1
|
Wu Y, Zhu Y, Zheng S, Mingxing D. Resveratrol alleviates depressive-like behavior via the activation of SIRT1/NF-κB signaling pathway in microglia. Future Sci OA 2025; 11:2463852. [PMID: 39967065 PMCID: PMC11845112 DOI: 10.1080/20565623.2025.2463852] [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: 10/02/2024] [Accepted: 01/15/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Currently, the pathogenesis of depression remains poorly understood, leading to many patients receiving ineffective treatment. Resveratrol has demonstrated beneficial effects in the prevention and treatment of depression. However, it remains unknown whether resveratrol administration can counteract depression-like behaviors by regulating the SIRT1/NF-κB signaling pathway. METHODOLOGY/PRINCIPAL FINDINGS Male C57BL/6 mice were randomly assigned to a control group, a depression group, and a resveratrol group. The depression model was established using chronic unpredictable mild stress (CUMS) for 5 weeks. Behavioral tests were conducted to assess depressive-like behaviors. The expression levels of SIRT1 and NF-κB in the hippocampus of mice and BV2 microglial cells were measured. After 5 weeks of modeling, the results indicated that mice in the depression group exhibited significant depressive-like behaviors and inhibited activation of the SIRT1/NF-κB signaling pathway. In contrast, resveratrol administration effectively reversed these changes. Results from in vitro experiments showed that LPS stimulation increased microglial activity and downregulated the SIRT1/NF-κB signaling pathway in microglia; however, resveratrol treatment mitigated these effects. CONCLUSIONS/SIGNIFICANCE Our findings suggested that resveratrol can alleviate CUMS-induced depression-like behaviors via the activation of the Sirt1/NF-κB pathway in microglia.
Collapse
Affiliation(s)
- Yuehong Wu
- Psychiatry department, Jinhua Second Hospital, Jinhua, Zhejiang Province, China
| | - Yixia Zhu
- Psychiatry department, Jinhua Second Hospital, Jinhua, Zhejiang Province, China
| | - Shun Zheng
- Psychiatry department, Jinhua Second Hospital, Jinhua, Zhejiang Province, China
| | - Ding Mingxing
- Medical Molecular Biology Laboratory, School of Medicine, Jinhua Polytechnic, Jinhua, Zhejiang Province, China
| |
Collapse
|
2
|
Coley-O'Rourke EJ, Lum GR, Pronovost GN, Yu LW, Özcan E, Yu KB, McDermott J, Chakhoyan A, Goldman E, Vuong HE, Paramo J, McCune S, Sejane K, Renwick S, Bode L, Chu A, Calkins KL, Hsiao EY. Murine maternal microbiome modifies adverse effects of protein undernutrition on offspring neurobehaviour. Nat Microbiol 2025:10.1038/s41564-025-02022-7. [PMID: 40514563 DOI: 10.1038/s41564-025-02022-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 04/24/2025] [Indexed: 06/16/2025]
Abstract
Protein undernutrition results in impaired growth and neurobehavioural development in children. However, the impact of timing, environmental factors and maternal versus neonatal influences are unclear. Here, using a mouse model of fetal growth restriction where maternal protein intake is limited during pregnancy, we show that adult offspring exhibit cognitive and anxiety-like behavioural abnormalities. Cross-fostering newborn mice to dams previously exposed to either low protein or standard diet reveals that behavioural impairments in adult offspring require diet-induced conditioning of both fetal development and maternal peripartum physiology. Maternal gut microbiome diversity is reduced, maternal immune, milk, and serum metabolomic profiles are altered, and widespread changes in fetal brain transcriptomic and metabolomic profiles are observed, including subsets of microbiome-dependent metabolites. Finally, we show that dam treatment with a cocktail of ten diet- and microbiome-dependent metabolites results in differential effects on fetal development and postnatal behaviour. Our study highlights the impact of prenatal maternal protein undernutrition on offspring neurobehavioural trajectories and the role of the maternal microbiome.
Collapse
Affiliation(s)
- Elena J Coley-O'Rourke
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, USA.
| | - Gregory R Lum
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, USA
| | - Geoffrey N Pronovost
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, USA
| | - Lewis W Yu
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, USA
| | - Ezgi Özcan
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, USA
| | - Kristie B Yu
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, USA
| | - Janet McDermott
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, USA
| | - Anna Chakhoyan
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, USA
| | - Eliza Goldman
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, USA
| | - Helen E Vuong
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, USA
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Jorge Paramo
- UCLA Goodman-Luskin Microbiome Center, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Sydney McCune
- Department of Pediatrics, Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence (LRF MOMI CORE) and Human Milk Institute (HMI), University of California San Diego, La Jolla, CA, USA
| | - Kristija Sejane
- Department of Pediatrics, Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence (LRF MOMI CORE) and Human Milk Institute (HMI), University of California San Diego, La Jolla, CA, USA
| | - Simone Renwick
- Department of Pediatrics, Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence (LRF MOMI CORE) and Human Milk Institute (HMI), University of California San Diego, La Jolla, CA, USA
| | - Lars Bode
- Department of Pediatrics, Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence (LRF MOMI CORE) and Human Milk Institute (HMI), University of California San Diego, La Jolla, CA, USA
| | - Alison Chu
- Division of Neonatology, Department of Pediatrics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Kara L Calkins
- Division of Neonatology, Department of Pediatrics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Elaine Y Hsiao
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, USA.
- UCLA Goodman-Luskin Microbiome Center, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
3
|
Salles J, Gorse T, Benvegnu G, Raynaud JP, Tauber M. Imprinting disorders as a window to understand pediatric feeding disorders. Orphanet J Rare Dis 2025; 20:247. [PMID: 40413507 PMCID: PMC12102808 DOI: 10.1186/s13023-025-03789-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 05/12/2025] [Indexed: 05/27/2025] Open
Abstract
Imprinting disorders are a group of rare congenital disorders characterized by common clinical features that affect growth, development, metabolism, and shared molecular abnormalities [1]. Patients with these disorders exhibit feeding difficulties and changes in social skills. Pediatric feeding disorders affect approximately 25% of children in the general population but have been difficult to understand and manage globally; indeed, they have traditionally been approached from different professional disciplines, each advocating its own unique method. An interdisciplinary consensus group recently introduced a more integrative definition of pediatric feeding disorders. From this new approach, we hypothesized that the imprinted genes may play an important role in the relationship between feeding and social development. In addition, we hypothesize in this letter that research on imprinting disorders may contribute to a better understanding of pediatric feeding disorders.
Collapse
Affiliation(s)
- Juliette Salles
- Service de psychiatrie d'urgence de crise et de liaison, CHU de Toulouse, Toulouse, France.
- Infinity (Toulouse Institute for Infectious and Inflammatory Diseases), INSERM UMR1291, CNRS UMR5051, Université Toulouse III, Toulouse, France.
| | - Thomas Gorse
- Service Universitaire de Psychiatrie de l'Enfant et de l'Adolescent (SUPEA), CHU de Toulouse, Toulouse, France
- CERPOP (Centre d'Epidémiologie et de Recherche en santé des POPulations), UMR1295, INSERM - Université Toulouse III Paul Sabatier, Toulouse, France
| | - Grégoire Benvegnu
- Service Universitaire de Psychiatrie de l'Enfant et de l'Adolescent (SUPEA), CHU de Toulouse, Toulouse, France
- Centre de compétences Maladies Rares à Expression Psychiatrique, Service Universitaire de Psychiatrie de l'Enfant et de l'Adolescent, CHU de Toulouse, Toulouse, France
- Centre de référence Maladies Rares (PRADORT, Syndrome de Prader‑Willi et autres formes rares d'obésité avec troubles du comportement alimentaire), Service d'Endocrinologie, Obésités, Maladies Osseuses, Génétique et Gynécologie Médicale, ENDO-ERN (European Reference Network on Rare Endocrine Conditions), Hôpital des Enfants, CHU de Toulouse, Toulouse, France
- CERPOP (Centre d'Epidémiologie et de Recherche en santé des POPulations), UMR1295, INSERM - Université Toulouse III Paul Sabatier, Toulouse, France
| | - Jean-Philippe Raynaud
- Service Universitaire de Psychiatrie de l'Enfant et de l'Adolescent (SUPEA), CHU de Toulouse, Toulouse, France
- CERPOP (Centre d'Epidémiologie et de Recherche en santé des POPulations), UMR1295, INSERM - Université Toulouse III Paul Sabatier, Toulouse, France
| | - Maithé Tauber
- Infinity (Toulouse Institute for Infectious and Inflammatory Diseases), INSERM UMR1291, CNRS UMR5051, Université Toulouse III, Toulouse, France
- Centre de Référence du Syndrome de Prader-Willi et Syndromes avec Troubles du Comportement Alimentaire, Hôpital des Enfants, Unité D'endocrinologie, Obésités, Maladies Osseuses, Génétique et Gynécologie Médicale, CHU de Toulouse, Toulouse, France
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Pearson G, Douglas J. Start 'em young, treat 'em right: How horses' early life experiences can set them up for success in life. Equine Vet J 2025; 57:540-545. [PMID: 40045757 DOI: 10.1111/evj.14485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Accepted: 01/23/2025] [Indexed: 04/11/2025]
Affiliation(s)
- Gemma Pearson
- The Horse Trust, Princes Risborough, Buckinghamshire, UK
- Royal (Dick) School of Veterinary Studies, Easter Bush Campus, University of Edinburgh, Midlothian, UK
| | | |
Collapse
|
7
|
Talukdar J, Megha, Choudhary H, Bhatnagar S, Pandit A, Mishra AK, Karmakar S, Sharan P. The Interplay of Chronic Stress and Cancer: Pathophysiology and Implications for Integrated Care. Cancer Rep (Hoboken) 2025; 8:e70143. [PMID: 40387308 PMCID: PMC12087007 DOI: 10.1002/cnr2.70143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 12/16/2024] [Accepted: 01/29/2025] [Indexed: 05/20/2025] Open
Abstract
BACKGROUND Cancer-associated depression is a multifaceted condition that arises from the interplay of biological, psychological, and social factors in individuals diagnosed with cancer. Understanding this condition involves exploring how cancer and its treatments can precipitate depressive symptoms and the mechanisms behind this association. Chronic stress, inflammation, and immunological responses play a crucial role in the development of both cancer and depression. The objective of this review is to describe and synthesize information on the complex interactions between chronic stress, inflammation, immunological responses, and cancer development. Additionally, it aims to review existing evidence regarding mechanisms such as neurotransmitter imbalances, structural brain changes, and genetic predispositions as key contributors to depression in cancer patients. RECENT FINDINGS A comprehensive literature search on Cancer-associated Depression was conducted in electronic databases, including APA PsycINFO, Medline, Google Scholar, Embase, PubMed, Scopus, and Web of Science. The research focused on understanding the potential relationship between stress-induced depression and cancer by examining neurochemical, anatomical, immunological, genetic, and psychological changes. The findings revealed a compilation of both quantitative and qualitative studies on depression in cancer patients. Evidence suggested a potential link between cancer-induced stress and depression, with increased levels of proinflammatory cytokines (such as IL-6) and dysregulation of neurotransmitters, including serotonin, contributing to the onset of depression. Furthermore, studies indicated that antidepressants, along with psychological interventions, were effective in managing depression among cancer patients. CONCLUSION This narrative review provides insights into the importance of integrating oncology and mental health services to address the psychosocial needs of cancer patients. Future research should focus on the bidirectional interactions between stress and cancer, aiming to improve cancer care by incorporating mental health support. Addressing the mental health aspects of cancer treatment can significantly enhance patient outcomes and overall quality of life.
Collapse
Affiliation(s)
- Joyeeta Talukdar
- Department of Bio‐ChemistryAll India Institute of Medical SciencesNew DelhiIndia
| | - Megha
- Department of PsychiatryAll India Institute of Medical SciencesNew DelhiIndia
| | - Hemant Choudhary
- Department of PsychiatryAll India Institute of Medical SciencesNew DelhiIndia
| | - Sushma Bhatnagar
- Department of Onco‐Anaesthesia & Palliative MedicineDR. B.R.A.I.R.C.H, All India Institute of Medical SciencesNew DelhiIndia
| | - Anuja Pandit
- Department of Onco‐Anaesthesia & Palliative MedicineDR. B.R.A.I.R.C.H, All India Institute of Medical SciencesNew DelhiIndia
| | - Ashwani Kumar Mishra
- National Drug Dependence Treatment CentreAll India Institute of Medical SciencesNew DelhiIndia
| | - Subhradip Karmakar
- Department of Bio‐ChemistryAll India Institute of Medical SciencesNew DelhiIndia
| | - Pratap Sharan
- Department of PsychiatryAll India Institute of Medical SciencesNew DelhiIndia
| |
Collapse
|
8
|
Wang M, Liu K, Guo D, Lv Y, Wang X. Arbovirus Infections and Epigenetic Mechanisms; a Potential Therapeutic Target. Rev Med Virol 2025; 35:e70033. [PMID: 40155348 DOI: 10.1002/rmv.70033] [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: 02/07/2025] [Revised: 03/06/2025] [Accepted: 03/11/2025] [Indexed: 04/01/2025]
Abstract
Arboviruses are a group of arthropod-borne viral pathogens that pose a significant threat to the public health system. The clinical manifestations associated with these viruses range from self-limiting infections to life-threatening disorders. As a group of systemic viral infections, arboviruses can affect various parts of human organ systems, such as the nervous system. In the nervous system, epigenetic mechanisms are involved in various mechanisms including adult neurogenesis, neuronal-glial differentiation, the regulation of neural behaviour and neural plasticity, as well as other brain functions such as memory, and cognition. Hence, epigenetic deregulation is a key factor in the aetiology of different neurological disorders that highlights the importance of studying the underlying mechanisms and risk factors to introduce effective therapeutic approaches. There is mounting evidence that arboviruses that affect the nervous system take advantage of various mechanisms to modulate epigenetic processes to regulate their life cycles. This phenomenon may affect the nervous system leading to neurotropic arboviral infection-associated neurological disorders. Hence, it is important to understand reciprocal interplays between neurotropic arboviral pathogens and epigenetic processes to better control these disorders. The present review provides an overview of different interactions of arboviruses with epigenetic mechanisms during neurotropic arboviral infections. It uniquely focuses on the interplay between epigenetic modifications and arboviral neurotropism, shedding light on potential therapeutic strategies that have not been comprehensively addressed before. Targeting virus-induced epigenetic alterations, such as miRNA regulation, could lead to novel antiviral therapies aimed at mitigating neuroinflammation and disease severity.
Collapse
Affiliation(s)
- Manhong Wang
- University Hospital, Jilin Normal University, Siping, China
| | - Kexin Liu
- Department of Pathology, Siping City Centeral People's Hospital, Siping, China
| | - Dan Guo
- University Hospital, Jilin Normal University, Siping, China
| | - Youjia Lv
- Department of Hepatology, Siping City Infectious Disease Hospital, Siping, China
| | - Xin Wang
- Student Affairs Office, Jilin Normal University, Siping, China
| |
Collapse
|
9
|
Lauby SC, Agarwal I, Lapp HE, Salazar M, Semyrenko S, Chauhan D, Margolis AE, Champagne FA. Interplay between prenatal bisphenol exposure, postnatal maternal care, and offspring sex in predicting DNA methylation relevant to anxiety-like behavior in rats. Horm Behav 2025; 172:105745. [PMID: 40273582 DOI: 10.1016/j.yhbeh.2025.105745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 04/04/2025] [Accepted: 04/14/2025] [Indexed: 04/26/2025]
Abstract
Prenatal exposure to endocrine disrupting chemicals, such as bisphenols, can alter neurodevelopmental trajectories and have a lasting neurobehavioral impact through epigenetic pathways. However, outcomes associated with prenatal bisphenol exposure may also be shaped by the postnatal environment and collectively these environmental effects may be sex-specific. Thus, an integrative research design that includes multiple early life exposures and considers sex differences may be essential for predicting outcomes. In the current study, we use a multivariate approach to examine the contributions of prenatal bisphenol exposure, postnatal maternal care, and offspring sex to variation in DNA methylation of well-studied candidate genes (NR3C1, BDNF, OXTR) in the ventral hippocampus and amygdala of adult Long-Evans rats. Main effects of postnatal maternal care and interactions with prenatal bisphenol exposure were consistently found for DNA methylation within the NR3C1 gene (ventral hippocampus) and within the BDNF and OXTR genes (amygdala). Sex-specific effects were also found across all analyses. Overall, our findings suggest that both early-life factors (prenatal and postnatal) and offspring sex contribute to variation in DNA methylation in genes and brain regions relevant for the expression of anxiety-like behavior. These results highlight the need to consider the brain region-specific effects of multiple exposures in males and females to understand the lasting effects of early environments.
Collapse
Affiliation(s)
- Samantha C Lauby
- Department of Psychology, College of Liberal Arts, University of Texas at Austin, Austin, TX, United States of America; Center for Molecular Carcinogenesis and Toxicology, University of Texas at Austin, Austin, TX, United States of America
| | - Isha Agarwal
- Department of Psychology, College of Liberal Arts, University of Texas at Austin, Austin, TX, United States of America
| | - Hannah E Lapp
- Department of Psychology, College of Liberal Arts, University of Texas at Austin, Austin, TX, United States of America
| | - Melissa Salazar
- Department of Psychology, College of Liberal Arts, University of Texas at Austin, Austin, TX, United States of America
| | - Sofiia Semyrenko
- Department of Psychology, College of Liberal Arts, University of Texas at Austin, Austin, TX, United States of America
| | - Danyal Chauhan
- Department of Psychology, College of Liberal Arts, University of Texas at Austin, Austin, TX, United States of America
| | - Amy E Margolis
- Department of Psychiatry, The Ohio State Univeristy, Columbus, OH, United States of America
| | - Frances A Champagne
- Department of Psychology, College of Liberal Arts, University of Texas at Austin, Austin, TX, United States of America; Center for Molecular Carcinogenesis and Toxicology, University of Texas at Austin, Austin, TX, United States of America.
| |
Collapse
|
10
|
McKenna BG, Lussier AA, Suderman MJ, Walton E, Simpkin AJ, Hüls A, Dunn EC. Strengthening Rigor and Reproducibility in Epigenome-Wide Association Studies of Social Exposures and Brain-Based Health Outcomes. Curr Environ Health Rep 2025; 12:19. [PMID: 40254641 PMCID: PMC12009779 DOI: 10.1007/s40572-024-00469-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2024] [Indexed: 04/22/2025]
Abstract
PURPOSE OF REVIEW Studies examining the effects of social factors on the epigenome have proliferated over the last two decades. Social epigenetics research to date has broadly demonstrated that social factors spanning childhood adversity, to neighborhood disadvantage, educational attainment, and economic instability are associated with alterations to DNA methylation that may have a functional impact on health. These relationships are particularly relevant to brain-based health outcomes such as psychiatric disorders, which are strongly influenced by social exposures and are also the leading cause of disability worldwide. However, social epigenetics studies are limited by the many challenges faced by both epigenome-wide association studies (EWAS) and the study of social factors. FINDINGS In this manuscript, we provide a framework to achieve greater rigor and reproducibility in social epigenetics research. We discuss current limitations of the social epigenetics field, as well as existing and new solutions to improve rigor and reproducibility. Readers will gain a better understanding of the current considerations and processes that could maximize rigor when conducting social epigenetics research, as well as the technologies and approaches that merit attention and investment to propel continued discovery into the future.
Collapse
Affiliation(s)
- Brooke G McKenna
- Center for Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA.
- Department of Sociology, Purdue University, West Lafayette, IN, USA.
| | - Alexandre A Lussier
- Center for Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Matthew J Suderman
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Esther Walton
- Department of Psychology, University of Bath, Bath, UK
| | - Andrew J Simpkin
- School of Mathematical and Statistical Sciences, University of Galway, Galway, Ireland
| | - Anke Hüls
- Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Erin C Dunn
- Center for Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Department of Sociology, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
11
|
Ferrer A, Labad J, Salvat-Pujol N, Urretavizcaya M, Obrador-Font R, Menchón JM, Palao D, Soriano-Mas C, Costas J, Barrachina M, Carracedo Á, Soria V. Genetic and epigenetic changes to the glucocorticoid receptor gene (NR3C1) and cognition in major depressive disorder. SPANISH JOURNAL OF PSYCHIATRY AND MENTAL HEALTH 2025:S2950-2853(25)00018-3. [PMID: 40189105 DOI: 10.1016/j.sjpmh.2024.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 11/14/2024] [Accepted: 12/18/2024] [Indexed: 04/16/2025]
Abstract
INTRODUCTION Many studies have found that hypothalamic-pituitary-adrenal (HPA) axis abnormalities are related to the pathophysiology of major depressive disorder (MDD) and cognitive functioning. Our aim was to assess the influence of genetic polymorphisms and methylation levels in three different promoter regions throughout the glucocorticoid receptor (GR) gene NR3C1 on cognitive performance in MDD. Plausible interactions with childhood adversity and mediation relationships between genetic and epigenetic variables were explored. MATERIALS AND METHODS The sample included a total of 64 MDD patients and 82 healthy controls. Child maltreatment and neurocognitive performance were assessed in all participants. HPA negative feedback was analyzed using the dexamethasone suppression test after the administration of 0.25mg of dexamethasone. A total of 23 single-nucleotide polymorphisms were genotyped, and methylation levels at several CpGs in exons 1D, 1F and 1H of the GR gene were measured. RESULTS Results show that, beyond the influence of other covariables, NR3C1 single-nucleotide polymorphisms and methylation levels predicted performance in executive functioning and working memory tasks. No significant interactions or mediation relationships were detected. CONCLUSIONS Results suggest that genetic variations and epigenetic regulation of the GR gene are relevant factors influencing cognitive performance in MDD and could emerge as significant biomarkers and therapeutic targets in mood disorders and other stress-related disorders.
Collapse
Affiliation(s)
- Alex Ferrer
- Department of Mental Health, Parc Taulí Hospital Universitari, Sabadell, Catalonia, Spain; Institut de Investigació i Innovació Parc Taulí (I3PT), Sabadell, Catalonia, Spain; Escoles Universitàries Gimbernat, Universitat Autònoma de Barcelona, Barcelona, Catalonia, Spain
| | - Javier Labad
- Institut de Investigació i Innovació Parc Taulí (I3PT), Sabadell, Catalonia, Spain; Department of Mental Health, Consorci Sanitari del Maresme, Mataró, Catalonia, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Neus Salvat-Pujol
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain; Department of Psychiatry, Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL), Neurosciences Group - Psychiatry and Mental Health, Barcelona, Catalonia, Spain; Hospital Universitari Institut Pere Mata, IISPV, Universitat Rovira i Virgili, Reus, Catalonia, Spain
| | - Mikel Urretavizcaya
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain; Department of Psychiatry, Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL), Neurosciences Group - Psychiatry and Mental Health, Barcelona, Catalonia, Spain; Department of Clinical Sciences, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Roser Obrador-Font
- Department of Psychiatry, Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL), Neurosciences Group - Psychiatry and Mental Health, Barcelona, Catalonia, Spain
| | - José M Menchón
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain; Department of Psychiatry, Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL), Neurosciences Group - Psychiatry and Mental Health, Barcelona, Catalonia, Spain; Department of Clinical Sciences, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Diego Palao
- Department of Mental Health, Parc Taulí Hospital Universitari, Sabadell, Catalonia, Spain; Institut de Investigació i Innovació Parc Taulí (I3PT), Sabadell, Catalonia, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain; Department of Psychiatry and Legal Medicine, Universitat Autònoma de Barcelona, Catalonia, Spain
| | - Carles Soriano-Mas
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain; Department of Psychiatry, Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL), Neurosciences Group - Psychiatry and Mental Health, Barcelona, Catalonia, Spain; Department of Social and Quantitative Psychology, Universitat de Barcelona, Catalonia, Spain
| | - Javier Costas
- Instituto de Investigación Sanitaria (IDIS) de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Galicia, A Coruña, Spain
| | - Marta Barrachina
- Department of Psychiatry, Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL), Neurosciences Group - Psychiatry and Mental Health, Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Ángel Carracedo
- Grupo de Medicina Xenómica, Universidade de Santiago de Compostela, Centro Nacional de Genotipado - Instituto Carlos III, Santiago de Compostela, A Coruña, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Virginia Soria
- Department of Mental Health, Parc Taulí Hospital Universitari, Sabadell, Catalonia, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain; Department of Psychiatry, Bellvitge University Hospital, Bellvitge Biomedical Research Institute (IDIBELL), Neurosciences Group - Psychiatry and Mental Health, Barcelona, Catalonia, Spain; Department of Psychiatry and Legal Medicine, Universitat Autònoma de Barcelona, Catalonia, Spain.
| |
Collapse
|
12
|
Milosavljevic S, Piroli MV, Sandago EJ, Piroli GG, Smith HH, Beggiato S, Frizzell N, Pocivavsek A. Parental kynurenine 3-monooxygenase genotype in mice directs sex-specific behavioral outcomes in offspring. Biol Sex Differ 2025; 16:22. [PMID: 40176166 PMCID: PMC11967062 DOI: 10.1186/s13293-025-00703-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 03/13/2025] [Indexed: 04/04/2025] Open
Abstract
BACKGROUND Disruptions in brain development can impact behavioral traits and increase the risk of neurodevelopmental conditions such as autism spectrum disorder, attention-deficit/hyperactivity disorder (ADHD), schizophrenia, and bipolar disorder, often in sex-specific ways. Dysregulation of the kynurenine pathway (KP) of tryptophan metabolism has been implicated in cognitive and neurodevelopmental disorders. Increased brain kynurenic acid (KYNA), a neuroactive metabolite implicated in cognition and sleep homeostasis, and variations in the Kmo gene, which encodes kynurenine 3-monooxygenase (KMO), have been identified in these patients. We hypothesize that parental Kmo genetics influence KP biochemistry, sleep behavior and brain energy demands, contributing to impairments in cognition and sleep in offspring through the influence of parental genotype and genetic nurture mechanisms. METHODS A mouse model of partial Kmo deficiency, Kmo heterozygous (HET-Kmo+/-), was used to examine brain KMO activity, KYNA levels, and sleep behavior in HET-Kmo+/- compared to wild-type control (WT-Control) mice. Brain mitochondrial respiration was assessed, and KP metabolites and corticosterone levels were measured in breast milk. Behavioral assessments were conducted on wild-type offspring from two parental groups: (i) WT-Control from WT-Control parents, (ii) wild-type Kmo (WT-Kmo+/+) from Kmo heterozygous parents (HET-Kmo+/-). All mice were C57Bl/6J background strain. Adult female and male offspring underwent behavioral testing for learning, memory, anxiety-like behavior and sleep-wake patterns. RESULTS HET-Kmo+/- mice exhibited reduced brain KMO activity, increased KYNA levels, and disrupted sleep architecture and electroencephalogram (EEG) power spectra. Mitochondrial respiration (Complex I and Complex II activity) and electron transport chain protein levels were impaired in the hippocampus of HET-Kmo+/- females. Breast milk from HET-Kmo+/- mothers increased kynurenine exposure during lactation but corticosterone levels were unchanged. Compared to WT-Control offspring, WT-Kmo+/+ females showed impaired spatial learning, heightened anxiety, reduced sleep and abnormal EEG spectral power. WT-Kmo+/+ males had deficits in reversal learning but no sleep disturbances or anxiety-like behaviors. CONCLUSIONS These findings suggest that Kmo deficiency impacts KP biochemistry, sleep behavior, and brain mitochondrial function. Even though WT-Kmo+/+ inherit identical genetic material as WT-Control, their development might be shaped by the parent's physiology, behavior, or metabolic state influenced by their Kmo genotype, leading to phenotypic sex-specific differences in offspring.
Collapse
Affiliation(s)
- Snezana Milosavljevic
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Building 1, D26, 6311 Garners Ferry Rd, Columbia, SC, 29209, USA
| | - Maria V Piroli
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Building 1, D26, 6311 Garners Ferry Rd, Columbia, SC, 29209, USA
| | - Emma J Sandago
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Building 1, D26, 6311 Garners Ferry Rd, Columbia, SC, 29209, USA
| | - Gerardo G Piroli
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Building 1, D26, 6311 Garners Ferry Rd, Columbia, SC, 29209, USA
| | - Holland H Smith
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Building 1, D26, 6311 Garners Ferry Rd, Columbia, SC, 29209, USA
| | - Sarah Beggiato
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Norma Frizzell
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Building 1, D26, 6311 Garners Ferry Rd, Columbia, SC, 29209, USA
| | - Ana Pocivavsek
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Building 1, D26, 6311 Garners Ferry Rd, Columbia, SC, 29209, USA.
| |
Collapse
|
13
|
Abdolmaleky HM, Nohesara S, Zhou JR, Thiagalingam S. Epigenetics in evolution and adaptation to environmental challenges: pathways for disease prevention and treatment. Epigenomics 2025; 17:317-333. [PMID: 39948759 PMCID: PMC11970782 DOI: 10.1080/17501911.2025.2464529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 02/04/2025] [Indexed: 04/02/2025] Open
Abstract
Adaptation to challenging environmental conditions is crucial for the survival/fitness of all organisms. Alongside genetic mutations that provide adaptive potential during environmental challenges, epigenetic modifications offer dynamic, reversible, and rapid mechanisms for regulating gene expression in response to environmental changes in both evolution and daily life, without altering DNA sequences or relying on accidental favorable mutations. The widespread conservation of diverse epigenetic mechanisms - like DNA methylation, histone modifications, and RNA interference across diverse species, including plants - underscores their significance in evolutionary biology. Remarkably, environmentally induced epigenetic alterations are passed to daughter cells and inherited transgenerationally through germline cells, shaping offspring phenotypes while preserving adaptive epigenetic memory. Throughout anthropoid evolution, epigenetic modifications have played crucial roles in: i) suppressing transposable elements and viral genomes intruding into the host genome; ii) inactivating one of the X chromosomes in female cells to balance gene dosage; iii) genetic imprinting to ensure expression from one parental allele; iv) regulating functional alleles to compensate for dysfunctional ones; and v) modulating the epigenome and transcriptome in response to influence from the gut microbiome among other functions. Understanding the interplay between environmental factors and epigenetic processes may provide valuable insights into developmental plasticity, evolutionary dynamics, and disease susceptibility.
Collapse
Affiliation(s)
- Hamid Mostafavi Abdolmaleky
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Medicine, Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Shabnam Nohesara
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Jin-Rong Zhou
- Nutrition/Metabolism Laboratory, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Sam Thiagalingam
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| |
Collapse
|
14
|
Hasegawa R, Nakaya K, Kanazawa M, Fukudo S. Corticotropin-releasing hormone receptor-1 antagonist attenuates visceral hypersensitivity induced by trinitrobenzene sulfonic acid colitis and maternal separation in rats. Biopsychosoc Med 2025; 19:5. [PMID: 40155981 PMCID: PMC11951537 DOI: 10.1186/s13030-025-00324-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 01/21/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND The prevailing paradigm for the etiology of irritable bowel syndrome is that transient noxious events lead to long-lasting sensitization of the neural pain circuit, despite complete resolution of the initiating event. In this study, we tested the hypotheses that (1) the combination of maternal separation (MS) and previous colorectal inflammation induces extensive visceral hypersensitivity in rats and (2) visceral hypersensitivity induced by maternal separation and previous colorectal inflammation in rats is mediated via the corticotropin-releasing hormone receptor-1 (CRH-R1) pathway. METHODS Male rat pups were separated from their dams from postnatal day 2 to postnatal day 21. Acute colitis was induced by colorectal administration of trinitrobenzene sulfonic acid (TNBS) or vehicle on postnatal day 8. On postnatal day 50, the visceromotor response was evaluated by electromyography of the abdominal muscle in response to graded (10-80 mmHg) and phasic colorectal distention (CRD) one time. The same experiments were repeated after administration of the selective CRH-R1 antagonist CP-154,526 (20 mg/kg) or vehicle at 45 min before CRD. RESULTS Compared with control rats, visceral perception was increased in MS + TNBS rats. MS + TNBS rats showed a significantly larger visceromotor response to phasic CRD with 40 mmHg, 60 mmHg, and 80 mmHg. Compared with vehicle administration in MS + TNBS rats, administration of CP-154,526 significantly attenuated this visceromotor response to CRD with 40 mmHg, 60 mmHg, and 80 mmHg. CONCLUSIONS These findings suggest that the combination of previous colitis and early life stress induce visceral hypersensitivity, and that the CRH-R1 pathway may play a role in this sensitization.
Collapse
Affiliation(s)
- Ryoko Hasegawa
- Department of Behavioral Medicine, Tohoku University Graduate School of Medicine, 2-1 Seiryo, Aoba, Sendai, 980-8575, Japan
| | - Kumi Nakaya
- Department of Behavioral Medicine, Tohoku University Graduate School of Medicine, 2-1 Seiryo, Aoba, Sendai, 980-8575, Japan
- Division of Epidemiology, School of Public Health, Tohoku University Graduate School of Medicine, Sendai, Japan
- Division of Personalized Prevention and Epidemiology Department of Preventive Medicine and Epidemiology Tohoku Medical Megabank Organization, Sendai, Japan
| | - Motoyori Kanazawa
- Department of Behavioral Medicine, Tohoku University Graduate School of Medicine, 2-1 Seiryo, Aoba, Sendai, 980-8575, Japan
- Department of Psychosomatic Medicine, Tohoku University Hospital, Sendai, Japan
| | - Shin Fukudo
- Department of Behavioral Medicine, Tohoku University Graduate School of Medicine, 2-1 Seiryo, Aoba, Sendai, 980-8575, Japan.
- Department of Psychosomatic Medicine, Tohoku University Hospital, Sendai, Japan.
- Research Center for Accelerator and Radioisotope Science, Tohoku University, Sendai, Japan.
- Department of Psychosomatic Medicine, Japanese Red Cross Ishinomaki Hospital, Ishinomaki, Japan.
| |
Collapse
|
15
|
Banushi B, Collova J, Milroy H. Epigenetic Echoes: Bridging Nature, Nurture, and Healing Across Generations. Int J Mol Sci 2025; 26:3075. [PMID: 40243774 PMCID: PMC11989090 DOI: 10.3390/ijms26073075] [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: 03/06/2025] [Revised: 03/19/2025] [Accepted: 03/25/2025] [Indexed: 04/18/2025] Open
Abstract
Trauma can impact individuals within a generation (intragenerational) and future generations (transgenerational) through a complex interplay of biological and environmental factors. This review explores the epigenetic mechanisms that have been correlated with the effects of trauma across generations, including DNA methylation, histone modifications, and non-coding RNAs. These mechanisms can regulate the expression of stress-related genes (such as the glucocorticoid receptor (NR3C1) and FK506 binding protein 5 (FKBP5) gene), linking trauma to biological pathways that may affect long-term stress regulation and health outcomes. Although research using model organisms has elucidated potential epigenetic mechanisms underlying the intergenerational effects of trauma, applying these findings to human populations remains challenging due to confounding variables, methodological limitations, and ethical considerations. This complexity is compounded by difficulties in establishing causality and in disentangling epigenetic influences from shared environmental factors. Emerging therapies, such as psychedelic-assisted treatments and mind-body interventions, offer promising avenues to address both the psychological and potential epigenetic aspects of trauma. However, translating these findings into effective interventions will require interdisciplinary methods and culturally sensitive approaches. Enriched environments, cultural reconnection, and psychosocial interventions have shown the potential to mitigate trauma's impacts within and across generations. By integrating biological, social, and cultural perspectives, this review highlights the critical importance of interdisciplinary frameworks in breaking cycles of trauma, fostering resilience, and advancing comprehensive healing across generations.
Collapse
Affiliation(s)
- Blerida Banushi
- School of Indigenous Studies, The University of Western Australia, Crawley, WA 6009, Australia; (J.C.); (H.M.)
| | | | | |
Collapse
|
16
|
Adar O, Shakargy JD, Ilan Y. The Constrained Disorder Principle: Beyond Biological Allostasis. BIOLOGY 2025; 14:339. [PMID: 40282204 PMCID: PMC12025142 DOI: 10.3390/biology14040339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 03/24/2025] [Accepted: 03/24/2025] [Indexed: 04/29/2025]
Abstract
The constrained disorder principle (CDP) defines complex biological systems based on inherent variability. Allostasis refers to the physiological processes that help maintain stability in response to changing environmental demands. Allostatic load describes the cumulative wear and tear on the body resulting from prolonged exposure to stress, and it has been suggested to mediate the relationship between stress and disease. This study presents the concepts of CDP and allostasis while discussing their similarities and differences. We reviewed the current literature on the potential benefits of introducing controlled doses of biological noise into interventions, which may enhance the effectiveness of therapies. The paper highlights the promising role of variability provided by a CDP-based second-generation artificial intelligence system in improving health outcomes.
Collapse
Affiliation(s)
- Ofek Adar
- Faculty of Medicine, Hebrew University, Jerusalem 9112001, Israel; (O.A.); (J.D.S.)
- Department of Medicine, Hadassah Medical Center, Jerusalem 9112001, Israel
| | - Josef Daniel Shakargy
- Faculty of Medicine, Hebrew University, Jerusalem 9112001, Israel; (O.A.); (J.D.S.)
- Department of Medicine, Hadassah Medical Center, Jerusalem 9112001, Israel
| | - Yaron Ilan
- Faculty of Medicine, Hebrew University, Jerusalem 9112001, Israel; (O.A.); (J.D.S.)
- Department of Medicine, Hadassah Medical Center, Jerusalem 9112001, Israel
| |
Collapse
|
17
|
Bottaccioli AG, Bologna M, Bottaccioli F. Rethinking Depression-Beyond Neurotransmitters: An Integrated Psychoneuroendocrineimmunology Framework for Depression's Pathophysiology and Tailored Treatment. Int J Mol Sci 2025; 26:2759. [PMID: 40141399 PMCID: PMC11943243 DOI: 10.3390/ijms26062759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/06/2025] [Accepted: 03/13/2025] [Indexed: 03/28/2025] Open
Abstract
It is known that the effectiveness of drug treatment for depression, ammine deficit based, is largely unsatisfactory. In this review, we examine the proposal of a precision therapy has emerged and has received a strong push by the identification of the role of inflammation in depression. However, precision psychiatry risks being caught in the reductionist trap of searching for the molecular switch that resets the whole system and switches off the disease. This is an illusion since the human being is complex and depression is a systemic and variable disorder. In this study, we show the inadequacy of the reductionist paradigm, and, at the same time, illustrate the superiority of the systemic paradigm centered on psychoneuroendocrineimmunology (PNEI). According to the PNEI paradigm, depression is a disease of the whole human being, caused by different sources working together: psychological, biological, and behavioral. This means knowing the biological and psychological history of the subject, identifying relational and biological crisis factors, and building personalized treatments targeting those factors with the tools of medicine and psychology, which are not reducible to the combination of drugs and psychotherapy. Our proposal presents a paradigm shift that is both theoretical and practical, which enables clinicians to assess patients experiencing depression in a unified way and treat them in an integrated manner.
Collapse
Affiliation(s)
- Anna Giulia Bottaccioli
- Department of Oncohematology, Clinical Psychology Graduated Course, University of Milan, I-20122 Milan, Italy
- Società Italiana di Psiconeuroendocrinoimmunologia, I-00195 Rome, Italy; (M.B.); (F.B.)
| | - Mauro Bologna
- Società Italiana di Psiconeuroendocrinoimmunologia, I-00195 Rome, Italy; (M.B.); (F.B.)
- Department of Medicine, Health, Life and Environment, University of L’Aquila, I-67100 L’Aquila, Italy
| | - Francesco Bottaccioli
- Società Italiana di Psiconeuroendocrinoimmunologia, I-00195 Rome, Italy; (M.B.); (F.B.)
- Post-Graduated Course of Psychoneuroendocrineimmunology, Humanitas University Consortium Rome, I-00193 Rome, Italy
| |
Collapse
|
18
|
Tonna M, Borrelli DF, Marchesi C, Gerra MC, Dallabona C. Childhood obsessive-compulsive disorder, epigenetics, and heterochrony: An evolutionary and developmental approach. Dev Psychopathol 2025:1-15. [PMID: 40099440 DOI: 10.1017/s0954579425000124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Childhood obsessive-compulsive disorder (OCD) stems from a bunch of restricted and repetitive behaviors, which are part of normal behavioral repertoire up to the age of 7. The persistence of compulsive-like behaviors after that age is often associated with unique comorbidity patterns, which are age-at-onset dependent and reflect different developmental stages. In particular, OCD synchronically co-occurs with a broad constellation of neurodevelopmental disorders, whereas diachronically it is related to an increased risk of major adult psychoses. Moreover, OCD is associated with trait-like sensory phenomena, suggesting a common disrupted sensorimotor grounding.The present study is aimed at exploring the hypothesis that this specific temporal and comorbidity OCD profile may be due to a developmental heterochronic mechanism of delay in attenuation of ontogenetically early behavioral patterns. The developmental shift of highly evolutionarily conserved behavioral phenotypes might be regulated by epigenetic changes induced by different conditions of sensory unbalance. This evolutionary and developmental model allows capturing childhood OCD in light of the ultimate causes of ritual behavior throughout phylogeny, namely its "homeostatic" function over conditions of unpredictability. Moreover, it may have important clinical implications, as OCD symptoms could represent putative biomarkers of early divergent developmental trajectories, with a pathoplastic effect on course and outcome.
Collapse
Affiliation(s)
- Matteo Tonna
- Department of Medicine and Surgery, Psychiatric Unit, University of Parma, Parma, Italy
- Department of Mental Health, Local Health Service, Parma, Italy
| | - Davide Fausto Borrelli
- Department of Medicine and Surgery, Psychiatric Unit, University of Parma, Parma, Italy
- Department of Mental Health, Local Health Service, Piacenza, Italy
| | - Carlo Marchesi
- Department of Medicine and Surgery, Psychiatric Unit, University of Parma, Parma, Italy
- Department of Mental Health, Local Health Service, Parma, Italy
| | - Maria Carla Gerra
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, PR, Italy
| | - Cristina Dallabona
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, PR, Italy
| |
Collapse
|
19
|
Villagrán-Silva F, Loren P, Sandoval C, Lanas F, Salazar LA. Circulating microRNAs as Potential Biomarkers of Overweight and Obesity in Adults: A Narrative Review. Genes (Basel) 2025; 16:349. [PMID: 40149500 PMCID: PMC11942292 DOI: 10.3390/genes16030349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/29/2025] Open
Abstract
In an obesogenic environment, such as the one we have been experiencing in recent decades, epigenetics provides answers to the relationship between hereditary and environmentally acquired patterns that have significantly contributed to the global rise in obesity prevalence. MicroRNA (miRNA) constitutes a diminutive non-coding small RNA molecule, 20 to 24 nucleotides in length, that functions as a regulator of gene regulation at the post-translational level. Circulating miRNAs (c-miRNAs) have been detected in multiple body fluids, including blood, plasma, serum, saliva, milk from breastfeeding mothers, and urine. These molecules hold significant therapeutic value and serve as extracellular biomarkers in metabolic diseases. They aid in the diagnosis and tracking of therapy responses, as well as dietary and physical habit modifications. Researchers have studied c-miRNAs as potential biomarkers for diagnosing and characterizing systemic diseases in people of all ages and backgrounds since then. These conditions encompass dyslipidemia, type 2 diabetes mellitus (T2DM), cardiovascular risk, metabolic syndrome, cardiovascular diseases, and obesity. This review therefore analyzes the usefulness of c-miRNAs as therapeutic markers over the past decades. It also provides an update on c-miRNAs associated with general obesity and overweight, as well as with the most prevalent pathologies in the adult population. It also examines the effect of different nutritional approaches and physical activity regarding the activity of miRNAs in circulation in adults with overweight or general obesity. All of this is done with the aim of evaluating their potential use as biomarkers in various research contexts related to overweight and obesity in adults.
Collapse
Affiliation(s)
- Francisca Villagrán-Silva
- Doctoral Program in Morphological Sciences, Faculty of Medicine, Universidad de la Frontera, Temuco 4811230, Chile;
- Center of Molecular Biology and Pharmacogenetics, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4811230, Chile; (P.L.); (F.L.)
| | - Pía Loren
- Center of Molecular Biology and Pharmacogenetics, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4811230, Chile; (P.L.); (F.L.)
| | - Cristian Sandoval
- Escuela de Tecnología Médica, Facultad de Salud, Universidad Santo Tomás, Los Carreras 753, Osorno 5310431, Chile;
- Department of Internal Medicine, Faculty of Medicine, Universidad de La Frontera, Temuco 4811230, Chile
| | - Fernando Lanas
- Center of Molecular Biology and Pharmacogenetics, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4811230, Chile; (P.L.); (F.L.)
- Department of Internal Medicine, Faculty of Medicine, Universidad de La Frontera, Temuco 4811230, Chile
| | - Luis A. Salazar
- Center of Molecular Biology and Pharmacogenetics, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4811230, Chile; (P.L.); (F.L.)
| |
Collapse
|
20
|
Chen H, Xu R, Wang J, Gao F, Lv Y, Li X, Li F, Zhao J, Zhang X, Wang J, Du R, Shi Y, Yu H, Ding S, Li W, Xiong J, Zheng J, Zhao L, Gao XY, Wang ZH. Maternal behavior promotes resilience to adolescent stress in mice through a microglia-neuron axis. Nat Commun 2025; 16:2333. [PMID: 40057602 PMCID: PMC11890579 DOI: 10.1038/s41467-025-57810-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 02/27/2025] [Indexed: 05/13/2025] Open
Abstract
Early life experience modulates resilience to stress in later life. Previous research implicated maternal care as a key mediator of behavioral responses to the adversity in adolescence, but details of molecular mechanisms remain elusive. Here, we show social stress activates transcription factor C/EBPβ in mPFC neurons of adolescent mice, which transcriptionally upregulates Dnm1l and promotes mitochondrial dysfunction, thereby conferring stress susceptibility in adolescent mice. Moreover, different maternal separation differentially regulates adolescent stress susceptibility. Mechanistically, this differential effect depends on maternal behavior-stimulated IGF-1, which inhibits neuronal C/EBPβ through mTORC1-induced C/EBPβ-LIP translation. Furthermore, we identify maternal behavior-stimulated IGF-1 is mainly released from mPFC microglia. Notably, increased maternal care under an environmental enrichment condition or maternal behavior impairment induced by repeated MPOAEsr1+ cells inhibition in dams prevents or promotes stress susceptibility via microglial-to-neuronal IGF-1-C/EBPβ-DRP1 signaling. In this work, these findings have unveiled molecular mechanisms by which maternal behavior promotes stress resilience in adolescents.
Collapse
Affiliation(s)
- Hongyu Chen
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ruifeng Xu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianhao Wang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan, China
| | - Feng Gao
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yida Lv
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiang Li
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan, China
| | - Fang Li
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan, China
| | - Junqin Zhao
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xi Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jiabei Wang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ruicheng Du
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yuke Shi
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hang Yu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shuai Ding
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wenxin Li
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jing Xiong
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jie Zheng
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing, China
| | - Liang Zhao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin-Ya Gao
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, China
- Laboratory of Neurology, Henan Provincial People's Hospital, Zhengzhou, China
| | - Zhi-Hao Wang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China.
- Center for Neurodegenerative Disease Research, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
21
|
Abouseta N, Gomaa N, Tassi A, Dixon SJ, Singh K, Pani SC. Profiling mRNA encoding glucocorticoid receptor α in saliva: Relationship to hair cortisol levels in individuals aged 15-25 years. Arch Oral Biol 2025; 171:106158. [PMID: 39672058 DOI: 10.1016/j.archoralbio.2024.106158] [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: 06/29/2024] [Revised: 11/17/2024] [Accepted: 12/03/2024] [Indexed: 12/15/2024]
Abstract
OBJECTIVE We assessed levels of mRNA encoding two glucocorticoid receptor (GR) isoforms (GRα and GRβ) in saliva and examined their relationship with hair cortisol levels and dental caries experience. DESIGN Adolescents and young adults were assessed for dental caries experience, and hair cortisol was measured by ELISA. RNA was extracted from whole saliva using TRIzol, followed by quantitative real-time PCR analysis of GRα, GRβ, and glyceraldehyde 3-phosphate dehydrogenase (GAPDH). RESULTS GRβ mRNA was not detectable in most samples, whereas GRα mRNA was observed in all samples. There were significantly lower levels of GRα mRNA in individuals with elevated hair cortisol levels than in those with normal cortisol levels. Levels of GRα mRNA did not differ significantly in individuals with dental caries experience compared to individuals with no caries experience. CONCLUSIONS We identified and quantified mRNA encoding GRα in saliva. Its levels were inversely associated with hair cortisol (a marker of chronic stress). Although caries experience was associated with hair cortisol levels, there was no significant association between GRα levels and caries experience. Chronic stress has been proposed to be associated with reduced expression of GRα and this association appears to hold for GRα mRNA levels in saliva.
Collapse
Affiliation(s)
- Naima Abouseta
- Department of Medical Biophysics, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada.
| | - Noha Gomaa
- Children's Health Research Institute, Lawson Health Research Institute, London, ON, Canada; Oral Diagnostic Sciences, and Epidemiology and Biostatistics, Schulich School of Medicine & Dentistry, University of Western Ontario, Lawson Health Research Institute, London, ON, Canada.
| | - Ali Tassi
- Graduate Orthodontics and Dentofacial Orthopaedics, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada.
| | - S Jeffrey Dixon
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada.
| | - Krishna Singh
- Department of Medical Biophysics, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada.
| | - Sharat C Pani
- Schulich Dentistry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada.
| |
Collapse
|
22
|
Hanson T, Spencer S, Harker SA, Barry F, Burton P, Beauchemin J, Mennenga SE, Braden BB, D'Sa V, Koinis-Mitchell D, Deoni SC, Lewis CR. Peripheral DNA Methylation of Cortisol- and Serotonin-Related Genes Predicts Hippocampal Volume in a Pediatric Population. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2025; 5:100421. [PMID: 39867566 PMCID: PMC11758844 DOI: 10.1016/j.bpsgos.2024.100421] [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: 07/01/2024] [Revised: 11/08/2024] [Accepted: 11/13/2024] [Indexed: 01/28/2025] Open
Abstract
Background Hippocampal volume increases throughout early development and is an important indicator of cognitive abilities and mental health. However, hippocampal development is highly vulnerable to exposures during development, as seen by smaller hippocampal volume and differential epigenetic programming in genes implicated in mental health. However, few studies have investigated hippocampal volume in relation to the peripheral epigenome across development, and even less is known about potential genetic moderators. Therefore, in this study, we explored relationships between hippocampal volume and peripheral DNA methylation of mental health-related genes, specifically NR3C1, FKBP5, and SLC6A4, throughout early development and whether these associations were moderated by age or genotype. Methods Bilateral hippocampal volume was computed from T2-weighted images through FreeSurfer, and DNA methylation was measured from saliva using the Illumina MethylationEPIC microarray in a pediatric population (N = 248, females = 112, meanage = 5.13 years, SDage = 3.60 years). Results Multiple linear regression and bootstrapping analyses revealed that DNA methylation of NR3C1, FKBP5, and SLC6A4 was associated with hippocampal volume and that these relationships were moderated by age and gene-specific variants. Conclusions These findings support the validity of peripheral DNA methylation profiles for indirectly assessing hippocampal volume and development and underscore the importance of genotype and age considerations in research. Therefore, peripheral epigenetic profiles may be a promising avenue for investigating the impacts of early-life stress on brain structure and subsequent mental health outcomes.
Collapse
Affiliation(s)
- Taena Hanson
- Department of Psychology, Arizona State University, Tempe, Arizona
| | - Sophia Spencer
- Department of Psychology, Arizona State University, Tempe, Arizona
| | | | - Fatoumata Barry
- Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - Phoebe Burton
- Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | | | | | - B. Blair Braden
- College of Health Solutions, Arizona State University, Tempe, Arizona
| | - Viren D'Sa
- Maternal, Newborn, and Child Health Discovery & Tools, Bill & Melinda Gates Foundation; Seattle, Washington; Providence, Rhode Island
| | - Daphne Koinis-Mitchell
- Maternal, Newborn, and Child Health Discovery & Tools, Bill & Melinda Gates Foundation; Seattle, Washington; Providence, Rhode Island
| | - Sean C.L. Deoni
- Maternal, Newborn, and Child Health Discovery & Tools, Bill & Melinda Gates Foundation; Seattle, Washington; Providence, Rhode Island
- Advanced Baby Imaging Laboratory, Rhode Island Hospital, Providence, Rhode Island
| | - Candace R. Lewis
- Department of Psychology, Arizona State University, Tempe, Arizona
- School of Life Sciences, Arizona State University, Tempe, Arizona
- Neurogenomics, Translational Genomics Research Institute, Phoenix, Arizona
| |
Collapse
|
23
|
Bowe C, Thomas C, Mackey P. Perspective to Practice: Theoretical Frameworks Explaining Intergenerational Trauma, Violence, and Maltreatment and Implications for the Therapeutic Response. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2025; 22:321. [PMID: 40238309 PMCID: PMC11942635 DOI: 10.3390/ijerph22030321] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 02/14/2025] [Accepted: 02/16/2025] [Indexed: 04/18/2025]
Abstract
Intergenerational trauma, violence, and maltreatment, in which symptoms or experiences of an ancestor's trauma repeat or otherwise manifest in subsequent generations, presents a weighty societal challenge to which a multiplicity of therapeutic intervention strategies have been applied. Theoretical perspectives are antecedent to clinical and social intervention, informing decisions in both policy and practice. However, these frequently remain subliminal or imperceptible in the discourse, resulting in interventions that remain somewhat dislocated from their theoretical foundations. This narrative review seeks to summarize and discuss each of these theories as they apply to intergenerational trauma, violence, and maltreatment, and to reveal their potential association with specific intervention models or approaches. It positions flexibility between theories and the integration of theories as opportunities to reach new and enhanced understandings and to engender distinctive therapeutic interventions. An enriched understanding of the theories explaining intergenerational trauma, violence, and maltreatment, a deeper appreciation for the pertinence of theory for practice, and an incitement to blend theoretical perspectives in unique ways is, herewith, reached.
Collapse
Affiliation(s)
- Crysta Bowe
- School of Social Work and Arts, Charles Sturt University, Barton, ACT 2600, Australia; (C.T.); (P.M.)
| | | | | |
Collapse
|
24
|
Grillo Balboa J, Colapietro AA, Cantarelli VI, Ponzio MF, Ceol Retamal MN, Pallarés ME, Antonelli MC, Chertoff M. Sex-Specific Outcomes in a Rat Model of Early-Life Stress Due to Adverse Caregiving. Neurotox Res 2025; 43:10. [PMID: 39964605 DOI: 10.1007/s12640-025-00731-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/29/2025] [Accepted: 01/30/2025] [Indexed: 05/03/2025]
Abstract
Early parental care is critical for the development of cortico-limbic circuits regulating stress responses and emotional well-being. Conversely, infant maltreatment can increase susceptibility to mood disorders-such as anxiety and depression-and impair stress-coping abilities. Here, we employed the Scarcity-Adversity Model (SAM) in rats, limiting nesting resources from postnatal days 8-12, to examine its effects on maternal and adult offspring behavior. SAM-exposed mothers exhibited fragmented care and increased violence towards pups. By postpartum day (PPD) 13, maternal fecal corticosterone metabolites (FCM) were elevated, indicating heightened stress. At weaning, SAM dams also showed increased anxiety-like behavior in the Elevated Plus Maze (EPM), suggesting significant emotional alterations. In adulthood, SAM-exposed offspring underwent anxiogenic tests. Both male and female SAM offspring showed increased latency to enter open arms and reduced risk-assessment in the EPM, though females displayed anxiolytic-like behavior in the Light-Dark Box. Male SAM rats had reduced locomotion in the Open Field, earlier onset and increased immobility in the Forced Swim, and increased latency to groom in the Sucrose Splash. When exposed to acute stress, male SAM rats had lower FCM levels, consistent with their passive stress reactivity. These findings confirm SAM induces long-lasting, sex-specific changes in risk-taking, novelty responsiveness, and stress reactions, underscoring the importance of early nurturing in promoting well-being and reducing psychopathological risk.
Collapse
Affiliation(s)
- Jazmín Grillo Balboa
- Laboratorio de Neuroepigenética y Adversidades Tempranas, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis", Facultad de Medicina, Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina
| | - Ailén A Colapietro
- Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis", Facultad de Medicina, Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina
| | - Verónica I Cantarelli
- Facultad de Ciencias Médicas, Instituto de Investigaciones en Ciencias de la Salud (INICSA-CONICET), Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Marina F Ponzio
- Facultad de Ciencias Médicas, Instituto de Investigaciones en Ciencias de la Salud (INICSA-CONICET), Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Marianela N Ceol Retamal
- Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis", Facultad de Medicina, Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina
| | - María E Pallarés
- Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis", Facultad de Medicina, Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina
| | - Marta C Antonelli
- Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis", Facultad de Medicina, Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina
| | - Mariela Chertoff
- Laboratorio de Neuroepigenética y Adversidades Tempranas, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina.
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
25
|
Taylor WW, Korobkova L, Bhinderwala N, Dias BG. Toward Understanding and Halting Legacies of Trauma. Biol Psychiatry 2025:S0006-3223(25)00108-8. [PMID: 39956254 DOI: 10.1016/j.biopsych.2025.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 02/02/2025] [Accepted: 02/10/2025] [Indexed: 02/18/2025]
Abstract
Echoes of natural and anthropogenic stressors not only reverberate within the physiology, biology, and neurobiology of the generation directly exposed to them but also within the biology of future generations. With the intent of understanding this phenomenon, significant efforts have been made to establish how exposure to psychosocial stress, chemicals, over- and undernutrition, and chemosensory experiences exert multigenerational influences. From these studies, we are gaining new appreciation for how negative environmental events experienced by one generation impact future generations. In this review, we first outline the need to operationally define dimensions of negative environmental events in the laboratory and the routes by which the impact of such events are felt through generations. Next, we discuss molecular processes that cause the effects of negative environmental events to be initiated in the exposed generation and then perpetuated across generations. Finally, we discuss how legacies of flourishing can be engineered to halt or reverse multigenerational influences of negative environmental events. In summary, this review synthesizes our current understanding of the concept, causes, and consequences of multigenerational echoes of stress and looks for opportunities to halt them.
Collapse
Affiliation(s)
- William Wesley Taylor
- Neuroscience Graduate Program, University of Southern California, Los Angeles, California; Developmental Neuroscience and Neurogenetics Program, The Saban Research Institute, Los Angeles, California
| | - Laura Korobkova
- Neuroscience Graduate Program, University of Southern California, Los Angeles, California; Developmental Neuroscience and Neurogenetics Program, The Saban Research Institute, Los Angeles, California
| | - Nabeel Bhinderwala
- Developmental Neuroscience and Neurogenetics Program, The Saban Research Institute, Los Angeles, California
| | - Brian George Dias
- Developmental Neuroscience and Neurogenetics Program, The Saban Research Institute, Los Angeles, California; Division of Endocrinology, Diabetes and Metabolism, Children's Hospital Los Angeles, Los Angeles, California; Department of Pediatrics, Keck School of Medicine of the University of Southern California, Los Angeles, California.
| |
Collapse
|
26
|
Vernick J, Martin C, Montelpare W, Dunham AE, Overall KL. Understanding the Influence of Early-Life Stressors on Social Interaction, Telomere Length, and Hair Cortisol Concentration in Homeless Kittens. Animals (Basel) 2025; 15:446. [PMID: 39943216 PMCID: PMC11815723 DOI: 10.3390/ani15030446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/14/2025] [Accepted: 02/04/2025] [Indexed: 02/16/2025] Open
Abstract
The early postnatal period is a critical neurodevelopmental stage characterized by rapid neural maturation and is adversely affected by early-life stressors. This study explored the behavioural, physiological, and epigenetic consequences of early-life stress in a population of homeless rescue kittens. This longitudinal study included 50 kittens rescued and placed into foster care by the Prince Edward Island Humane Society. They underwent behavioural testing at 8, 10, and 12 weeks of age. Hair cortisol concentration was measured at 8 weeks and served as a physiological marker of the previous 3 months' cumulative stress response, which, for these kittens, included the late gestation period. A blood sample for relative telomere length measurement was taken at 10-12 weeks to estimate epigenetic changes as young kittens. Data were analyzed with respect to age and performance in all repeated measures tests, status as a stray or a surrender, and the presence of the dam in their foster homes. As expected, the performance of kittens in all tests changed over the 5 weeks of testing. Kittens separated from their mothers exhibited significantly higher hair cortisol concentrations (p = 0.02) and elongated relative telomere lengths (p = 0.04). No correlation was found between hair cortisol concentration and relative telomere lengths (p = 0.99). These results support the need for further study on the effects of epigenetics and early-life stress, both in kittens and across species.
Collapse
Affiliation(s)
- Jennifer Vernick
- Department of Health Management, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, PE C1A 4P3, Canada
| | - Chelsea Martin
- Department of Microbiology and Pathology, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, PE C1A 4P3, Canada;
| | - William Montelpare
- Department of Applied Human Sciences, Faculty of Science and Faculty of Nursing, University of Prince Edward Island, Charlottetown, PE C1A 4P3, Canada;
| | - Arthur E. Dunham
- Biology Department, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Karen L. Overall
- Department of Health Management, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, PE C1A 4P3, Canada
| |
Collapse
|
27
|
Neri M, Brovelli A, Castro S, Fraisopi F, Gatica M, Herzog R, Mediano PAM, Mindlin I, Petri G, Bor D, Rosas FE, Tramacere A, Estarellas M. A Taxonomy of Neuroscientific Strategies Based on Interaction Orders. Eur J Neurosci 2025; 61:e16676. [PMID: 39906974 DOI: 10.1111/ejn.16676] [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: 08/19/2024] [Revised: 11/15/2024] [Accepted: 12/29/2024] [Indexed: 02/06/2025]
Abstract
In recent decades, neuroscience has advanced with increasingly sophisticated strategies for recording and analysing brain activity, enabling detailed investigations into the roles of functional units, such as individual neurons, brain regions and their interactions. Recently, new strategies for the investigation of cognitive functions regard the study of higher order interactions-that is, the interactions involving more than two brain regions or neurons. Although methods focusing on individual units and their interactions at various levels offer valuable and often complementary insights, each approach comes with its own set of limitations. In this context, a conceptual map to categorize and locate diverse strategies could be crucial to orient researchers and guide future research directions. To this end, we define the spectrum of orders of interaction, namely, a framework that categorizes the interactions among neurons or brain regions based on the number of elements involved in these interactions. We use a simulation of a toy model and a few case studies to demonstrate the utility and the challenges of the exploration of the spectrum. We conclude by proposing future research directions aimed at enhancing our understanding of brain function and cognition through a more nuanced methodological framework.
Collapse
Affiliation(s)
- Matteo Neri
- Institut de Neurosciences de la Timone, Aix-Marseille Université, UMR 7289 CNRS, Marseille, France
| | - Andrea Brovelli
- Institut de Neurosciences de la Timone, Aix-Marseille Université, UMR 7289 CNRS, Marseille, France
| | - Samy Castro
- Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR 7364, Strasbourg, France
- Institut de Neurosciences Des Systèmes (INS), Aix-Marseille Université, UMR 1106, Marseille, France
| | - Fausto Fraisopi
- Institute for Advanced Study, Aix-Marseille University, Marseille, France
| | - Marilyn Gatica
- NPLab, Network Science Institute, Northeastern University London, London, UK
| | - Ruben Herzog
- DreamTeam, Paris Brain Institute (ICM), Paris, France
| | - Pedro A M Mediano
- Department of Computing, Imperial College London, London, UK
- Division of Psychology and Language Sciences, University College London, London, UK
| | - Ivan Mindlin
- DreamTeam, Paris Brain Institute (ICM), Paris, France
- PICNIC lab, Paris Brain Institute (ICM), Paris, France
| | - Giovanni Petri
- NPLab, Network Science Institute, Northeastern University London, London, UK
- Department of Physics, Northeastern University, Boston, Massachusetts, USA
- NPLab, CENTAI Institute, Turin, Italy
| | - Daniel Bor
- Department of Psychology, School of Biological and Behavioural Sciences, Queen Mary University of London, London, UK
- Department of Psychology, University of Cambridge, Cambridge, UK
| | - Fernando E Rosas
- Sussex Centre for Consciousness Science and Sussex AI, Department of Informatics, University of Sussex, Brighton, UK
- Center for Psychedelic Research and Centre for Complexity Science, Department of Brain Science, Imperial College London, London, UK
- Centre for Eudaimonia and Human Flourishing, University of Oxford, Oxford, UK
- Principles of Intelligent Behavior in Biological and Social Systems (PIBBSS), Prague, Czechia
| | - Antonella Tramacere
- Department of Philosophy, Communication and Performing Arts, Roma Tre University, Rome, Italy
| | - Mar Estarellas
- Department of Psychology, School of Biological and Behavioural Sciences, Queen Mary University of London, London, UK
- Department of Psychology, University of Cambridge, Cambridge, UK
| |
Collapse
|
28
|
Iannuzzi V, Narboux-Nême N, Lehoczki A, Levi G, Giuliani C. Stay social, stay young: a bioanthropological outlook on the processes linking sociality and ageing. GeroScience 2025; 47:721-744. [PMID: 39527178 PMCID: PMC11872968 DOI: 10.1007/s11357-024-01416-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
In modern human societies, social interactions and pro-social behaviours are associated with better individual and collective health, reduced mortality, and increased longevity. Conversely, social isolation is a predictor of shorter lifespan. The biological processes through which sociality affects the ageing process, as well as healthspan and lifespan, are still poorly understood. Unveiling the physiological, neurological, genomic, epigenomic, and evolutionary mechanisms underlying the association between sociality and longevity may open new perspectives to understand how lifespan is determined in a broader socio/evolutionary outlook. Here we summarize evidence showing how social dynamics can shape the evolution of life history traits through physiological and genetic processes directly or indirectly related to ageing and lifespan. We start by reviewing theories of ageing that incorporate social interactions into their model. Then, we address the link between sociality and lifespan from two separate points of view: (i) considering evidences from comparative evolutionary biology and bioanthropology that demonstrates how sociality contributes to natural variation in lifespan over the course of human evolution and among different human groups in both pre-industrial and post-industrial society, and (ii) discussing the main physiological, neurological, genetic, and epigenetic molecular processes at the interface between sociality and ageing. We highlight that the exposure to chronic social stressors deregulates neurophysiological and immunological pathways and promotes accelerated ageing and thereby reducing lifespan. In conclusion, we describe how sociality and social dynamics are intimately embedded in human biology, influencing healthy ageing and lifespan, and we highlight the need to foster interdisciplinary approaches including social sciences, biological anthropology, human ecology, physiology, and genetics.
Collapse
Affiliation(s)
- Vincenzo Iannuzzi
- Laboratory of Molecular Anthropology & Centre for Genome Biology, Department of Biological, Geological and Environmental Sciences, University of Bologna, Via Selmi 3, 40126, Bologna, Italy
| | - Nicolas Narboux-Nême
- Physiologie Moléculaire Et Adaptation, CNRS UMR7221, Département AVIV, Muséum National d'Histoire Naturelle, Paris, France
| | - Andrea Lehoczki
- Doctoral College, Health Sciences Program, Semmelweis University, Budapest, Hungary
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Giovanni Levi
- Physiologie Moléculaire Et Adaptation, CNRS UMR7221, Département AVIV, Muséum National d'Histoire Naturelle, Paris, France.
| | - Cristina Giuliani
- Laboratory of Molecular Anthropology & Centre for Genome Biology, Department of Biological, Geological and Environmental Sciences, University of Bologna, Via Selmi 3, 40126, Bologna, Italy.
| |
Collapse
|
29
|
Cheng Y, Miller MJ, Lei F. Molecular Innovations Shaping Beak Morphology in Birds. Annu Rev Anim Biosci 2025; 13:99-119. [PMID: 39546421 DOI: 10.1146/annurev-animal-030424-074906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
The beak, a pivotal evolutionary trait characterized by high morphological diversity and plasticity, has enabled birds to survive mass extinction events and subsequently radiate into diverse ecological niches worldwide. This remarkable ecological adaptability underscores the importance of uncovering the molecular mechanisms shaping avian beak morphology, particularly benefiting from the rapidly advancing archives of genomics and epigenomics. We review the latest advancements in understanding how genetic and epigenetic innovations control or regulate beak development and drive beak morphological adaptation and diversification over the past two decades. We conclude with several recommendations for future endeavors, expanding to more bird lineages, with a focus on beak shape and the lower beak, and conducting functional experiments. By directing research efforts toward these aspects and integrating advanced omics techniques, the complex molecular mechanisms involved in avian beak evolution and morphogenesis will be deeply interpreted.
Collapse
Affiliation(s)
- Yalin Cheng
- Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing, China;
- College of Life Science, Hebei University, Baoding, China
| | | | - Fumin Lei
- University of Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing, China;
| |
Collapse
|
30
|
Guo H, Gao J, Gong L, Wang Y. Multi-omics analysis reveals novel causal pathways in psoriasis pathogenesis. J Transl Med 2025; 23:100. [PMID: 39844246 PMCID: PMC11752815 DOI: 10.1186/s12967-025-06099-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 01/08/2025] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND To elucidate the genetic and molecular mechanisms underlying psoriasis by employing an integrative multi-omics approach, using summary-data-based Mendelian randomization (SMR) to infer causal relationships among DNA methylation, gene expression, and protein levels in relation to psoriasis risk. METHODS We conducted SMR analyses integrating genome-wide association study (GWAS) summary statistics with methylation quantitative trait loci (mQTL), expression quantitative trait loci (eQTL), and protein quantitative trait loci (pQTL) data. Publicly available datasets were utilized, including psoriasis GWAS data from the European Molecular Biology Laboratory-European Bioinformatics Institute and the UK Biobank. Heterogeneity in dependent instruments (HEIDI) test and colocalization analyses were performed to identify shared causal variants, and multi-omics integration was employed to construct potential regulatory pathways. RESULTS Our analyses identified significant causal associations between DNA methylation, gene expression, protein abundance, and psoriasis risk. We discovered two pathways involving the long non-coding RNA RP11-977G19.11 and apolipoprotein F (APOF). Methylation at sites cg26804944 and cg02705573 was negatively associated with RP11-977G19.11 expression. Reduced expression of RP11-977G19.11 was linked to increased APOF levels, which were positively associated with a higher risk of psoriasis. Methylation at sites cg00172967, cg00294382, and cg24773560 was positively associated with RP11-977G19.11 expression. Elevated expression of RP11-977G19.11 was associated with decreased APOF levels, reducing the risk of psoriasis. Colocalization analysis highlighted APOF as a key protein in psoriasis pathogenesis. Validation using skin tissue, EBV-transformed lymphocytes data and inflammation-related protein panels confirmed the associations of RP11-977G19.11 and APOF with psoriasis. CONCLUSIONS Our multi-omics analysis provides preliminary evidence for potential molecular mechanisms in psoriasis pathogenesis. Through the integration of GWAS and molecular QTL data, we identify candidate pathways that may be relevant to disease biology. While these findings require extensive experimental validation, they offer a framework for future investigations into the molecular basis of psoriasis.
Collapse
Affiliation(s)
- Hua Guo
- Department of Academic Research, The Second Hospital of Shandong University, Jinan, Shandong, China
- School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Jinyang Gao
- School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Liping Gong
- Department of Academic Research, The Second Hospital of Shandong University, Jinan, Shandong, China.
| | - Yanqing Wang
- Department of Academic Research, The Second Hospital of Shandong University, Jinan, Shandong, China.
- School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
- Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
31
|
Masson BA, Kiridena P, Lu D, Kleeman EA, Reisinger SN, Qin W, Davies WJ, Muralitharan RR, Jama HA, Antonacci S, Marques FZ, Gubert C, Hannan AJ. Depletion of the paternal gut microbiome alters sperm small RNAs and impacts offspring physiology and behavior in mice. Brain Behav Immun 2025; 123:290-305. [PMID: 39293692 DOI: 10.1016/j.bbi.2024.09.020] [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: 05/16/2024] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 09/20/2024] Open
Abstract
The paternal environment prior to conception has been demonstrated to influence offspring physiology and behavior, with the sperm epigenome (including noncoding RNAs) proposed as a potential facilitator of non-genetic inheritance. Whilst the maternal gut microbiome has been established as an important influence on offspring development, the impact of the paternal gut microbiome on offspring development, health and behavior is largely unknown. Gut microbiota have major influences on immunity, and thus we hypothesized that they may be relevant to paternal immune activation (PIA) modulating epigenetic inheritance in mice. Therefore, male C57BL/6J mice (F0) were orally administered non-absorbable antibiotics via drinking water in order to substantially deplete their gut microbiome. Four weeks after administration of the antibiotics (gut microbiome depletion), F0 male mice were then mated with naïve female mice. The F1 offspring of the microbiome-depleted males had reduced body weight as well as altered gut morphology (shortened colon length). F1 females showed significant alterations in affective behaviors, including measures of anxiety and depressive-like behaviors, indicating altered development. Analysis of small noncoding RNAs in the sperm of F0 mice revealed that gut microbiome depletion is associated with differential expression of 8 different PIWI-interacting RNAs (piRNAs), each of which has the potential to modulate the expression of multiple downstream gene targets, and thus influence epigenetic inheritance and offspring development. This study demonstrates that the gut-germline axis influences sperm small RNA profiles and offspring physiology, with specific impacts on offspring affective and/or coping behaviors. These findings may have broader implications for other animal species with comparable gut microbiota, intergenerational epigenetics and developmental biology, including humans.
Collapse
Affiliation(s)
- Bethany A Masson
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia; Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Pamudika Kiridena
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia; Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Da Lu
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia; Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Elizabeth A Kleeman
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia; Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Sonali N Reisinger
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Wendy Qin
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - William J Davies
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia; Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC, Australia
| | - Rikeish R Muralitharan
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash, Clayton, Australia; Victorian Heart Institute, Monash University, Clayton, Australia
| | - Hamdi A Jama
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash, Clayton, Australia
| | - Simona Antonacci
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash, Clayton, Australia
| | - Francine Z Marques
- Hypertension Research Laboratory, School of Biological Sciences, Faculty of Science, Monash, Clayton, Australia; Heart Failure Research Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia; Victorian Heart Institute, Monash University, Clayton, Australia
| | - Carolina Gubert
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia.
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia; Florey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia; Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
32
|
Forum DMK, Bjerregaard C, Thomsen PH. The significance of DNA methylation of the NR3C1 gene encoding the glucocorticoid receptor for developing resilience in individuals exposed to early life stress. Nord J Psychiatry 2025; 79:1-14. [PMID: 39773140 DOI: 10.1080/08039488.2024.2436987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 11/27/2024] [Accepted: 11/28/2024] [Indexed: 01/11/2025]
Abstract
PURPOSE To analyze and interpret why some individuals are resilient to ELS while others are susceptible, resulting in psychiatric outcome later in life, with a focus on the role of DNAm of the NR3C1 gene as a mediating mechanism between ELS and the risk of psychiatric outcomes. We hypothesized that a high level of mental resilience to ELS, expressed as lower incidence of psychiatric outcomes, was associated with attenuated NR3C1 DNAm levels. MATERIALS AND METHODS The first authors conducted a systematic search on PubMed to identify primary research studies. Abstract were screened and full-text were reviewed to assess the eligibility for inclusion. Consensus on assessment was reached after discussion of eligibility criteria. Studies were sorted based on whether they investigated the association between ELS and NR3C1 DNAm in 1) individuals exposed compared to unexposed to ELS both without a psychiatric outcome or in 2) individuals exposed to ELS with a psychiatric outcome compared to exposed individuals without a psychiatric outcome. RESULTS AND CONCLUSION Seven studies met the eligibility criteria. The results were inconsistent; two studies supported our hypothesis, two studies indicated that increased NR3C1 DNAm mediated resilience to ELS, and three studies found no association.
Collapse
Affiliation(s)
- Ditte Mathilde Klith Forum
- Department of Child and Adolescent Psychiatry, Research Unit, Aarhus University Hospital, Psychiatry, Aarhus, Denmark
| | - Camilla Bjerregaard
- Department of Child and Adolescent Psychiatry, Research Unit, Aarhus University Hospital, Psychiatry, Aarhus, Denmark
| | - Per Hove Thomsen
- Department of Child and Adolescent Psychiatry, Research Unit, Aarhus University Hospital, Psychiatry, Aarhus, Denmark
| |
Collapse
|
33
|
Pacheco HA, Hernandez RO, Chen SY, Neave HW, Pempek JA, Brito LF. Invited review: Phenotyping strategies and genetic background of dairy cattle behavior in intensive production systems-From trait definition to genomic selection. J Dairy Sci 2025; 108:6-32. [PMID: 39389298 DOI: 10.3168/jds.2024-24953] [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: 03/25/2024] [Accepted: 09/14/2024] [Indexed: 10/12/2024]
Abstract
Understanding and assessing dairy cattle behavior is critical for developing sustainable breeding programs and management practices. The behavior of individual animals can provide valuable information on their health and welfare status, improve reproductive management, and predict efficiency traits such as feed efficiency and milking efficiency. Routine genetic evaluations of animal behavior traits can contribute to optimizing breeding and management strategies for dairy cattle but require the identification of traits that capture the most important biological processes involved in behavioral responses. These traits should be heritable, repeatable, and measured in noninvasive and cost-effective ways in many individuals from the breeding populations or related reference populations. Although behavior traits are heritable in dairy cattle populations, they are highly polygenic, with no known major genes influencing their phenotypic expression. Genetically selecting dairy cattle based on their behavior can be advantageous because of their relationship with other key traits such as animal health, welfare, and productive efficiency, as well as animal and handler safety. Trait definition and longitudinal data collection are still key challenges for breeding for behavioral responses in dairy cattle. However, the more recent developments and adoption of precision technologies in dairy farms provide avenues for more objective phenotyping and genetic selection of behavior traits. Furthermore, there is still a need to standardize phenotyping protocols for existing traits and develop guidelines for recording novel behavioral traits and integrating multiple data sources. This review gives an overview of the most common indicators of dairy cattle behavior, summarizes the main methods used for analyzing animal behavior in commercial settings, describes the genetic and genomic background of previously defined behavioral traits, and discusses strategies for breeding and improving behavior traits coupled with future opportunities for genetic selection for improved behavioral responses.
Collapse
Affiliation(s)
- Hendyel A Pacheco
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907
| | - Rick O Hernandez
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907
| | - Shi-Yi Chen
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907; Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan 611130, China
| | - Heather W Neave
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907
| | - Jessica A Pempek
- USDA-ARS, Livestock Behavior Research Unit, West Lafayette, IN 47907
| | - Luiz F Brito
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907.
| |
Collapse
|
34
|
Venditti S. Remodeling the Epigenome Through Meditation: Effects on Brain, Body, and Well-being. Subcell Biochem 2025; 108:231-260. [PMID: 39820865 DOI: 10.1007/978-3-031-75980-2_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Epigenetic mechanisms are key processes that constantly reshape genome activity carrying out physiological responses to environmental stimuli. Such mechanisms regulate gene activity without modifying the DNA sequence, providing real-time adaptation to changing environmental conditions. Both favorable and unfavorable lifestyles have been shown to influence body and brain by means of epigenetics, leaving marks on the genome that can either be rapidly reversed or persist in time and even be transmitted trans-generationally. Among virtuous habits, meditation seemingly represents a valuable way of activating inner resources to cope with adverse experiences. While unhealthy habits, stress, and traumatic early-life events may favor the onset of diseases linked to inflammation, neuroinflammation, and neuroendocrine dysregulation, the practice of mindfulness-based techniques was associated with the alleviation of many of the above symptoms, underlying the importance of lifestyles for health and well-being. Meditation influences brain and body systemwide, eliciting structural/morphological changes as well as modulating the levels of circulating factors and the expression of genes linked to the HPA axis and the immune and neuroimmune systems. The current chapter intends to give an overview of pioneering research showing how meditation can promote health through epigenetics, by reshaping the profiles of the three main epigenetic markers, namely DNA methylation, histone modifications, and non-coding RNAs.
Collapse
Affiliation(s)
- Sabrina Venditti
- Department of Biology and Biotechnologies C. Darwin, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
35
|
Chera A, Stancu-Cretu M, Zabet NR, Bucur O. Shedding light on DNA methylation and its clinical implications: the impact of long-read-based nanopore technology. Epigenetics Chromatin 2024; 17:39. [PMID: 39734197 DOI: 10.1186/s13072-024-00558-2] [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: 08/08/2024] [Accepted: 11/01/2024] [Indexed: 12/31/2024] Open
Abstract
DNA methylation is an essential epigenetic mechanism for regulation of gene expression, through which many physiological (X-chromosome inactivation, genetic imprinting, chromatin structure and miRNA regulation, genome defense, silencing of transposable elements) and pathological processes (cancer and repetitive sequences-associated diseases) are regulated. Nanopore sequencing has emerged as a novel technique that can analyze long strands of DNA (long-read sequencing) without chemically treating the DNA. Interestingly, nanopore sequencing can also extract epigenetic status of the nucleotides (including both 5-Methylcytosine and 5-hydroxyMethylcytosine), and a large variety of bioinformatic tools have been developed for improving its detection properties. Out of all genomic regions, long read sequencing provides advantages in studying repetitive elements, which are difficult to characterize through other sequencing methods. Transposable elements are repetitive regions of the genome that are silenced and usually display high levels of DNA methylation. Their demethylation and activation have been observed in many cancers. Due to their repetitive nature, it is challenging to accurately estimate DNA methylation levels within transposable elements using short sequencing technologies. The advantage to sequence native DNA (without PCR amplification biases or harsh bisulfite treatment) and long and ultra long reads coupled with epigenetic states of the DNA allows to accurately estimate DNA methylation levels in transposable elements. This is a big step forward for epigenomic studies, and unsolved questions regarding gene expression and transposable elements silencing through DNA methylation can now be answered.
Collapse
Affiliation(s)
- Alexandra Chera
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
- Carol Davila Nephrology Clinical Hospital, Bucharest, Romania
| | | | - Nicolae Radu Zabet
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK.
| | - Octavian Bucur
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania.
- Genomics Research and Development Institute, Bucharest, Romania.
- Victor Babes National Institute of Pathology, Bucharest, Romania.
| |
Collapse
|
36
|
Demirkan A, van Dongen J, Finnicum CT, Westra HJ, Jankipersadsing S, Willemsen G, Ijzerman RG, Boomsma DI, Ehli EA, Bonder MJ, Fu J, Franke L, Wijmenga C, de Geus EJC, Kurilshikov A, Zhernakova A. Linking the gut microbiome to host DNA methylation by a discovery and replication epigenome-wide association study. BMC Genomics 2024; 25:1224. [PMID: 39702006 DOI: 10.1186/s12864-024-11136-x] [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: 04/19/2024] [Accepted: 12/06/2024] [Indexed: 12/21/2024] Open
Abstract
Microbiome influences multiple human systems, but its effects on gene methylation is unknown. We investigated the relations between gene methylation in blood and the abundance of common gut bacteria profiled by 16s rRNA gene sequencing in two population-based Dutch cohorts: LifeLines-Deep (LLD, n = 616, discovery) and the Netherlands Twin Register (NTR, n = 296, replication). In LLD, we also explored microbial pathways using data generated by shotgun metagenomic sequencing (n = 683). Methylation in both cohorts was profiled in blood samples using the Illumina 450K array. Discovery and replication analysis identified two independent CpGs associated with the genus Eggerthella: cg16586104 (Pmeta-analysis = 3.21 × 10-11) and cg12234533 (Pmeta-analysis = 4.29 × 10-10). We also show that microbiome can mediate the effect of environmental factors on host gene methylation. In this first association study linking epigenome to microbiome, we found and replicated the associations of two CpGs to the abundance of genus Eggerthella and identified microbiome as a mediator of the exposome. These associations are observational and suggest further investigation in larger and longitudinal set-ups.
Collapse
Affiliation(s)
- Ayşe Demirkan
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
- Department of Clinical and Experimental Medicine, Section of Statistical Multi-omics, School of Biosciences and Medicine & People-Centered AI institute University of Surrey, Guildford, United Kingdom.
| | - Jenny van Dongen
- Biological Psychology, Vrije Universiteit, Amsterdam, the Netherlands
- Amsterdam Public Health Research Institute, Amsterdam MC, Amsterdam, the Netherlands
| | - Casey T Finnicum
- Avera Institute of Human Genetics, Avera McKennan Hospital & University Health Center, Sioux Falls, Sioux Falls, SD, USA
| | - Harm-Jan Westra
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Soesma Jankipersadsing
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Gonneke Willemsen
- Biological Psychology, Vrije Universiteit, Amsterdam, the Netherlands
- Amsterdam Public Health Research Institute, Amsterdam MC, Amsterdam, the Netherlands
| | - Richard G Ijzerman
- Department of Endocrinology, Amsterdam University Medical Center, location AMC, Amsterdam, the Netherlands
| | - Dorret I Boomsma
- Amsterdam Public Health Research Institute, Amsterdam MC, Amsterdam, the Netherlands
- Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Erik A Ehli
- Avera Institute of Human Genetics, Avera McKennan Hospital & University Health Center, Sioux Falls, Sioux Falls, SD, USA
| | - Marc Jan Bonder
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jingyuan Fu
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Lude Franke
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Cisca Wijmenga
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Eco J C de Geus
- Biological Psychology, Vrije Universiteit, Amsterdam, the Netherlands
- Amsterdam Public Health Research Institute, Amsterdam MC, Amsterdam, the Netherlands
| | - Alexander Kurilshikov
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Alexandra Zhernakova
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
37
|
Mundorf A, Freund N. Effects of Early Stress Exposure on Anxiety-like Behavior and MORC1 Expression in Rats. Biomolecules 2024; 14:1587. [PMID: 39766294 PMCID: PMC11674774 DOI: 10.3390/biom14121587] [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: 09/17/2024] [Revised: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
Exposure to stress during early and late childhood can lead to long-lasting neurobiological and behavioral impairments. Although sensitive periods for stress exposure are well established, less is known about the trajectory of induced alterations throughout development. In this study, we investigated the impact of maternal separation (MS), social isolation, and their combination on anxiety-like behavior and gene expression across developmental stages. Sprague Dawley rats were exposed to one or both stressors and later assessed for anxiety-like behavior in juvenility, adolescence, and adulthood. mRNA levels of Morc1, a gene linked to early-life stress and depression, were measured in the medial prefrontal cortex to assess developmental changes. The results showed that MS had age- and sex-dependent effects on anxiety-like behavior. Juveniles exhibited less anxiety after MS, while adolescents showed more pronounced behavioral changes following social isolation. No behavioral changes were observed in adults. Males exhibited greater anxiety-like behavior than females in adolescence and adulthood, but not in juvenility. Female adults exposed to both MS and social isolation had significantly lower Morc1 expression compared to controls. These findings highlight the dynamic effects of early stress across the lifespan, underscoring the critical role of adolescence and differential stress susceptibility by age and sex.
Collapse
Affiliation(s)
- Annakarina Mundorf
- Division of Experimental and Molecular Psychiatry, Department of Psychiatry, Psychotherapy and Preventive Medicine, LWL University Hospital, Ruhr University Bochum, 44801 Bochum, Germany
- Institute for Systems Medicine and Department of Human Medicine, MSH Medical School, 20457 Hamburg, Germany
| | - Nadja Freund
- Division of Experimental and Molecular Psychiatry, Department of Psychiatry, Psychotherapy and Preventive Medicine, LWL University Hospital, Ruhr University Bochum, 44801 Bochum, Germany
| |
Collapse
|
38
|
Chao S, Lu J, Li LJ, Guo HY, Xu K, Wang N, Zhao SX, Jin XW, Wang SG, Yin S, Shen W, Zhao MH, Huang GA, Sun QY, Ge ZJ. Maternal obesity may disrupt offspring metabolism by inducing oocyte genome hyper-methylation via increased DNMTs. eLife 2024; 13:RP97507. [PMID: 39642055 PMCID: PMC11623932 DOI: 10.7554/elife.97507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2024] Open
Abstract
Maternal obesity has deleterious effects on the process of establishing oocyte DNA methylation; yet the underlying mechanisms remain unclear. Here, we found that maternal obesity disrupted the genomic methylation of oocytes using a high-fat diet (HFD) induced mouse model, at least a part of which was transmitted to the F2 oocytes and livers via females. We further examined the metabolome of serum and found that the serum concentration of melatonin was reduced. Exogenous melatonin treatment significantly reduced the hyper-methylation of HFD oocytes, and the increased expression of DNMT3a and DNMT1 in HFD oocytes was also decreased. These suggest that melatonin may play a key role in the disrupted genomic methylation in the oocytes of obese mice. To address how melatonin regulates the expression of DNMTs, the function of melatonin was inhibited or activated upon oocytes. Results revealed that melatonin may regulate the expression of DNMTs via the cAMP/PKA/CREB pathway. These results suggest that maternal obesity induces genomic methylation alterations in oocytes, which can be partly transmitted to F2 in females, and that melatonin is involved in regulating the hyper-methylation of HFD oocytes by increasing the expression of DNMTs via the cAMP/PKA/CREB pathway.
Collapse
Affiliation(s)
- Shuo Chao
- College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural UniversityQingdaoChina
| | - Jun Lu
- College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural UniversityQingdaoChina
| | - Li-Jun Li
- College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural UniversityQingdaoChina
| | - Hong-Yan Guo
- College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural UniversityQingdaoChina
| | - Kuipeng Xu
- College of Horticulture, Qingdao Agricultural UniversityQingdaoChina
| | - Ning Wang
- College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural UniversityQingdaoChina
| | - Shu-Xian Zhao
- College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural UniversityQingdaoChina
| | - Xiao-Wen Jin
- College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural UniversityQingdaoChina
| | - Shao-Ge Wang
- College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural UniversityQingdaoChina
| | - Shen Yin
- College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural UniversityQingdaoChina
| | - Wei Shen
- College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural UniversityQingdaoChina
| | - Ming-Hui Zhao
- College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural UniversityQingdaoChina
| | - Gui-An Huang
- College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural UniversityQingdaoChina
| | - Qing-Yuan Sun
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General HospitalGuangzhouChina
| | - Zhao-Jia Ge
- College of Life Sciences, Institute of Reproductive Sciences, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural UniversityQingdaoChina
| |
Collapse
|
39
|
Mormede E, Mormede P. Genetic Variation of Hypothalamic-Pituitary-Adrenal Axis Activity in Farm Animals and Beyond. Neuroendocrinology 2024; 115:128-137. [PMID: 39626641 DOI: 10.1159/000542831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 11/14/2024] [Indexed: 01/21/2025]
Abstract
BACKGROUND Many experimental data in several species clearly demonstrate the important genetic contribution to variations in HPA axis activity. The influence of corticosteroid hormones on adaptive processes and on production traits such as growth rate, feed efficiency, carcass composition, and meat quality is a strong impetus to the search for the molecular bases of these differences for efficient genetic selection. SUMMARY Three main sources of genetic variability have been documented so far in farm animal species, the adrenal cortex sensitivity to ACTH-regulating corticosteroid hormone production, the bioavailability of corticosteroid hormones and especially corticosteroid-binding globulin capacity, and glucocorticoid receptor function. The effect of single mutations may be dependent on the genetic background, and genetic variation of cortisol levels may have different functional consequences depending on the molecular mechanisms responsible for this change. KEY MESSAGES Understanding the genetic basis of HPA axis activity allows the development of genomic tools and breeding technologies aimed at improving adaptive capacity and stress tolerance in farm animals and their use as valuable models for the genetic study of the HPA axis and the correlation with adaptation, metabolism, and other functions regulated by adrenal hormones, and associated pathologies (obesity, cardiovascular, etc.). The next step will be to explore HPA axis variability from a system genetics perspective including the multiple sources of variation and their interactions. This multifactorial approach is a prerequisite to the use of the HPA axis phenotypes in the genetic selection for more productive and robust animals, with a high level of production of quality products.
Collapse
Affiliation(s)
- Elena Mormede
- GenPhySE, Université de Toulouse, INRAE, INPT, ENVT, Toulouse, France
| | - Pierre Mormede
- GenPhySE, Université de Toulouse, INRAE, INPT, ENVT, Toulouse, France
| |
Collapse
|
40
|
Giannakopoulos A, Chrysis D. Reversibility of disturbed pituitary function in pediatric conditions with psychological stressors: implications for clinical practice. Hormones (Athens) 2024; 23:709-716. [PMID: 38421589 DOI: 10.1007/s42000-024-00536-z] [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: 12/04/2023] [Accepted: 02/13/2024] [Indexed: 03/02/2024]
Abstract
The complex communication network between the central nervous system and the hypothalamic-pituitary axis forms the basis of endocrine functional plasticity, which facilitates adaptation to changing internal and external conditions, but also makes it vulnerable to the negative effects of stressful psychological factors. Herein, clinical conditions such as functional hypothalamic amenorrhea, eating disorders, growth faltering, post-traumatic stress disorder, and pubertal disorders that may emerge during childhood or adolescence, their origin possibly including psychological stressors, are analyzed regarding their genetic susceptibility and reversibility of endocrine function. A discussion on the optimization of therapeutic management defined by managing stress and maximizing the degree and rate of reversibility follows.
Collapse
Affiliation(s)
- Aristeidis Giannakopoulos
- Division of Pediatric Endocrinology, Department of Pediatrics, Medical School of Patras, University Hospital, Rio, 26504, Patras, Greece.
| | - Dionisios Chrysis
- Division of Pediatric Endocrinology, Department of Pediatrics, Medical School of Patras, University Hospital, Rio, 26504, Patras, Greece
| |
Collapse
|
41
|
Gescher DM, Schanze D, Vavra P, Wolff P, Zimmer-Bensch G, Zenker M, Frodl T, Schmahl C. Differential methylation of OPRK1 in borderline personality disorder is associated with childhood trauma. Mol Psychiatry 2024; 29:3734-3741. [PMID: 38862675 PMCID: PMC11609100 DOI: 10.1038/s41380-024-02628-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 05/21/2024] [Accepted: 05/28/2024] [Indexed: 06/13/2024]
Abstract
According to a growing body of neurobiological evidence, the core symptoms of borderline personality disorder (BPD) may be linked to an opioidergic imbalance between the hedonic and stimulatory activity of mu opioid receptors (MOR) and the reward system inhibiting effects of kappa opioid receptors (KOR). Childhood trauma (CT), which is etiologically relevant to BPD, is also likely to lead to epigenetic and neurobiological adaptations by extensive activation of the stress and endogenous opioid systems. In this study, we investigated the methylation differences in the promoter of the KOR gene (OPRK1) in subjects with BPD (N = 47) and healthy controls (N = 48). Comparing the average methylation rates of regulatorily relevant subregions (specified regions CGI-1, CGI-2, EH1), we found no differences between BPD and HC. Analyzing individual CG nucleotides (N = 175), we found eight differentially methylated CG sites, all of which were less methylated in BPD, with five showing highly interrelated methylation rates. This differentially methylated region (DMR) was found on the falling slope (5') of the promoter methylation gap, whose effect is enhanced by the DMR hypomethylation in BPD. A dimensional assessment of the correlation between disease severity and DMR methylation rate revealed DMR hypomethylation to be negatively associated with BPD symptom severity (measured by BSL-23). Finally, analyzing the influence of CT on DMR methylation, we found DMR hypomethylation to correlate with physical and emotional neglect in childhood (quantified by CTQ). Thus, the newly identified DMR may be a biomarker of the risks caused by CT, which likely epigenetically contribute to the development of BPD.
Collapse
MESH Headings
- Humans
- Borderline Personality Disorder/genetics
- Female
- DNA Methylation/genetics
- Male
- Receptors, Opioid, kappa/genetics
- Receptors, Opioid, kappa/metabolism
- Adult
- Promoter Regions, Genetic/genetics
- Epigenesis, Genetic/genetics
- Receptors, Opioid, mu/genetics
- Receptors, Opioid, mu/metabolism
- Child Abuse/psychology
- Adverse Childhood Experiences
- Child
- Middle Aged
- Young Adult
Collapse
Affiliation(s)
- Dorothee Maria Gescher
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, RWTH Aachen University, Aachen, Germany.
- Department for General Psychiatry, Center of Psychosocial Medicine, Medical Faculty, Heidelberg University, Heidelberg, Germany.
- Department of Psychiatry and Psychotherapy, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany.
| | - Denny Schanze
- Institute of Human Genetics, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Peter Vavra
- Department of Biological Psychology, Institute of Psychology, Otto-von-Guericke University, Magdeburg, Germany
| | - Philip Wolff
- Division of Neuroepigenetics, Institute of Zoology (Biology II), RWTH Aachen University, Aachen, Germany
| | - Geraldine Zimmer-Bensch
- Division of Neuroepigenetics, Institute of Zoology (Biology II), RWTH Aachen University, Aachen, Germany
| | - Martin Zenker
- Institute of Human Genetics, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Thomas Frodl
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, RWTH Aachen University, Aachen, Germany
- Department of Psychiatry and Psychotherapy, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
- German Center for Mental Health (DZPG), Jena-Magdeburg-Halle, Germany
- Center for Intervention and Research on adaptive and maladaptive brain Circuits underlying mental health (C-I-R-C), Jena-Magdeburg-Halle, Germany
| | - Christian Schmahl
- Department of Psychosomatic Medicine and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
42
|
Martinez ME, Karaczyn A, Wu Z, Bennett CA, Matoin KL, Daigle HM, Hernandez A. Transgenerational epigenetic self-memory of Dio3 dosage is associated with Meg3 methylation and altered growth trajectories and neonatal hormones. Epigenetics 2024; 19:2376948. [PMID: 38991122 PMCID: PMC11244338 DOI: 10.1080/15592294.2024.2376948] [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: 01/30/2024] [Accepted: 07/02/2024] [Indexed: 07/13/2024] Open
Abstract
Intergenerational and transgenerational epigenetic effects resulting from conditions in previous generations can contribute to environmental adaptation as well as disease susceptibility. Previous studies in rodent and human models have shown that abnormal developmental exposure to thyroid hormone affects endocrine function and thyroid hormone sensitivity in later generations. Since the imprinted type 3 deiodinase gene (Dio3) regulates sensitivity to thyroid hormones, we hypothesize its epigenetic regulation is altered in descendants of thyroid hormone overexposed individuals. Using DIO3-deficient mice as a model of developmental thyrotoxicosis, we investigated Dio3 total and allelic expression and growth and endocrine phenotypes in descendants. We observed that male and female developmental overexposure to thyroid hormone altered total and allelic Dio3 expression in genetically intact descendants in a tissue-specific manner. This was associated with abnormal growth and neonatal levels of thyroid hormone and leptin. Descendant mice also exhibited molecular abnormalities in the Dlk1-Dio3 imprinted domain, including increased methylation in Meg3 and altered foetal brain expression of other genes of the Dlk1-Dio3 imprinted domain. These molecular abnormalities were also observed in the tissues and germ line of DIO3-deficient ancestors originally overexposed to thyroid hormone in utero. Our results provide a novel paradigm of epigenetic self-memory by which Dio3 gene dosage in a given individual, and its dependent developmental exposure to thyroid hormone, influences its own expression in future generations. This mechanism of epigenetic self-correction of Dio3 expression in each generation may be instrumental in descendants for their adaptive programming of developmental growth and adult endocrine function.
Collapse
Affiliation(s)
- M. Elena Martinez
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, Scarborough, ME, USA
| | - Aldona Karaczyn
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, Scarborough, ME, USA
| | - Zhaofei Wu
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, Scarborough, ME, USA
| | - Christian A. Bennett
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, Scarborough, ME, USA
| | - Kassey L. Matoin
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, Scarborough, ME, USA
| | - Heather M. Daigle
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, Scarborough, ME, USA
| | - Arturo Hernandez
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, Scarborough, ME, USA
- Graduate School for Biomedical Sciences and Engineering, University of Maine, Orono, ME, USA
- Department of Medicine, Tufts University School of Medicine, Boston, MA, USA
| |
Collapse
|
43
|
Sullivan EL, Bogdan R, Bakhireva L, Levitt P, Jones J, Sheldon M, Croff JM, Thomason M, Lo JO, MacIntyre L, Shrivastava S, Cioffredi LA, Edlow AG, Howell BR, Chaiyachati BH, Lashley-Simms N, Molloy K, Lam C, Stoermann AM, Trinh T, Ambalavanan N, Neiderhiser JM. Biospecimens in the HEALthy Brain and Child Development (HBCD) Study: Rationale and protocol. Dev Cogn Neurosci 2024; 70:101451. [PMID: 39326174 PMCID: PMC11460495 DOI: 10.1016/j.dcn.2024.101451] [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: 03/06/2024] [Revised: 07/17/2024] [Accepted: 09/13/2024] [Indexed: 09/28/2024] Open
Abstract
The HEALthy Brain and Child Development (HBCD) Study, a multi-site prospective longitudinal cohort study, will examine human brain, cognitive, behavioral, social, and emotional development beginning prenatally and planned through early childhood. The longitudinal collection of biological samples from over 7000 birthing parents and their children within the HBCD study enables research on pre- and postnatal exposures (e.g., substance use, toxicants, nutrition), and biological processes (e.g., genetics, epigenetic signatures, proteins, metabolites) on neurobehavioral developmental outcomes. The following biosamples are collected from the birthing parent: 1) blood (i.e., whole blood, serum, plasma, buffy coat, and dried blood spots) during pregnancy, 2) nail clippings during pregnancy and one month postpartum, 3) urine during pregnancy, and 4) saliva during pregnancy and at in-person postnatal assessments. The following samples are collected from the child at in-person study assessments: 1) saliva, 2) stool, and 3) urine. Additionally, placenta tissue, cord blood, and cord tissue are collected by a subset of HBCD sites. Here, we describe the rationale for the collection of these biospecimens, their current and potential future uses, the collection protocol, and collection success rates during piloting. This information will assist research teams in the planning of future studies utilizing this collection of biological samples.
Collapse
Affiliation(s)
- Elinor L Sullivan
- Departments of Psychiatry and Behavioral Neuroscience, Center for Mental Health Innovation, Oregon Health & Science University, Portland, OR, USA.
| | - Ryan Bogdan
- Department of Psychological & Brain Sciences, Washington University in Saint Louis, Saint Louis, MO, USA.
| | - Ludmila Bakhireva
- Substance Use Research and Education (SURE) Center, College of Pharmacy, University of New Mexico, Albuquerque, NM, USA.
| | - Pat Levitt
- Department of Pediatrics, Keck School of Medicine of University of Southern California, Los Angeles, CA, USA; Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Joseph Jones
- United States Drug Testing Laboratories, Des Plaines, IL, USA
| | | | - Julie M Croff
- Department of Rural Health, Oklahoma State University Center for Health Sciences, Tulsa, OK, USA
| | - Moriah Thomason
- Department of Child and Adolescent Psychiatry & Department of Population Health, New York University Langone Health, New York City, NY, USA
| | - Jamie O Lo
- Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR, USA
| | - Leigh MacIntyre
- McGill University, Montreal, QC, Canada; Lasso Informatics, Montreal, QC, Canada
| | | | - Leigh-Anne Cioffredi
- Dept of Pediatrics, Larner College of Medicine at the University of Vermont, Burlington, VT, USA; Vermont Children's Hospital, Burlington, VT, USA
| | - Andrea G Edlow
- Department of Obstetrics and Gynecology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Brittany R Howell
- Fralin Biomedical Research Institute at VTC, Virginia Tech, Roanoke, VA, USA; Department of Human Development and Family Science, Virginia Tech, Blacksburg, VA, USA
| | - Barbara H Chaiyachati
- Dept of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA; PolicyLab & Clinical Futures, Children's Hospital of Philadelphia, Philadelphia, USA
| | - Nicole Lashley-Simms
- Department of Psychological & Brain Sciences, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Kelly Molloy
- Departments of Psychiatry and Behavioral Neuroscience, Center for Mental Health Innovation, Oregon Health & Science University, Portland, OR, USA
| | - Cris Lam
- University of California, San Diego, San Diego, CA, USA
| | | | - Thanh Trinh
- University of California, San Diego, San Diego, CA, USA
| | | | | |
Collapse
|
44
|
Alberry B, Silveira PP. Early environmental influences on brain development and executive function. Brain Cogn 2024; 182:106241. [PMID: 39542747 DOI: 10.1016/j.bandc.2024.106241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Affiliation(s)
- Bonnie Alberry
- Department of Psychiatry, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Patrícia Pelufo Silveira
- Department of Psychiatry, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada; Ludmer Centre for Neuroinformatics and Mental Health, Douglas Research Centre, McGill University, Montreal, QC, Canada.
| |
Collapse
|
45
|
Olasunkanmi OI, Aremu J, Wong ML, Licinio J, Zheng P. Maternal gut-microbiota impacts the influence of intrauterine environmental stressors on the modulation of human cognitive development and behavior. J Psychiatr Res 2024; 180:307-326. [PMID: 39488009 DOI: 10.1016/j.jpsychires.2024.10.028] [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: 07/18/2023] [Revised: 11/01/2023] [Accepted: 10/23/2024] [Indexed: 11/04/2024]
Abstract
This review examines the longstanding debate of nature and intrauterine environmental challenges that shapes human development and behavior, with a special focus on the influence of maternal prenatal gut microbes. Recent research has revealed the critical role of the gut microbiome in human neurodevelopment, and evidence suggest that maternal microbiota can impact fetal gene and microenvironment composition, as well as immunophysiology and neurochemical responses. Furthermore, intrauterine neuroepigenetic regulation may be influenced by maternal microbiota, capable of having long-lasting effects on offspring behavior and cognition. By examining the complex relationship between maternal prenatal gut microbes and human development, this review highlights the importance of early-life environmental factors in shaping neurodevelopment and cognition.
Collapse
Affiliation(s)
- Oluwatayo Israel Olasunkanmi
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Institute for Brain Science and Disease, Chongqing Medical University, Chongqing, China; Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education) Chongqing Medical University, Chongqing, China.
| | - John Aremu
- Department of Neuroscience, Chongqing Medical University, Chongqing, China
| | - Ma-Li Wong
- Department of Psychiatry, College of Medicine, Upstate Medical University, Syracuse, NY, USA
| | - Julio Licinio
- Department of Psychiatry, College of Medicine, Upstate Medical University, Syracuse, NY, USA.
| | - Peng Zheng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Institute for Brain Science and Disease, Chongqing Medical University, Chongqing, China; Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education) Chongqing Medical University, Chongqing, China.
| |
Collapse
|
46
|
Costa CE, Watowich MM, Goldman EA, Sterner KN, Negron-Del Valle JE, Phillips D, Platt ML, Montague MJ, Brent LJN, Higham JP, Snyder-Mackler N, Lea AJ. Genetic Architecture of Immune Cell DNA Methylation in the Rhesus Macaque. Mol Ecol 2024:e17576. [PMID: 39582237 DOI: 10.1111/mec.17576] [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: 12/04/2023] [Revised: 06/23/2024] [Accepted: 10/18/2024] [Indexed: 11/26/2024]
Abstract
Genetic variation that impacts gene regulation, rather than protein function, can have strong effects on trait variation both within and between species. Epigenetic mechanisms, such as DNA methylation, are often an important intermediate link between genotype and phenotype, yet genetic effects on DNA methylation remain understudied in natural populations. To address this gap, we used reduced representation bisulfite sequencing to measure DNA methylation levels at 555,856 CpGs in peripheral whole blood of 573 samples collected from free-ranging rhesus macaques (Macaca mulatta) living on the island of Cayo Santiago, Puerto Rico. We used allele-specific methods to map cis-methylation quantitative trait loci (meQTL) and tested for effects of 243,389 single nucleotide polymorphisms (SNPs) on local DNA methylation levels. Of 776,092 tested SNP-CpG pairs, we identified 516,213 meQTL, with 69.12% of CpGs having at least one meQTL (FDR < 5%). On average, meQTL explained 21.2% of nearby methylation variance, significantly more than age or sex. meQTL were enriched in genomic compartments where methylation is likely to impact gene expression, for example, promoters, enhancers and binding sites for methylation-sensitive transcription factors. In support, using mRNA-seq data from 172 samples, we confirmed 332 meQTL as whole blood cis-expression QTL (eQTL) in the population, and found meQTL-eQTL genes were enriched for immune response functions, like antigen presentation and inflammation. Overall, our study takes an important step towards understanding the genetic architecture of DNA methylation in natural populations, and more generally points to the biological mechanisms driving phenotypic variation in our close relatives.
Collapse
Affiliation(s)
- Christina E Costa
- Department of Anthropology, New York University, New York, New York, USA
- New York Consortium in Evolutionary Primatology, New York, New York, USA
| | - Marina M Watowich
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, USA
| | | | - Kirstin N Sterner
- Department of Anthropology, University of Oregon, Eugene, Oregon, USA
| | - Josue E Negron-Del Valle
- School of Life Sciences, Arizona State University, Tempe, Arizona, USA
- Center for Evolution and Medicine, Arizona State University, Tempe, Arizona, USA
| | - Daniel Phillips
- School of Life Sciences, Arizona State University, Tempe, Arizona, USA
- Center for Evolution and Medicine, Arizona State University, Tempe, Arizona, USA
| | - Michael L Platt
- Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michael J Montague
- Department of Neuroscience, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | - James P Higham
- Department of Anthropology, New York University, New York, New York, USA
- New York Consortium in Evolutionary Primatology, New York, New York, USA
| | - Noah Snyder-Mackler
- School of Life Sciences, Arizona State University, Tempe, Arizona, USA
- Center for Evolution and Medicine, Arizona State University, Tempe, Arizona, USA
- School of Human Evolution and Social Change, Arizona State University, Tempe, Arizona, USA
- Neurodegenerative Disease Research Center, Arizona State University, Tempe, Arizona, USA
| | - Amanda J Lea
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
47
|
Hong SH, Hardi FA, Tillem S, Goetschius LG, Brooks-Gunn J, McLoyd V, Lopez-Duran NL, Mitchell C, Hyde LW, Monk CS. Mother-child closeness and adolescent structural neural networks: a prospective longitudinal study of low-income families. Soc Cogn Affect Neurosci 2024; 19:nsae083. [PMID: 39512200 PMCID: PMC11631430 DOI: 10.1093/scan/nsae083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 09/17/2024] [Accepted: 11/07/2024] [Indexed: 11/15/2024] Open
Abstract
Mother-child closeness, a mutually trusting and affectionate bond, is an important factor in shaping positive youth development. However, little is known about the neural pathways through which mother-child closeness is related to brain organization. Utilizing a longitudinal sample primarily from low-income families (N = 181; 76% African American youth and 54% female), this study investigated the associations between mother-child closeness at ages 9 and 15 years and structural connectivity organization (network integration, robustness, and segregation) at age 15 years. The assessment of mother-child closeness included perspectives from both mother and child. The results revealed that greater mother-child closeness is linked with increased global efficiency and transitivity, but not with modularity. Specifically, both the mother's and child's reports of closeness at age 15 years predicted network metrics, but report at age 9 years did not. Our findings suggest that mother-child closeness is associated with neural white matter organization, as adolescents who experienced greater mother-child closeness displayed topological properties indicative of more integrated and robust structural networks.
Collapse
Affiliation(s)
- Sunghyun H Hong
- Department of Psychology, University of Michigan, Ann Arbor, MI 48109, United States
- School of Social Work, University of Michigan, Ann Arbor, MI 48109, United States
| | - Felicia A Hardi
- Department of Psychology, University of Michigan, Ann Arbor, MI 48109, United States
| | - Scott Tillem
- Department of Psychology, University of Michigan, Ann Arbor, MI 48109, United States
| | - Leigh G Goetschius
- The Hilltop Institute, University of Maryland Baltimore County, Baltimore, MD 21250, United States
| | - Jeanne Brooks-Gunn
- Teachers College, Columbia University, New York, NY 10027, United States
- College of Physicians and Surgeons, Columbia University, New York, NY 10032, United States
| | - Vonnie McLoyd
- Department of Psychology, University of Michigan, Ann Arbor, MI 48109, United States
| | - Nestor L Lopez-Duran
- Department of Psychology, University of Michigan, Ann Arbor, MI 48109, United States
| | - Colter Mitchell
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI 48104, United States
- Population Studies Center, Institute for Social Research, University of Michigan, Ann Arbor, MI 48106, United States
| | - Luke W Hyde
- Department of Psychology, University of Michigan, Ann Arbor, MI 48109, United States
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI 48104, United States
| | - Christopher S Monk
- Department of Psychology, University of Michigan, Ann Arbor, MI 48109, United States
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI 48104, United States
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, United States
- Department of Psychiatry, University of Michigan, Ann Arbor, MI 48109, United States
| |
Collapse
|
48
|
Arusha KS, Boadi KD, Ellah SS, Kim D, Bauer CM. Sibling presence during fostering ameliorates endocrine stress profile changes in a social rodent species (Octodon degus) in a sex-specific manner. Horm Behav 2024; 166:105660. [PMID: 39500218 DOI: 10.1016/j.yhbeh.2024.105660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 10/30/2024] [Accepted: 11/01/2024] [Indexed: 11/29/2024]
Abstract
During early life, disruption of the parent-offspring bond can substantially impact development of offspring physiology and behavior. In rodents, it has been well-documented that parental separation, reduction in parental care, and cross-fostering can affect development of the endocrine stress response. For social species, however, several social factors may mitigate the stress of cross-fostering, such as remaining with other known adult caregivers or siblings. In this study, we cross-fostered a social rodent species (Octodon degus) with or without their siblings at postnatal day (PND) 8 and measured their endocrine stress response immediately after fostering (PND9) and at weaning (PND28). We found that female singly-fostered offspring displayed elevated baseline cortisol levels and reduced weight gain immediately after fostering. At weaning, female singly-fostered offspring continued to display elevated baseline cortisol levels compared to non-fostered female offspring, while singly-fostered males demonstrated weaker cortisol negative feedback strength compared to male offspring that were not fostered or were fostered with their siblings. These results suggest that sibling presence may help mitigate the stress of fostering, and that future studies should further examine other social conditions that may help reduce developmental consequences of long-term parental bond disruption.
Collapse
Affiliation(s)
- Kaja S Arusha
- Department of Biology, Swarthmore College, Swarthmore, PA, USA
| | - Krystle D Boadi
- Department of Biology, Swarthmore College, Swarthmore, PA, USA
| | - Sabrina S Ellah
- Department of Biology, Swarthmore College, Swarthmore, PA, USA
| | - Daniela Kim
- Department of Biology, Swarthmore College, Swarthmore, PA, USA
| | - Carolyn M Bauer
- Department of Biology, Swarthmore College, Swarthmore, PA, USA.
| |
Collapse
|
49
|
Vetrovoy OV, Potapova SS, Stratilov VA, Tyulkova EI. Prenatal Hypoxia Predisposes to Impaired Expression of the chrna4 and chrna7 Genes in Adult Rats without Affecting Acetylcholine Metabolism during Embryonic Development. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:1950-1960. [PMID: 39647824 DOI: 10.1134/s0006297924110099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/28/2024] [Accepted: 06/03/2024] [Indexed: 12/10/2024]
Abstract
Previous studies have shown that the combined effect of fetal hypoxia and maternal stress hormones predetermines tendency to nicotine addiction in adulthood. This study in rats aimed to investigate the effect of prenatal severe hypoxia (PSH) on acetylcholine metabolism in the developing brain, as well as on expression of acetylcholine receptors chrna4 and chrna7 in both the developing brain and adult brain structures following nicotine consumption. In the developing brain of PSH rats, no changes were found in the activity of choline acetyltransferase (ChAT) and acetylcholinesterase (AChE) or disturbances in the acetylcholine levels. However, decreased chrna4 expression was detected on the day 15 of pregnancy, while elevation in the chrna7 expression was observed on the days 15 and 16 of embryogenesis. In adulthood, the consequences of PSH were manifested as decreased expression of chrna4 in the medial prefrontal cortex (PFC), nucleus accumbens (NAacc), and hypothalamus (HT), decreased expression of chrna7 in the PFC and hippocampus (HPC). Whereas, nicotine consumption did not decrease the expression levels of chrna4 and chrna7 compared to the control group in the adult PSH rats. Thus, prenatal hypoxia predisposes to impaired expression of the chrna4 and chrna7 genes in adult rats without affecting acetylcholine metabolism during embryonic development.
Collapse
Affiliation(s)
- Oleg V Vetrovoy
- Laboratory of Regulation of Brain Neuronal Functions, Pavlov Institute of Physiology, Russian Academy of Sciences, Saint Petersburg, 199034, Russia.
| | - Sofiia S Potapova
- Laboratory of Regulation of Brain Neuronal Functions, Pavlov Institute of Physiology, Russian Academy of Sciences, Saint Petersburg, 199034, Russia
| | - Viktor A Stratilov
- Laboratory of Regulation of Brain Neuronal Functions, Pavlov Institute of Physiology, Russian Academy of Sciences, Saint Petersburg, 199034, Russia
| | - Ekaterina I Tyulkova
- Laboratory of Regulation of Brain Neuronal Functions, Pavlov Institute of Physiology, Russian Academy of Sciences, Saint Petersburg, 199034, Russia
| |
Collapse
|
50
|
Gilbert SF. "When does human life begin?" teaching human embryology in the context of the American abortion debate. Dev Biol 2024; 515:102-111. [PMID: 39004200 DOI: 10.1016/j.ydbio.2024.07.003] [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: 03/13/2024] [Revised: 07/02/2024] [Accepted: 07/11/2024] [Indexed: 07/16/2024]
Abstract
The Dobbs decision of the United States Supreme Court and the actions of several state legislatures have made it risky, if not outright dangerous, to teach factual material concerning human embryology. At some state universities, for instance, if a professor's lecture is felt to teach or discuss abortion (as it might when teaching about tubal pregnancies, hydatidiform moles, or eneuploidy), that instructor risks imprisonment for up to 14 years (Gyori, 2023). Some states' new censorship rules have thus caused professors to drop modules on abortion from numerous science and humanities courses. In most states, instructors can still teach about human embryonic development and not risk putting their careers or livelihoods in jeopardy. However, even in many of these institutions, students can bring a professor to a disciplinary hearing by claiming that the instructor failed to provide ample trigger warnings on such issues. This essay attempts to provide some strategies wherein human embryology and the ethical issues surrounding it might be taught and students may be given resources to counter unscientific falsehoods about fertilization and human development. This essay provides evidence for teaching the following propositions. Mis-information about human biology and medicine is rampant on the internet, and there are skills that can be taught to students that will help them determine which sites should trusted. This is a skill that needs to be taught as part of science courses.
Collapse
Affiliation(s)
- Scott F Gilbert
- Swarthmore College, Swarthmore, PA, 19081, USA; University of Helsinki, Helsinki, Finland.
| |
Collapse
|