional ontology databases, which contain curated gene sets representing well-studied pathways. This dependence can cause neuroscience studies to be interpreted in terms of functional pathways documented in better studied tissues (e.g., liver) and topics (e.g., cancer), and systematically emphasizes well-studied genes, leaving other findings in the obscurity of the brain "ignorome". To address this issue, we compiled a curated database of 918 gene sets related to nervous system function, tissue, and cell types ("Brain.GMT") that can be used within common analysis pipelines (GSEA, limma, edgeR) to interpret results from three species (rat, mouse, human). Brain.GMT includes brain-related gene sets curated from the Molecular Signatures Database (MSigDB) and extracted from public databases (GeneWeaver, Gemma, DropViz, BrainInABlender, HippoSeq) and published studies containing differential expression results. Although Brain.GMT is still undergoing development and currently only represents a fraction of available brain gene sets, "brain ignorome" genes are already better represented than in traditional Gene Ontology databases. Moreover, Brain.GMT substantially improves the quantity and quality of gene sets identified as enriched with differential expression in neuroscience studies, enhancing interpretation.
Collapse
Affiliation(s)
- Megan H Hagenauer
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor; MI 48109, USA
| | - Yusra Sannah
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor; MI 48109, USA
| | - Elaine K Hebda-Bauer
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor; MI 48109, USA
| | - Cosette Rhoads
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor; MI 48109, USA
- National Institutes of Health, Bethesda, MD 20892, USA
| | - Angela M O'Connor
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor; MI 48109, USA
| | - Stanley J Watson
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor; MI 48109, USA
| | - Huda Akil
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor; MI 48109, USA
| |
Collapse
|
28
|
Zhang Y, Jiang M, Wang S, Xiang X, He W, Du J, Hei M. Effect of family integrated care on stress in mothers of preterm infants: A multicenter cluster randomized controlled trial. J Affect Disord 2024; 350:304-312. [PMID: 38232775 DOI: 10.1016/j.jad.2024.01.102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 11/23/2023] [Accepted: 01/09/2024] [Indexed: 01/19/2024]
Abstract
BACKGROUND Reducing mother-infant separation in early life is a key breakthrough in the care improvement model in the neonatal intensive care unit (NICU). Previously, we reported effect of family integrated care (FICare) on clinical outcomes of preterm infants. We further clarify effect of FICare on maternal stress. METHODS Mothers of preterm infants at eleven NICUs were randomized to the FICare group and the control group. The primary outcome was the reduction in Parental Stress Scale: NICU (PSS:NICU) score from enrollment to discharge. RESULTS Total of 601 mothers (298 in FICare and 303 in control groups) enrolled. There was no significant difference in PSS:NICU score between the 2 groups at enrollment (P = 0.824), and the FICare group had lower scores at discharge (P < 0.001). PSS:NICU scores of both groups were significantly decreased at discharge compared to at enrollment (P < 0.001), and the reduction was greater in the FICare group (P < 0.001). After applying linear regressions to adjust for potential confounders, results remained unchanged (adjusted P < 0.001). PSS:NICU score reductions from enrollment to discharge were positively correlated with maternal age in the control group (ρ = 0.147, P = 0.011). LIMITATIONS This study was limited to post-hoc analyses and did not include follow-up to evaluate long-term effects. CONCLUSIONS FICare is helpful for reducing maternal stress in preterm infants in the NICU. Older mothers tend to have limited improvements in stress after traditional nonparent care, which suggests that they may benefit more from the FICare model.
Collapse
Affiliation(s)
- Yuan Zhang
- Neonatal Center, Beijing Children's Hospital, Capital Medical University, Beijing, China; National Center for Children's Health, Beijing, China
| | - Min Jiang
- Neonatal Center, Beijing Children's Hospital, Capital Medical University, Beijing, China; National Center for Children's Health, Beijing, China
| | - Shu Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xiying Xiang
- Neonatal Center, Beijing Children's Hospital, Capital Medical University, Beijing, China; National Center for Children's Health, Beijing, China
| | - Wenwen He
- Neonatal Center, Beijing Children's Hospital, Capital Medical University, Beijing, China; National Center for Children's Health, Beijing, China
| | - Juan Du
- Neonatal Center, Beijing Children's Hospital, Capital Medical University, Beijing, China; National Center for Children's Health, Beijing, China
| | - Mingyan Hei
- Neonatal Center, Beijing Children's Hospital, Capital Medical University, Beijing, China; National Center for Children's Health, Beijing, China.
| |
Collapse
|
29
|
Bennett SN, Chang AB, Rogers FD, Jones P, Peña CJ. Thyroid hormones mediate the impact of early-life stress on ventral tegmental area gene expression and behavior. Horm Behav 2024; 159:105472. [PMID: 38141539 PMCID: PMC10922504 DOI: 10.1016/j.yhbeh.2023.105472] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 11/17/2023] [Accepted: 12/12/2023] [Indexed: 12/25/2023]
Abstract
Proper thyroid function is essential to the developing brain, including dopamine neuron differentiation, growth, and maintenance. Stress across the lifespan impacts thyroid hormone signaling and anxiety disorders and depression have been associated with thyroid dysfunction (both hypo- and hyper-active). However, less is known about how stress during postnatal development impacts thyroid function and related brain development. Our previous work in mice demonstrated that early-life stress (ELS) transiently impinged on expression of a transcription factor in dopamine neurons, Otx2, shown to be regulated by thyroid hormones. We hypothesized that thyroid hormone signaling may link experience of ELS with transcriptional dysregulation within the dopaminergic midbrain, and ultimately behavior. Here, we find that ELS transiently increases thyroid-stimulating hormone levels (inversely related to thyroid signaling) in both male and female mice at P21, an effect which recovers by adolescence. We next tested whether transient treatment of ELS mice with synthetic thyroid hormone (levothyroxine, LT4) could ameliorate the impact of ELS on sensitivity to future stress, and on expression of genes related to dopamine neuron development and maintenance, thyroid signaling, and plasticity within the ventral tegmental area. Among male mice, but not females, juvenile LT4 treatment prevented hypersensitivity to adult stress. We also found that rescuing developmental deficits in thyroid hormone signaling after ELS restored levels of some genes altered directly by ELS, and prevented alterations in expression of other genes sensitive to the second hit of adult stress. These findings suggest that thyroid signaling mediates the deleterious impact of ELS on VTA development, and that temporary treatment of hypothyroidism after ELS may be sufficient to prevent future stress hypersensitivity.
Collapse
Affiliation(s)
| | - Austin B Chang
- Princeton Neuroscience Institute, Princeton University, USA
| | - Forrest D Rogers
- Princeton Neuroscience Institute, Princeton University, USA; Department of Molecular Biology, Princeton University, USA
| | - Parker Jones
- Princeton Neuroscience Institute, Princeton University, USA
| | | |
Collapse
|
30
|
Kuhlman KR. Pitfalls and potential: Translating the two-hit model of early life stress from pre-clinical non-human experiments to human samples. Brain Behav Immun Health 2024; 35:100711. [PMID: 38169793 PMCID: PMC10758720 DOI: 10.1016/j.bbih.2023.100711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/08/2023] [Accepted: 12/04/2023] [Indexed: 01/05/2024] Open
Abstract
Exposure to early life stress (ELS) has been linked to at least double the risk of psychopathology as well as higher morbidity and earlier mortality across the lifespan. For this reason, the field of developmental psychopathology has spent decades identifying factors that explain which individuals are at risk for negative health outcomes. Preclinical experiments in this field commonly test the "two-hit hypothesis", which explores how ELS potentiates vulnerability to pathogenic physiological and behavioral outcomes when an individual is exposed to a stressor later in development. Yet, translation of the two-hit hypothesis to humans is conceptually and practically challenging, thus impeding progress in the field. This review summarizes the two-hit hypothesis used in preclinical experiments as it pertains to two putative pathways linking ELS to psychopathology: the innate immune and neuroendocrine systems. This review also identifies important considerations when translating this model to humans and provides several recommendations. Specifically, attention to the "biological salience" of different forms of ELA and the concordance of that salience with later probes of the system are needed. Further, the consequences of ELS may be context-specific rather than ubiquitous, at least among young people. Within this conceptualization, "second hits" may be best operationalized using standardized acute challenges to the innate immune and neuroendocrine systems (e.g., psychosocial stress). Third, more explicit reporting of sex differences in the human literature is needed. Finally, preclinical experimental designs that more accurately reflect the natural occurrence of ELS in community samples will more effectively advance the understanding of developmental mechanisms that occur as a consequence of ELS.
Collapse
Affiliation(s)
- Kate Ryan Kuhlman
- Department of Psychological Science, School of Social Ecology, University of California Irvine, USA
- Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, USA
| |
Collapse
|
31
|
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
|
32
|
Gyles TM, Nestler EJ, Parise EM. Advancing preclinical chronic stress models to promote therapeutic discovery for human stress disorders. Neuropsychopharmacology 2024; 49:215-226. [PMID: 37349475 PMCID: PMC10700361 DOI: 10.1038/s41386-023-01625-0] [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: 03/31/2023] [Revised: 05/08/2023] [Accepted: 05/19/2023] [Indexed: 06/24/2023]
Abstract
There is an urgent need to develop more effective treatments for stress-related illnesses, which include depression, post-traumatic stress disorder, and anxiety. We view animal models as playing an essential role in this effort, but to date, such approaches have generally not succeeded in developing therapeutics with new mechanisms of action. This is partly due to the complexity of the brain and its disorders, but also to inherent difficulties in modeling human disorders in rodents and to the incorrect use of animal models: namely, trying to recapitulate a human syndrome in a rodent which is likely not possible as opposed to using animals to understand underlying mechanisms and evaluating potential therapeutic paths. Recent transcriptomic research has established the ability of several different chronic stress procedures in rodents to recapitulate large portions of the molecular pathology seen in postmortem brain tissue of individuals with depression. These findings provide crucial validation for the clear relevance of rodent stress models to better understand the pathophysiology of human stress disorders and help guide therapeutic discovery. In this review, we first discuss the current limitations of preclinical chronic stress models as well as traditional behavioral phenotyping approaches. We then explore opportunities to dramatically enhance the translational use of rodent stress models through the application of new experimental technologies. The goal of this review is to promote the synthesis of these novel approaches in rodents with human cell-based approaches and ultimately with early-phase proof-of-concept studies in humans to develop more effective treatments for human stress disorders.
Collapse
Affiliation(s)
- Trevonn M Gyles
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Eric J Nestler
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Eric M Parise
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
33
|
Seah C, Signer R, Deans M, Bader H, Rusielewicz T, Hicks EM, Young H, Cote A, Townsley K, Xu C, Hunter CJ, McCarthy B, Goldberg J, Dobariya S, Holtzherimer PE, Young KA, Noggle SA, Krystal JH, Paull D, Girgenti MJ, Yehuda R, Brennand KJ, Huckins LM. Common genetic variation impacts stress response in the brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.27.573459. [PMID: 38234801 PMCID: PMC10793429 DOI: 10.1101/2023.12.27.573459] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
To explain why individuals exposed to identical stressors experience divergent clinical outcomes, we determine how molecular encoding of stress modifies genetic risk for brain disorders. Analysis of post-mortem brain (n=304) revealed 8557 stress-interactive expression quantitative trait loci (eQTLs) that dysregulate expression of 915 eGenes in response to stress, and lie in stress-related transcription factor binding sites. Response to stress is robust across experimental paradigms: up to 50% of stress-interactive eGenes validate in glucocorticoid treated hiPSC-derived neurons (n=39 donors). Stress-interactive eGenes show brain region- and cell type-specificity, and, in post-mortem brain, implicate glial and endothelial mechanisms. Stress dysregulates long-term expression of disorder risk genes in a genotype-dependent manner; stress-interactive transcriptomic imputation uncovered 139 novel genes conferring brain disorder risk only in the context of traumatic stress. Molecular stress-encoding explains individualized responses to traumatic stress; incorporating trauma into genomic studies of brain disorders is likely to improve diagnosis, prognosis, and drug discovery.
Collapse
|
34
|
Faraji J, Bettenson D, Yong VW, Metz GAS. Early life stress aggravates disease pathogenesis in mice with experimental autoimmune encephalomyelitis: Support for a two-hit hypothesis of multiple sclerosis etiology. J Neuroimmunol 2023; 385:578240. [PMID: 37951203 DOI: 10.1016/j.jneuroim.2023.578240] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/27/2023] [Accepted: 11/05/2023] [Indexed: 11/13/2023]
Abstract
Vision problems are one of the earliest diagnosed symptoms of multiple sclerosis (MS). The onset and progression of vision loss and the underlying pathogenesis in MS may be influenced by cumulative psychophysiological stress. Here, we used a two-hit model of stress in female mice to determine if early life stress (ELS, the first hit) influences the response to an immunization that induces experimental autoimmune encephalomyelitis (EAE, the second hit) later in life. We hypothesized that ELS caused by animal transportation from a vendor during early postnatal development represents a co-factor which can exacerbate the clinical severity of EAE. Indeed, adult EAE mice with a history of ELS displayed more severe clinical signs and delayed recovery compared to non-stressed EAE mice. ELS also diminished visual acuity measured by optokinetic responses, as well as locomotion and exploratory behaviours in EAE mice. Notably, ELS accelerated vision loss and caused earlier onset of visual impairments in EAE. Exacerbated functional impairments in stressed EAE mice were highly correlated with circulating corticosterone levels. The findings show that the progression of induced EAE in adulthood can be significantly impacted by adverse early life experiences. These observations emphasize the importance of comprehensive behavioural testing, including non-motor functions, to enhance the translational value of preclinical animal models of MS. Moreover, shipment stress of laboratory animals should be considered a necessary variable in preclinical MS research. The consideration of cumulative lifetime stresses provides a new perspective of MS pathogenesis within a personalized medicine framework.
Collapse
Affiliation(s)
- Jamshid Faraji
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Lethbridge, Alberta T1K 3M4, Canada.
| | - Dennis Bettenson
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Lethbridge, Alberta T1K 3M4, Canada
| | - V Wee Yong
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Gerlinde A S Metz
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Lethbridge, Alberta T1K 3M4, Canada; Southern Alberta Genome Sciences Centre, University of Lethbridge, Lethbridge, Alberta T1K 3M4, Canada.
| |
Collapse
|
35
|
Parel ST, Bennett SN, Cheng CJ, Timmermans OC, Fiori LM, Turecki G, Peña CJ. Transcriptional signatures of early-life stress and antidepressant treatment efficacy. Proc Natl Acad Sci U S A 2023; 120:e2305776120. [PMID: 38011563 DOI: 10.1073/pnas.2305776120] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 07/10/2023] [Indexed: 11/29/2023] Open
Abstract
Individuals with a history of early-life stress (ELS) tend to have an altered course of depression and lower treatment response rates. Research suggests that ELS alters brain development, but the molecular changes in the brain following ELS that may mediate altered antidepressant response have not been systematically studied. Sex and gender also impact the risk of depression and treatment response. Here, we leveraged existing RNA sequencing datasets from 1) blood samples from depressed female- and male-identifying patients treated with escitalopram or desvenlafaxine and assessed for treatment response or failure; 2) the nucleus accumbens (NAc) of female and male mice exposed to ELS and/or adult stress; and 3) the NAc of mice after adult stress, antidepressant treatment with imipramine or ketamine, and assessed for treatment response or failure. We find that transcriptomic signatures of adult stress after a history of ELS correspond with transcriptomic signatures of treatment nonresponse, across species and multiple classes of antidepressants. Transcriptomic correspondence with treatment outcome was stronger among females and weaker among males. We next pharmacologically tested these predictions in our mouse model of early-life and adult social defeat stress and treatment with either chronic escitalopram or acute ketamine. Among female mice, the strongest predictor of behavior was an interaction between ELS and ketamine treatment. Among males, however, early experience and treatment were poor predictors of behavior, mirroring our bioinformatic predictions. These studies provide neurobiological evidence for molecular adaptations in the brain related to sex and ELS that contribute to antidepressant treatment response.
Collapse
Affiliation(s)
- Sero Toriano Parel
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544
| | - Shannon N Bennett
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544
| | - Cindy J Cheng
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544
| | | | - Laura M Fiori
- Douglas Institute, Department of Psychiatry, McGill University, Montreal, QC H4H 1R3, Canada
| | - Gustavo Turecki
- Douglas Institute, Department of Psychiatry, McGill University, Montreal, QC H4H 1R3, Canada
| | | |
Collapse
|
36
|
Kos A, Lopez JP, Bordes J, de Donno C, Dine J, Brivio E, Karamihalev S, Luecken MD, Almeida-Correa S, Gasperoni S, Dick A, Miranda L, Büttner M, Stoffel R, Flachskamm C, Theis FJ, Schmidt MV, Chen A. Early life adversity shapes social subordination and cell type-specific transcriptomic patterning in the ventral hippocampus. SCIENCE ADVANCES 2023; 9:eadj3793. [PMID: 38039370 PMCID: PMC10691768 DOI: 10.1126/sciadv.adj3793] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 11/01/2023] [Indexed: 12/03/2023]
Abstract
Adverse events in early life can modulate the response to additional stressors later in life and increase the risk of developing psychiatric disorders. The underlying molecular mechanisms responsible for these effects remain unclear. Here, we uncover that early life adversity (ELA) in mice leads to social subordination. Using single-cell RNA sequencing (scRNA-seq), we identified cell type-specific changes in the transcriptional state of glutamatergic and GABAergic neurons in the ventral hippocampus of ELA mice after exposure to acute social stress in adulthood. These findings were reflected by an alteration in excitatory and inhibitory synaptic transmission induced by ELA in response to acute social stress. Finally, enhancing the inhibitory network function through transient diazepam treatment during an early developmental sensitive period reversed the ELA-induced social subordination. Collectively, this study significantly advances our understanding of the molecular, physiological, and behavioral alterations induced by ELA, uncovering a previously unknown cell type-specific vulnerability to ELA.
Collapse
Affiliation(s)
- Aron Kos
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Juan Pablo Lopez
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Joeri Bordes
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, Munich, Germany
| | - Carlo de Donno
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Department of Mathematics, Technische Universität München, Munich, Germany
| | - Julien Dine
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Elena Brivio
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), Munich, Germany
| | - Stoyo Karamihalev
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), Munich, Germany
| | - Malte D. Luecken
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Lung Health and Immunity, Helmholtz Munich, Munich, Germany
| | | | - Serena Gasperoni
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Alec Dick
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Lucas Miranda
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), Munich, Germany
- Department of Statistical Genetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Maren Büttner
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Department of Mathematics, Technische Universität München, Munich, Germany
| | - Rainer Stoffel
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Cornelia Flachskamm
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Fabian J. Theis
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Department of Mathematics, Technische Universität München, Munich, Germany
| | - Mathias V. Schmidt
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, Munich, Germany
| | - Alon Chen
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
37
|
Ye L, Wu J, Liu Z, Deng D, Bai S, Yang L, Xuan Y, Liu Z, Shi Y, Liu Z, Zhang R, Zhao J. Si-Ni-San alleviates early life stress-induced depression-like behaviors in adolescence via modulating Rac1 activity and associated spine plasticity in the nucleus accumbens. Front Pharmacol 2023; 14:1274121. [PMID: 38026979 PMCID: PMC10646421 DOI: 10.3389/fphar.2023.1274121] [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: 08/07/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Background: Early life stress (ELS) is a major risk factor for depression in adolescents. The nucleus accumbens (NAc) is a key center of the reward system, and spine remodeling in the NAc contributes to the development of depression. The Si-Ni-San formula (SNS) is a fundamental prescription for treating depression in traditional Chinese medicine. However, little is known about the effects of SNS on behavioral abnormalities and spine plasticity in the NAc induced by ELS. Purpose: This study aimed to investigate the therapeutic effect and the modulatory mechanism of SNS on abnormal behaviors and spine plasticity in the NAc caused by ELS. Methods: We utilized a model of ELS that involved maternal separation with early weaning to explore the protective effects of SNS on adolescent depression. Depressive-like behaviors were evaluated by the sucrose preference test, the tail suspension test, and the forced swimming test; anxiety-like behaviors were monitored by the open field test and the elevated plus maze. A laser scanning confocal microscope was used to analyze dendritic spine remodeling in the NAc. The activity of Rac1 was detected by pull-down and Western blot tests. Viral-mediated gene transfer of Rac1 was used to investigate its role in ELS-induced depression-like behaviors in adolescence. Results: ELS induced depression-like behaviors but not anxiety-like behaviors in adolescent mice, accompanied by an increase in stubby spine density, a decrease in mushroom spine density, and decreased Rac1 activity in the NAc. Overexpression of constitutively active Rac1 in the NAc reversed depression-related behaviors, leading to a decrease in stubby spine density and an increase in mushroom spine density. Moreover, SNS attenuated depression-like behavior in adolescent mice and counteracted the spine abnormalities in the NAc induced by ELS. Additionally, SNS increased NAc Rac1 activity, and the inhibition of Rac1 activity weakened the antidepressant effect of SNS. Conclusion: These results suggest that SNS may exert its antidepressant effects by modulating Rac1 activity and associated spine plasticity in the NAc.
Collapse
Affiliation(s)
- Lihong Ye
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiayi Wu
- School of Fundamental Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zuyi Liu
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Di Deng
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shasha Bai
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lei Yang
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yao Xuan
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zehao Liu
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yafei Shi
- School of Fundamental Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhongqiu Liu
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Rong Zhang
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jinlan Zhao
- Guangdong Provincial Key Laboratory of Translational Cancer Research of Chinese Medicines, Joint International Research Laboratory of Translational Cancer Research of Chinese Medicines, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
38
|
Wang F, Yang X, Ren Z, Chen C, Liu C. Alternative splicing in mouse brains affected by psychological stress is enriched in the signaling, neural transmission and blood-brain barrier pathways. Mol Psychiatry 2023; 28:4707-4718. [PMID: 37217679 DOI: 10.1038/s41380-023-02103-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 04/26/2023] [Accepted: 05/03/2023] [Indexed: 05/24/2023]
Abstract
Psychological stress increases the risk of major psychiatric disorders. Psychological stress on mice was reported to induce differential gene expression (DEG) in mice brain regions. Alternative splicing is a fundamental aspect of gene expression and has been associated with psychiatric disorders but has not been investigated in the stressed brain yet. This study investigated changes in gene expression and splicing under psychological stress, the related pathways, and possible relationship with psychiatric disorders. RNA-seq raw data of 164 mouse brain samples from 3 independent datasets with stressors including chronic social defeat stress (CSDS), early life stress (ELS), and two-hit stress of combined CSDS and ELS were collected. There were more changes in splicing than in gene expression in the ventral hippocampus and medial prefrontal cortex, but stress-induced changes of individual genes by differential splicing and differential expression could not be replicated. In contrast, pathway analyses produced robust findings: stress-induced differentially spliced genes (DSGs) were reproducibly enriched in neural transmission and blood-brain barrier systems, and DEGs were reproducibly enriched in stress response-related functions. The hub genes of DSG-related PPI networks were enriched in synaptic functions. The corresponding human homologs of stress-induced DSGs were robustly enriched in AD-related DSGs as well as BD and SCZ in GWAS. These results suggested that stress-induced DSGs from different datasets belong to the same biological system throughout the stress response process, resulting in consistent stress response effects.
Collapse
Affiliation(s)
- Feiran Wang
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, and Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiuju Yang
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, and Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zongyao Ren
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, and Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chao Chen
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, and Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center on Mental Disorders, The Second Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - Chunyu Liu
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, and Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China.
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, 13210, USA.
| |
Collapse
|
39
|
Nieves GM, Bravo M, Bath KG. Early life adversity ablates sex differences in active versus passive threat responding in mice. Stress 2023; 26:2244598. [PMID: 37624104 PMCID: PMC10529224 DOI: 10.1080/10253890.2023.2244598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 07/28/2023] [Indexed: 08/26/2023] Open
Abstract
Early life adversity (ELA) heightens the risk for anxiety disorders (which are characterized by heightened fear and avoidance behaviors), with females being twice as likely as males to develop pathology. Pavlovian fear conditioning tasks have been used to study possible mechanisms supporting endophenotypes of pathology. Identification of sex and ELA selective effects on the nature of behavioral responding in these paradigms may provide a unique window into coping strategies in response to learned fear to guide more mechanistic studies. The goals of this study were two-fold; First, to test if male and female mice employed different coping strategies in response to threat learning using different conditioning parameters (low, medium, and high intensity foot shocks). Second, to test if ELA in the form of limited bedding and nesting (LBN) altered the behavioral response of mice to conditioning. Mice received 6 tone/foot-shock pairings at one of three different foot-shock intensities (0.35 mA; 0.57 mA; 0.7 mA). Freezing, darting, and foot-shock reactivity were measured across trials. During conditioning, control-reared female mice exhibited significantly higher rates of darting behavior compared to control males at nearly all shock intensities tested. LBN rearing decreased the proportion of darting females to levels observed in males. Thus, ELA in the form of LBN significantly diminished the recruitment of active versus passive coping strategies in female mice but did not generally change male responding. Additional work will be required to understand the neural basis of these behavioral effects. Findings extending from this work have the potential to shed light on how ELA impacts trajectories of regional brain development with implications for sex-selective risk for behavioral endophenotypes associated with pathology and possibly symptom presentation.
Collapse
Affiliation(s)
- Gabriela Manzano Nieves
- Department of Psychiatry, Sackler Institute for Developmental Psychobiology, and Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10021
| | - Marilyn Bravo
- David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA, 90095
| | - Kevin G. Bath
- Division of Developmental Neuroscience, New York State Psychiatric Institute/Research Foundation for Mental Hygiene, 1051 Riverside Drive, New York, NY, 10032
- Department of Psychiatry, Columbia University Medical College, New York, NY 1003
| |
Collapse
|
40
|
Rocks D, Jaric I, Bellia F, Cham H, Greally JM, Suzuki M, Kundakovic M. Early-life stress and ovarian hormones alter transcriptional regulation in the nucleus accumbens resulting in sex-specific responses to cocaine. Cell Rep 2023; 42:113187. [PMID: 37777968 PMCID: PMC10753961 DOI: 10.1016/j.celrep.2023.113187] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/29/2023] [Accepted: 09/12/2023] [Indexed: 10/03/2023] Open
Abstract
Early-life stress and ovarian hormones contribute to increased female vulnerability to cocaine addiction. Here, we reveal molecular substrates in the reward area, the nucleus accumbens, through which these female-specific factors affect immediate and conditioning responses to cocaine. We find shared involvement of X chromosome inactivation-related and estrogen signaling-related gene regulation in enhanced conditioning responses following early-life stress and during the low-estrogenic state in females. Low-estrogenic females respond to acute cocaine by opening neuronal chromatin enriched for the sites of ΔFosB, a transcription factor implicated in chronic cocaine response and addiction. Conversely, high-estrogenic females respond to cocaine by preferential chromatin closing, providing a mechanism for limiting cocaine-driven chromatin and synaptic plasticity. We find that physiological estrogen withdrawal, early-life stress, and absence of one X chromosome all nullify the protective effect of a high-estrogenic state on cocaine conditioning in females. Our findings offer a molecular framework to enable understanding of sex-specific neuronal mechanisms underlying cocaine use disorder.
Collapse
Affiliation(s)
- Devin Rocks
- Department of Biological Sciences, Fordham University, Bronx, NY, USA
| | - Ivana Jaric
- Department of Biological Sciences, Fordham University, Bronx, NY, USA
| | - Fabio Bellia
- Department of Biological Sciences, Fordham University, Bronx, NY, USA
| | - Heining Cham
- Department of Psychology, Fordham University, Bronx, NY, USA
| | - John M Greally
- Center for Epigenomics, Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Masako Suzuki
- Center for Epigenomics, Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Nutrition, Texas A&M University, College Station, TX, USA
| | - Marija Kundakovic
- Department of Biological Sciences, Fordham University, Bronx, NY, USA.
| |
Collapse
|
41
|
Rombaut C, Roura-Martinez D, Lepolard C, Gascon E. Brief and long maternal separation in C57Bl6J mice: behavioral consequences for the dam and the offspring. Front Behav Neurosci 2023; 17:1269866. [PMID: 37936649 PMCID: PMC10626007 DOI: 10.3389/fnbeh.2023.1269866] [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: 07/31/2023] [Accepted: 10/09/2023] [Indexed: 11/09/2023] Open
Abstract
Introduction Animal models, especially rodents, have become instrumental to experimentally investigate the effects of an adverse post-natal environment on the developing brain. For this purpose, maternal separation (MS) paradigms have been widely used in the last decades. Nonetheless, how MS affects maternal behavior and, ultimately, the offspring depend on multiple variables. Methods To gain further insights into the consequences of MS, we decided to thoroughly measure and compare the effects of short (15 min, 3 times/day) vs. long (3 h, 1 time/day) separation on multiple maternally-associated behaviors and across the entire post-natal period. Results Compared to unhandled control litters, our results confirmed previous studies and indicated that SMS enhanced the time and variety of maternal care whereas LMS resulted in poor caregiving. We also showed that SMS-accrued caregiving persisted during the whole post-natal period. In contrast, LMS effects on maternal behavior were restricted to the early life (P2-P10). Finally, we also analyzed the behavioral consequences of these different rearing social environments on the offspring. We found that MS has profound effects in social tasks. We showed that affiliative touch, a type of prosocial behavior that provides comfort to others, is particularly sensitive to the modification of maternal caregiving. Discussion Our results provide further support to the contention that interactions during the early post-natal period critically contribute to emotional processing and brain co-construction.
Collapse
Affiliation(s)
| | | | | | - Eduardo Gascon
- Aix Marseille Univ, CNRS, INT, Inst Neurosci Timone, Marseille, France
| |
Collapse
|
42
|
Battaglia M, Rossignol O, Lorenzo LE, Deguire J, Godin AG, D’Amato FR, De Koninck Y. Enhanced harm detection following maternal separation: Transgenerational transmission and reversibility by inhaled amiloride. SCIENCE ADVANCES 2023; 9:eadi8750. [PMID: 37792939 PMCID: PMC10550232 DOI: 10.1126/sciadv.adi8750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/01/2023] [Indexed: 10/06/2023]
Abstract
Early-life adversities are associated with altered defensive responses. Here, we demonstrate that the repeated cross-fostering (RCF) paradigm of early maternal separation is associated with enhancements of distinct homeostatic reactions: hyperventilation in response to hypercapnia and nociceptive sensitivity, among the first generation of RCF-exposed animals, as well as among two successive generations of their normally reared offspring, through matrilineal transmission. Parallel enhancements of acid-sensing ion channel 1 (ASIC1), ASIC2, and ASIC3 messenger RNA transcripts were detected transgenerationally in central neurons, in the medulla oblongata, and in periaqueductal gray matter of RCF-lineage animals. A single, nebulized dose of the ASIC-antagonist amiloride renormalized respiratory and nociceptive responsiveness across the entire RCF lineage. These findings reveal how, following an early-life adversity, a biological memory reducible to a molecular sensor unfolds, shaping adaptation mechanisms over three generations. Our findings are entwined with multiple correlates of human anxiety and pain conditions and suggest nebulized amiloride as a therapeutic avenue.
Collapse
Affiliation(s)
- Marco Battaglia
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Child Youth and Emerging Adult Programme, Centre for Addiction and Mental Health, Toronto, ON, Canada
- CERVO Brain Research Centre, Québec Mental Health Institute, Québec City, QC, Canada
- Department of Psychiatry and Neuroscience, Université Laval, Québec City, QC, Canada
| | - Orlane Rossignol
- CERVO Brain Research Centre, Québec Mental Health Institute, Québec City, QC, Canada
| | - Louis-Etienne Lorenzo
- CERVO Brain Research Centre, Québec Mental Health Institute, Québec City, QC, Canada
| | - Jasmin Deguire
- CERVO Brain Research Centre, Québec Mental Health Institute, Québec City, QC, Canada
| | - Antoine G. Godin
- CERVO Brain Research Centre, Québec Mental Health Institute, Québec City, QC, Canada
- Department of Psychiatry and Neuroscience, Université Laval, Québec City, QC, Canada
| | - Francesca R. D’Amato
- Institute of Biochemistry and Cell Biology, National Research Council, Rome, Italy
| | - Yves De Koninck
- CERVO Brain Research Centre, Québec Mental Health Institute, Québec City, QC, Canada
- Department of Psychiatry and Neuroscience, Université Laval, Québec City, QC, Canada
| |
Collapse
|
43
|
Kong CH, Park K, Kim DY, Kim JY, Kang WC, Jeon M, Min JW, Lee WH, Jung SY, Ryu JH. Effects of oleanolic acid and ursolic acid on depression-like behaviors induced by maternal separation in mice. Eur J Pharmacol 2023; 956:175954. [PMID: 37541369 DOI: 10.1016/j.ejphar.2023.175954] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/11/2023] [Accepted: 08/01/2023] [Indexed: 08/06/2023]
Abstract
Oleanolic acid (OA) and ursolic acid (UA) are structural isomeric triterpenoids. Both triterpenoids have been reported to be able to improve depression. However, no studies have compared their effects in the same system. Whether OA or UA could ameliorate depression-like behaviors in maternal separation (MS)-induced depression-like model was investigated. MS model is a well-accepted mouse model that can reflect the phenotype and pathogenesis of depression. Depression is a mental illness caused by neuroinflammation or changes in neuroplasticity in certain brain regions, such as the prefrontal cortex and hippocampus. Depression-like behaviors were measured using splash test or forced swimming test. In addition, anxiety-like behaviors were also measured using the open field test or elevated plus-maze test. MS-treated female mice showed greater depression-like behaviors than male mice, and that OA improved several depression-like behaviors, whereas UA only relieved anxiety-like behavior of MS-treated mice. Microglial activation, expression levels of TNF-α, and mRNA levels of IDO1 were increased in the hippocampi of MS-treated female mice. However, OA and UA treatments attenuated such increases. In addition, expression levels of synaptophysin and PSD-95 were decreased in the hippocampi of MS-treated female mice. These decreased expression levels of synaptophysin were reversed by both OA and UA treatments, although decreased PSD-95 expression levels were only reversed by OA treatment. Our findings suggest that MS cause depression-like behaviors through female-specific neuroinflammation, changes of tryptophan metabolism, and alterations of synaptic plasticity. Our findings also suggest that OA could reverse MS-induced depression-like behaviors more effectively than UA.
Collapse
Affiliation(s)
- Chang Hyeon Kong
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Keontae Park
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Do Yeon Kim
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Jae Youn Kim
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Woo Chang Kang
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Mijin Jeon
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Ji Won Min
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Won Hyung Lee
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Seo Yun Jung
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Jong Hoon Ryu
- Department of Biomedical and Pharmaceutical Sciences, Kyung Hee University, Seoul, 02447, Republic of Korea; Department of Oriental Pharmaceutical Science, Kyung Hee University, Seoul, 02447, Republic of Korea.
| |
Collapse
|
44
|
Zhang X, Eladawi MA, Ryan WG, Fan X, Prevoznik S, Devale T, Ramnani B, Malathi K, Sibille E, Mccullumsmith R, Tomoda T, Shukla R. Ribosomal dysregulation: A conserved pathophysiological mechanism in human depression and mouse chronic stress. PNAS NEXUS 2023; 2:pgad299. [PMID: 37822767 PMCID: PMC10563789 DOI: 10.1093/pnasnexus/pgad299] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 09/05/2023] [Indexed: 10/13/2023]
Abstract
The underlying biological mechanisms that contribute to the heterogeneity of major depressive disorder (MDD) presentation remain poorly understood, highlighting the need for a conceptual framework that can explain this variability and bridge the gap between animal models and clinical endpoints. Here, we hypothesize that comparative analysis of molecular data from different experimental systems of chronic stress, and MDD has the potential to provide insight into these mechanisms and address this gap. Thus, we compared transcriptomic profiles of brain tissue from postmortem MDD subjects and from mice exposed to chronic variable stress (CVS) to identify orthologous genes. Ribosomal protein genes (RPGs) were down-regulated, and associated ribosomal protein (RP) pseudogenes were up-regulated in both conditions. A seeded gene co-expression analysis using altered RPGs common between the MDD and CVS groups revealed that down-regulated RPGs homeostatically regulated the synaptic changes in both groups through a RP-pseudogene-driven mechanism. In vitro analysis demonstrated that the RPG dysregulation was a glucocorticoid-driven endocrine response to stress. In silico analysis further demonstrated that the dysregulation was reversed during remission from MDD and selectively responded to ketamine but not to imipramine. This study provides the first evidence that ribosomal dysregulation during stress is a conserved phenotype in human MDD and chronic stress-exposed mouse. Our results establish a foundation for the hypothesis that stress-induced alterations in RPGs and, consequently, ribosomes contribute to the synaptic dysregulation underlying MDD and chronic stress-related mood disorders. We discuss the role of ribosomal heterogeneity in the variable presentations of depression and other mood disorders.
Collapse
Affiliation(s)
- Xiaolu Zhang
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Centre, Shreveport, LA 71105, USA
| | - Mahmoud Ali Eladawi
- Department of Neurosciences, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - William George Ryan
- Department of Neurosciences, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Xiaoming Fan
- Department of Medicine, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Stephen Prevoznik
- Department of Neurosciences, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Trupti Devale
- Department of Biological Sciences, College of Natural Sciences and Mathematics, University of Toledo, Toledo, OH 43614, USA
| | - Barkha Ramnani
- Department of Biological Sciences, College of Natural Sciences and Mathematics, University of Toledo, Toledo, OH 43614, USA
| | - Krishnamurthy Malathi
- Department of Biological Sciences, College of Natural Sciences and Mathematics, University of Toledo, Toledo, OH 43614, USA
| | - Etienne Sibille
- Centre for Addiction and Mental Health, Campbell Family Mental Health Research Institute, Toronto, ON M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON M5T 1R8, Canada
| | - Robert Mccullumsmith
- Department of Neurosciences, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
- Neurosciences Institute, ProMedica, Toledo, OH 43614, USA
| | - Toshifumi Tomoda
- Centre for Addiction and Mental Health, Campbell Family Mental Health Research Institute, Toronto, ON M5T 1R8, Canada
| | - Rammohan Shukla
- Department of Zoology and Physiology, University of Wyoming, Laramie, WY 82071, USA
| |
Collapse
|
45
|
Mujtaba S, Patro IK, Patro N. Multiple Early Life Stressors as Risk Factors for Neurodevelopmental Abnormalities in the F1 Wistar Rats. Brain Sci 2023; 13:1360. [PMID: 37891729 PMCID: PMC10605318 DOI: 10.3390/brainsci13101360] [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: 07/07/2023] [Revised: 09/07/2023] [Accepted: 09/20/2023] [Indexed: 10/29/2023] Open
Abstract
Cumulative exposure to multiple early life stressors is expected to affect behavioral development, causing increased susceptibility to neuropsychiatric disorders. The present study was designed to mimic such conditions in a rat model to study behavioral impairments during adolescence and adulthood. Female Wistar rats (n = 32; 140-150 gm) were switched to a low protein (LP; 8% protein) or control (20% protein) diet 15 days prior to conception, and then the diet regime was maintained throughout the experimental period. Pups born to control and LP dams were intraperitoneally injected with deltamethrin (DLT-pyrethroid insecticide; 0.7 mg/kg body weight; PND 1 to 7), lipopolysaccharide (LPS-bacterial endotoxin; 0.3 mg/kg body weight; PND 3 and 5), or DLT+LPS, on designated days forming eight experimental groups (Control, LP, Control+LPS, LP+LPS, Control+DLT, LP+DLT, Control+DLT+LPS and LP+DLT+LPS). Neurobehavioral assessments were performed in F1 rats (1, 3, 6 months) by open field, elevated plus maze, light and dark box, and rotarod tests. LP rats were found to be highly susceptible to either singular or cumulative exposure as compared to their age-matched control counterparts, showing significantly severe behavioral abnormalities, such as hyperactivity, attention deficits and low anxiety, the hallmark symptoms of neuropsychiatric disorders like schizophrenia and ADHD, suggesting thereby that early life multi-hit exposure may predispose individuals to developmental disorders.
Collapse
Affiliation(s)
- Syed Mujtaba
- School of Studies in Neuroscience, Jiwaji University, Gwalior 474011, India; (S.M.); (I.K.P.)
- School of Studies in Zoology, Jiwaji University, Gwalior 474011, India
| | - Ishan Kumar Patro
- School of Studies in Neuroscience, Jiwaji University, Gwalior 474011, India; (S.M.); (I.K.P.)
- School of Studies in Zoology, Jiwaji University, Gwalior 474011, India
| | - Nisha Patro
- School of Studies in Neuroscience, Jiwaji University, Gwalior 474011, India; (S.M.); (I.K.P.)
| |
Collapse
|
46
|
Patterson SK, Petersen RM, Brent LJN, Snyder-Mackler N, Lea AJ, Higham JP. Natural Animal Populations as Model Systems for Understanding Early Life Adversity Effects on Aging. Integr Comp Biol 2023; 63:681-692. [PMID: 37279895 PMCID: PMC10503476 DOI: 10.1093/icb/icad058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/25/2023] [Accepted: 05/30/2023] [Indexed: 06/08/2023] Open
Abstract
Adverse experiences in early life are associated with aging-related disease risk and mortality across many species. In humans, confounding factors, as well as the difficulty of directly measuring experiences and outcomes from birth till death, make it challenging to identify how early life adversity impacts aging and health. These challenges can be mitigated, in part, through the study of non-human animals, which are exposed to parallel forms of adversity and can age similarly to humans. Furthermore, studying the links between early life adversity and aging in natural populations of non-human animals provides an excellent opportunity to better understand the social and ecological pressures that shaped the evolution of early life sensitivities. Here, we highlight ongoing and future research directions that we believe will most effectively contribute to our understanding of the evolution of early life sensitivities and their repercussions.
Collapse
Affiliation(s)
- Sam K Patterson
- Department of Anthropology, New York University, New York City, 10003, USA
| | - Rachel M Petersen
- Department of Biological Science, Vanderbilt University, Nashville, 37232, USA
| | - Lauren J N Brent
- Department of Psychology, University of Exeter, Exeter, EX4 4QG, United Kingdom
| | - Noah Snyder-Mackler
- School of Life Sciences, Center for Evolution and Medicine, and School of Human Evolution and Social Change, Arizona State University, Tempe, 85281, USA
| | - Amanda J Lea
- Department of Biological Science, Vanderbilt University, Nashville, 37232, USA
- Child and Brain Development Program, Canadian Institute for Advanced Study, Toronto, M5G 1M1, Canada
| | - James P Higham
- Department of Anthropology, New York University, New York City, 10003, USA
| |
Collapse
|
47
|
Fitzgerald E, Arcego DM, Shen MJ, O'Toole N, Wen X, Nagy C, Mostafavi S, Craig K, Silveira PP, Rayan NA, Diorio J, Meaney MJ, Zhang TY. Sex and cell-specific gene expression in corticolimbic brain regions associated with psychiatric disorders revealed by bulk and single-nuclei RNA sequencing. EBioMedicine 2023; 95:104749. [PMID: 37549631 PMCID: PMC10432187 DOI: 10.1016/j.ebiom.2023.104749] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 06/28/2023] [Accepted: 07/25/2023] [Indexed: 08/09/2023] Open
Abstract
BACKGROUND There are sex-specific differences in the prevalence, symptomology and course of psychiatric disorders. However, preclinical models have primarily used males, such that the molecular mechanisms underlying sex-specific differences in psychiatric disorders are not well established. METHODS In this study, we compared transcriptome-wide gene expression profiles in male and female rats within the corticolimbic system, including the cingulate cortex, nucleus accumbens medial shell (NAcS), ventral dentate gyrus and the basolateral amygdala (n = 22-24 per group/region). FINDINGS We found over 3000 differentially expressed genes (DEGs) in the NAcS between males and females. Of these DEGs in the NAcS, 303 showed sex-dependent conservation DEGs in humans and were significantly enriched for gene ontology terms related to blood vessel morphogenesis and regulation of cell migration. Single nuclei RNA sequencing in the NAcS of male and female rats identified widespread sex-dependent expression, with genes upregulated in females showing a notable enrichment for synaptic function. Female upregulated genes in astrocytes, Drd3+MSNs and oligodendrocyte were also enriched in several psychiatric genome-wide association studies (GWAS). INTERPRETATION Our data provide comprehensive evidence of sex- and cell-specific molecular profiles in the NAcS. Importantly these differences associate with anxiety, bipolar disorder, schizophrenia, and cross-disorder, suggesting an intrinsic molecular basis for sex-based differences in psychiatric disorders that strongly implicates the NAcS. FUNDING This work was supported by funding from the Hope for Depression Research Foundation (MJM).
Collapse
Affiliation(s)
- Eamon Fitzgerald
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, H4H 1R3, Canada; Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montréal, H4H 1R3, Canada
| | - Danusa Mar Arcego
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, H4H 1R3, Canada; Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montréal, H4H 1R3, Canada
| | - Mo Jun Shen
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Nicholas O'Toole
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, H4H 1R3, Canada; Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montréal, H4H 1R3, Canada
| | - Xianglan Wen
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, H4H 1R3, Canada; Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montréal, H4H 1R3, Canada
| | - Corina Nagy
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, H4H 1R3, Canada
| | - Sara Mostafavi
- Paul G. Allen School of Computer Science and Engineering, University of Washington, 185 E Stevens Way NE, Seattle, WA 9819, USA
| | - Kelly Craig
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, H4H 1R3, Canada
| | - Patricia Pelufo Silveira
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, H4H 1R3, Canada; Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montréal, H4H 1R3, Canada; Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Nirmala Arul Rayan
- Translational Neuroscience Program, Singapore Institute for Clinical Sciences and Brain - Body Initiative, Agency for Science, Technology and Research (A∗STAR), Singapore
| | - Josie Diorio
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, H4H 1R3, Canada
| | - Michael J Meaney
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, H4H 1R3, Canada; Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montréal, H4H 1R3, Canada; Translational Neuroscience Program, Singapore Institute for Clinical Sciences and Brain - Body Initiative, Agency for Science, Technology and Research (A∗STAR), Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Tie-Yuan Zhang
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, H4H 1R3, Canada; Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montréal, H4H 1R3, Canada.
| |
Collapse
|
48
|
Yuan M, Yang B, Rothschild G, Mann JJ, Sanford LD, Tang X, Huang C, Wang C, Zhang W. Epigenetic regulation in major depression and other stress-related disorders: molecular mechanisms, clinical relevance and therapeutic potential. Signal Transduct Target Ther 2023; 8:309. [PMID: 37644009 PMCID: PMC10465587 DOI: 10.1038/s41392-023-01519-z] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 05/14/2023] [Accepted: 05/31/2023] [Indexed: 08/31/2023] Open
Abstract
Major depressive disorder (MDD) is a chronic, generally episodic and debilitating disease that affects an estimated 300 million people worldwide, but its pathogenesis is poorly understood. The heritability estimate of MDD is 30-40%, suggesting that genetics alone do not account for most of the risk of major depression. Another factor known to associate with MDD involves environmental stressors such as childhood adversity and recent life stress. Recent studies have emerged to show that the biological impact of environmental factors in MDD and other stress-related disorders is mediated by a variety of epigenetic modifications. These epigenetic modification alterations contribute to abnormal neuroendocrine responses, neuroplasticity impairment, neurotransmission and neuroglia dysfunction, which are involved in the pathophysiology of MDD. Furthermore, epigenetic marks have been associated with the diagnosis and treatment of MDD. The evaluation of epigenetic modifications holds promise for further understanding of the heterogeneous etiology and complex phenotypes of MDD, and may identify new therapeutic targets. Here, we review preclinical and clinical epigenetic findings, including DNA methylation, histone modification, noncoding RNA, RNA modification, and chromatin remodeling factor in MDD. In addition, we elaborate on the contribution of these epigenetic mechanisms to the pathological trait variability in depression and discuss how such mechanisms can be exploited for therapeutic purposes.
Collapse
Affiliation(s)
- Minlan Yuan
- Mental Health Center and Psychiatric Laboratory, the State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Biao Yang
- Department of Abdominal Oncology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Gerson Rothschild
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, 10032, USA
| | - J John Mann
- Department of Psychiatry, Columbia University, New York, NY, 10032, USA
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, NY, 10032, USA
- Department of Radiology, Columbia University, New York, NY, 10032, USA
| | - Larry D Sanford
- Sleep Research Laboratory, Center for Integrative Neuroscience and Inflammatory Diseases, Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Xiangdong Tang
- Sleep Medicine Center, Department of Respiratory and Critical Care Medicine, Mental Health Center, Translational Neuroscience Center, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Canhua Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chuang Wang
- Department of Pharmacology, and Provincial Key Laboratory of Pathophysiology in School of Medicine, Ningbo University, Ningbo, Zhejiang, 315211, China.
| | - Wei Zhang
- Mental Health Center and Psychiatric Laboratory, the State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Medical Big Data Center, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
49
|
Bennett SN, Chang AB, Rogers FD, Jones P, Peña CJ. Thyroid hormones mediate the impact of early-life stress on ventral tegmental area gene expression and behavior. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.25.554785. [PMID: 37662236 PMCID: PMC10473690 DOI: 10.1101/2023.08.25.554785] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Proper thyroid function is essential to the developing brain, including dopamine neuron differentiation, growth, and maintenance. Stress across the lifespan impacts thyroid hormone signaling and anxiety disorders and depression have been associated with thyroid dysfunction (both hypo- and hyper-active). However, less is known about how stress during postnatal development impacts thyroid function and related brain development. Our previous work in mice demonstrated that early-life stress (ELS) transiently impinged on expression of a transcription factor in dopamine neurons shown to be regulated by thyroid hormones. We hypothesized that thyroid hormone signaling may link experience of ELS with transcriptional dysregulation within the dopaminergic midbrain, and ultimately behavior. Here, we find that ELS transiently increases thyroid-stimulating hormone levels (inversely related to thyroid signaling) in both male and female mice at P21, an effect which recovers by adolescence. We next tested whether transient treatment of ELS mice with synthetic thyroid hormone (levothyroxine, LT4) could ameliorate the impact of ELS on sensitivity to future stress, and on expression of genes related to dopamine neuron development and maintenance, thyroid signaling, and plasticity within the ventral tegmental area. Among male mice, but not females, juvenile LT4 treatment prevented hypersensitivity to adult stress. We also found that rescuing developmental deficits in thyroid hormone signaling after ELS restored levels of some genes altered directly by ELS, and prevented alterations in expression of other genes sensitive to the second hit of adult stress. These findings suggest that thyroid signaling mediates the deleterious impact of ELS on VTA development, and that temporary treatment of hypothyroidism after ELS may be sufficient to prevent future stress hypersensitivity.
Collapse
|
50
|
Balouek JA, Mclain CA, Minerva AR, Rashford RL, Bennett SN, Rogers FD, Peña CJ. Reactivation of Early-Life Stress-Sensitive Neuronal Ensembles Contributes to Lifelong Stress Hypersensitivity. J Neurosci 2023; 43:5996-6009. [PMID: 37429717 PMCID: PMC10451005 DOI: 10.1523/jneurosci.0016-23.2023] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 06/01/2023] [Accepted: 06/17/2023] [Indexed: 07/12/2023] Open
Abstract
Early-life stress (ELS) is one of the strongest lifetime risk factors for depression, anxiety, suicide, and other psychiatric disorders, particularly after facing additional stressful events later in life. Human and animal studies demonstrate that ELS sensitizes individuals to subsequent stress. However, the neurobiological basis of such stress sensitization remains largely unexplored. We hypothesized that ELS-induced stress sensitization would be detectable at the level of neuronal ensembles, such that cells activated by ELS would be more reactive to adult stress. To test this, we leveraged transgenic mice to genetically tag, track, and manipulate experience-activated neurons. We found that in both male and female mice, ELS-activated neurons within the nucleus accumbens (NAc), and to a lesser extent the medial prefrontal cortex, were preferentially reactivated by adult stress. To test whether reactivation of ELS-activated ensembles in the NAc contributes to stress hypersensitivity, we expressed hM4Dis receptor in control or ELS-activated neurons of pups and chemogenetically inhibited their activity during experience of adult stress. Inhibition of ELS-activated NAc neurons, but not control-tagged neurons, ameliorated social avoidance behavior following chronic social defeat stress in males. These data provide evidence that ELS-induced stress hypersensitivity is encoded at the level of corticolimbic neuronal ensembles.SIGNIFICANCE STATEMENT Early-life stress enhances sensitivity to stress later in life, yet the mechanisms of such stress sensitization are largely unknown. Here, we show that neuronal ensembles in corticolimbic brain regions remain hypersensitive to stress across the life span, and quieting these ensembles during experience of adult stress rescues stress hypersensitivity.
Collapse
Affiliation(s)
- Julie-Anne Balouek
- Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey 08544
| | - Christabel A Mclain
- Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey 08544
| | - Adelaide R Minerva
- Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey 08544
| | - Rebekah L Rashford
- Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey 08544
| | - Shannon N Bennett
- Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey 08544
| | - Forrest D Rogers
- Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey 08544
| | | |
Collapse
|