201
|
Rakovski C, Lalli M, Gu J, Hobson M, Wollenhaupt-Aguiar B, Minuzzi L, Kapczinski F, de Azevedo Cardoso T, Frey BN. Childhood maltreatment as a predictor of substance use/misuse among youth: A systematic review and meta-analysis. Neurosci Biobehav Rev 2024; 166:105873. [PMID: 39243876 DOI: 10.1016/j.neubiorev.2024.105873] [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: 06/19/2024] [Revised: 08/07/2024] [Accepted: 09/01/2024] [Indexed: 09/09/2024]
Abstract
This systematic review and meta-analysis aimed to comprehensively describe whether experiencing a variety of childhood maltreatment types predicts a variety of substance use/misuse types among youth, beyond the narrow scope covered in previous systematic reviews on similar topics. A literature search was conducted in June, 2022 using PubMed, PsycInfo, and Embase. 58 studies (total participant n=170,749) were included. These studies were primarily organized by substance type outcomes including alcohol (n=43), cannabis (n=25), unspecified substances (n=25), and other specific substances (n=10). Results were further stratified by maltreatment type. For specific maltreatment and substance type combinations, the majority of studies indicated that childhood maltreatment was a significant predictor of substance use/misuse in youth. Of the 10 meta-analyses we conducted, significant associations were found for the majority (9/10) of maltreatment and substance type combinations. For instance, unspecified childhood maltreatment increased the probability of youth alcohol use by about four times, which was the highest relative risk found. In conclusion, this study shows that childhood maltreatment is a predictor of youth substance use/misuse.
Collapse
Affiliation(s)
- Coral Rakovski
- Neuroscience Graduate Program, McMaster University, Hamilton, ON, Canada
| | - Mikayla Lalli
- Department of Kinesiology, McMaster University, Hamilton, ON, Canada
| | - Jessica Gu
- Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Madison Hobson
- Honours Integrated Science Program, School of Interdisciplinary Science, McMaster University, Hamilton, ON, Canada
| | - Bianca Wollenhaupt-Aguiar
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada; Mood Disorders Treatment and Research Centre and Women's Health Concerns Clinic, St. Joseph's Healthcare, Hamilton, ON, Canada
| | - Luciano Minuzzi
- Neuroscience Graduate Program, McMaster University, Hamilton, ON, Canada; Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada; Mood Disorders Treatment and Research Centre and Women's Health Concerns Clinic, St. Joseph's Healthcare, Hamilton, ON, Canada
| | - Flavio Kapczinski
- Neuroscience Graduate Program, McMaster University, Hamilton, ON, Canada; Department of Psychiatry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Taiane de Azevedo Cardoso
- The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Australia
| | - Benicio N Frey
- Neuroscience Graduate Program, McMaster University, Hamilton, ON, Canada; Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada; Mood Disorders Treatment and Research Centre and Women's Health Concerns Clinic, St. Joseph's Healthcare, Hamilton, ON, Canada.
| |
Collapse
|
202
|
Jean KR, Dotson VM. Dementia: Common Syndromes and Modifiable Risk and Protective Factors. Neurol Clin 2024; 42:793-807. [PMID: 39343475 DOI: 10.1016/j.ncl.2024.05.005] [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: 10/01/2024]
Abstract
Dementia is an umbrella term for multiple conditions that lead to progressive cognitive decline and impaired activities of daily living. Neuropsychological evaluation is essential for characterizing the distinct cognitive and behavioral profile that can aid in the diagnostic process and treatment planning for dementia. Modifiable risk factors for dementia such as nutrition, physical activity, sleep, cognitive and social engagement, and stress provide important avenues for prevention. Neurologists and other health care providers can help patients reduce their risk for dementia by providing them with education about modifiable factors and connecting them to resources to empower them to engage in brain-healthy behavior.
Collapse
Affiliation(s)
- Kharine R Jean
- Department of Psychology, Georgia State University, PO Box 5010, Atlanta, GA 30302-5010, USA
| | - Vonetta M Dotson
- Department of Psychology, Georgia State University, PO Box 5010, Atlanta, GA 30302-5010, USA; Gerontology Institute, Georgia State University, PO Box 3984, Atlanta, GA 30302-3984, USA.
| |
Collapse
|
203
|
Lotter LD, Nehls S, Losse E, Dukart J, Chechko N. Temporal dissociation between local and global functional adaptations of the maternal brain to childbirth: a longitudinal assessment. Neuropsychopharmacology 2024; 49:1809-1818. [PMID: 38769432 PMCID: PMC11473773 DOI: 10.1038/s41386-024-01880-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 04/24/2024] [Accepted: 04/29/2024] [Indexed: 05/22/2024]
Abstract
The maternal brain undergoes significant reorganization during birth and the postpartum period. However, the temporal dynamics of these changes remain unclear. Using resting-state functional magnetic resonance imaging, we report on local and global brain function alterations in 75 mothers in their first postpartum week, compared to 23 nulliparous women. In a subsample followed longitudinally for the next six months, we observed a temporal and spatial dissociation between changes observed at baseline (cluster mass permutation: pFWE < 0.05). Local activity and connectivity changes in widespread neocortical regions persisted throughout the studied time period (ANCOVAs vs. controls: pFDR < 0.05), with preliminary evidence linking these alterations to behavioral and psychological adaptations (interaction effect with postpartum time: uncorrected p < 0.05). In contrast, the initially reduced whole-brain connectivity of putamen-centered subcortical areas returned to control levels within six to nine weeks postpartum (linear and quadratic mixed linear models: pFDR < 0.05). The whole-brain spatial colocalization with hormone receptor distributions (Spearman correlations: pFDR < 0.05) and preliminary blood hormone associations (interaction effect with postpartum time: uncorrected p < 0.05) suggested that the postpartum restoration of progesterone levels may underlie this rapid normalization. These observations enhance our understanding of healthy maternal brain function, contributing to the identification of potential markers for pathological postpartum adaptation processes, which in turn could underlie postpartum psychiatric disorders.
Collapse
Affiliation(s)
- Leon D Lotter
- Institute of Neuroscience and Medicine, Brain & Behavior (INM-7), Research Centre Jülich, Jülich, Germany.
- Institute of Systems Neuroscience, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
- Max Planck School of Cognition; Stephanstrasse 1A, 04103, Leipzig, Germany.
| | - Susanne Nehls
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, RWTH Aachen, Aachen, Germany
- Institute of Neuroscience and Medicine, JARA-Institute Brain Structure Function Relationship (INM-10), Research Centre Jülich, Jülich, Germany
| | - Elena Losse
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, RWTH Aachen, Aachen, Germany
| | - Juergen Dukart
- Institute of Neuroscience and Medicine, Brain & Behavior (INM-7), Research Centre Jülich, Jülich, Germany
- Institute of Systems Neuroscience, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Natalya Chechko
- Institute of Neuroscience and Medicine, Brain & Behavior (INM-7), Research Centre Jülich, Jülich, Germany.
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, RWTH Aachen, Aachen, Germany.
- Institute of Neuroscience and Medicine, JARA-Institute Brain Structure Function Relationship (INM-10), Research Centre Jülich, Jülich, Germany.
| |
Collapse
|
204
|
Atila C, Mekkattu S, Murugesu R, Gaisl O, Varghese N, Eckert A, Christ-Crain M. Plasma oxytocin levels in response to glucagon in patients with arginine vasopressin deficiency (central diabetes insipidus) and healthy controls. Endocrine 2024; 86:774-781. [PMID: 38935296 PMCID: PMC11489228 DOI: 10.1007/s12020-024-03920-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 06/04/2024] [Indexed: 06/28/2024]
Abstract
PURPOSE We recently demonstrated an additional oxytocin (OT) deficiency in patients with arginine vasopressin (AVP) deficiency (central diabetes insipidus) by using 3,4-methylenedioxy-methamphetamine (MDMA) as a novel provocation test. However, the implication of the MDMA provocation test in clinical practice might be challenging. Glucagon effectively stimulates vasopressinergic neurons with a strong increase in plasma copeptin. We therefore hypothesized that this provocation test might also stimulate OT. METHODS This is a predefined secondary analysis of a prospective double-blind, randomised, placebo-controlled cross-over trial involving ten patients with AVP deficiency and ten sex- and body-mass index-matched healthy participants at the University Hospital Basel, Switzerland. Each participant underwent the glucagon test (s.c. injection of 1 mg glucagon) and placebo test (s.c. injection of 0.9% normal saline). Plasma OT levels were measured at baseline, 60, 120 and 180 min after injection. The primary objective was to determine whether glucagon stimulates OT and whether OT levels differ between patients with AVP deficiency and healthy participants. The primary outcome (maximum change in OT within 180 min) was compared between groups and conditions using a linear mixed effects model. RESULTS In healthy participants, the median OT at baseline was 82.7 pg/ml [62.3-94.3] and slightly increased to a maximum of 93.3 pg/ml [87.2-121.1] after injection of glucagon, resulting in a change increase of 24.9 pg/ml [5.1-27.8]. Similarly, in patients with AVP deficiency, the median OT at baseline was 73.9 pg/ml [65.3-81.6] and slightly increased after glucagon injection to 114.9 pg/ml [70.9-140.9], resulting in a change increase of 36.8 pg/ml [-2.2 to 51.2]. The results from the mixed model showed no effect between glucagon compared to placebo on OT (difference: -0.5 pg/ml; 95%-CI [-25, 24]; p = 0.97) and no significant treatment-by-group interaction effect between patients compared to healthy participants (interaction: 28 pg/ml; 95%-CI [-7, 62]; p = 0.13). CONCLUSION We found no effect of glucagon on plasma OT levels and no difference between patients with AVP deficiency and healthy participants.
Collapse
Affiliation(s)
- Cihan Atila
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, Basel, Switzerland
- Department of Clinical Research, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Shalini Mekkattu
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, Basel, Switzerland
- Department of Clinical Research, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Rakithan Murugesu
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, Basel, Switzerland
- Department of Clinical Research, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Odile Gaisl
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, Basel, Switzerland
- Department of Clinical Research, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Nimmy Varghese
- Psychiatric University Hospital, University of Basel, Basel, Switzerland
- Research Cluster, Molecular & Cognitive Neuroscience, Division of Neurobiology, University of Basel, 4002, Basel, Switzerland
| | - Anne Eckert
- Psychiatric University Hospital, University of Basel, Basel, Switzerland
- Research Cluster, Molecular & Cognitive Neuroscience, Division of Neurobiology, University of Basel, 4002, Basel, Switzerland
| | - Mirjam Christ-Crain
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, Basel, Switzerland.
- Department of Clinical Research, University Hospital Basel, University of Basel, Basel, Switzerland.
| |
Collapse
|
205
|
Geloso MC, Zupo L, Corvino V. Crosstalk between peripheral inflammation and brain: Focus on the responses of microglia and astrocytes to peripheral challenge. Neurochem Int 2024; 180:105872. [PMID: 39362496 DOI: 10.1016/j.neuint.2024.105872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/18/2024] [Accepted: 09/30/2024] [Indexed: 10/05/2024]
Abstract
A growing body of evidence supports the link between peripheral inflammation and impairment of neurologic functions, including mood and cognitive abilities. The pathogenic event connecting peripheral inflammation and brain dysfunction is represented by neuroinflammation, a pathogenic phenomenon that provides an important contribution to neurodegeneration and cognitive decline also in Alzheimer's, Parkinson's, Huntington's diseases, as well as in Multiple Sclerosis. It is driven by resident brain immune cells, microglia and astrocytes, that acquire an activated phenotype in response to proinflammatory molecules moving from the periphery to the brain parenchyma. Although a huge progress has been made in clarifying cellular and molecular mechanisms bridging peripheral and central inflammation, a clear picture has not been achieved so far. Therefore, experimental models are of crucial relevance to clarify knowledge gaps in this regard. Many findings demonstrate that systemic inflammation induced by pathogen-associated molecular patterns, such as lipopolysaccharide (LPS), is able to trigger neuroinflammation. Therefore, LPS-administration is widely considered a useful tool to study this phenomenon. On this basis, the present review will focus on in vivo studies based on acute and subacute effects of systemic administration of LPS, with special attention on the state of art of microglia and astrocyte response to peripheral challenge.
Collapse
Affiliation(s)
- Maria Concetta Geloso
- Department of Neuroscience, Section of Human Anatomy, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy; Gemelli Science and Technology Park (GSTeP)-Organoids Research Core Facility, Fondazione Policlinico Agostino Gemelli IRCCS, Rome, Italy.
| | - Luca Zupo
- Department of Neuroscience, Section of Human Anatomy, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy
| | - Valentina Corvino
- Department of Neuroscience, Section of Human Anatomy, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy
| |
Collapse
|
206
|
Li X, Takahashi N, Narita A, Nakamura Y, Sakurai-Yageta M, Murakami K, Ishikuro M, Obara T, Kikuya M, Ueno F, Metoki H, Ohseto H, Takahashi I, Nakamura T, Warita N, Shoji T, Yu Z, Ono C, Kobayashi N, Kikuchi S, Matsuki T, Nagami F, Ogishima S, Sugawara J, Hoshiai T, Saito M, Fuse N, Kinoshita K, Yamamoto M, Yaegashi N, Ozaki N, Tamiya G, Kuriyama S, Tomita H. Identification of risk loci for postpartum depression in a genome-wide association study. Psychiatry Clin Neurosci 2024; 78:712-720. [PMID: 39287932 DOI: 10.1111/pcn.13731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/30/2024] [Accepted: 08/06/2024] [Indexed: 09/19/2024]
Abstract
AIM Genome-wide association studies (GWAS) of postpartum depression (PPD) based on accumulated cohorts with multiple ethnic backgrounds have failed to identify significantly associated loci. Herein, we conducted a GWAS of Japanese perinatal women along with detailed confounding information to uncover PPD-associated loci. METHODS The first and second cohorts (n = 9260 and n = 8582 perinatal women enrolled in the Tohoku Medical Megabank Project) and the third cohort (n = 997), recruited at Nagoya University, underwent genotyping. Of them, 1421, 1264, and 225 were classified as PPD based on the Edinburgh Postnatal Depression Scale 1 month after delivery. The most influential confounding factors of genetic liability to PPD were selected, and logistic regression analyses were performed to evaluate genetic associations with PPD after adjusting for confounders. RESULTS A meta-analysis of GWAS results from the three cohorts identified significant associations between PPD and the following loci (P < 5 × 10-8) by integrating the number of deliveries and the number of family members living together as the most influential confounders: rs377546683 at DAB1, rs11940752 near UGT8, rs141172317, rs117928019, rs76631412, rs118131805 at DOCK2, rs188907279 near ZNF572, rs504378, rs690150, rs491868, rs689917, rs474978, rs690118, rs690253 near DIRAS2, rs1435984417 at ZNF618, rs57705782 near PTPRM, and rs185293917 near PDGFB. Pathway analyses indicated that SNPs suggestively associated with PPD were mostly over-represented in categories including long-term depression, GnRH signaling, glutamatergic synapse, oxytocin signaling, and Rap1 signaling. CONCLUSION The current GWAS study identified eight loci significantly associated with PPD, which may clarify the genetic structure underlying its pathogenesis.
Collapse
Affiliation(s)
- Xue Li
- Department of Psychiatry, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Regional Alliance for Promoting Liaison Psychiatry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Nagahide Takahashi
- Department of Child and Adolescent Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Akira Narita
- Department of Integrative Genomics, Tohoku University Tohoku Medical Megabank Organization, Sendai, Japan
| | - Yukako Nakamura
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mika Sakurai-Yageta
- Department of Integrative Genomics, Tohoku University Tohoku Medical Megabank Organization, Sendai, Japan
| | - Keiko Murakami
- Department of Preventive Medicine and Epidemiology, Tohoku University Tohoku Medical Megabank Organization, Sendai, Japan
| | - Mami Ishikuro
- Department of Preventive Medicine and Epidemiology, Tohoku University Tohoku Medical Megabank Organization, Sendai, Japan
| | - Taku Obara
- Department of Preventive Medicine and Epidemiology, Tohoku University Tohoku Medical Megabank Organization, Sendai, Japan
| | - Masahiro Kikuya
- Department of Preventive Medicine and Epidemiology, Tohoku University Tohoku Medical Megabank Organization, Sendai, Japan
- Department of Hygiene and Public Health, Teikyo University School of Medicine, Tokyo, Japan
| | - Fumihiko Ueno
- Department of Preventive Medicine and Epidemiology, Tohoku University Tohoku Medical Megabank Organization, Sendai, Japan
| | - Hirohito Metoki
- Department of Preventive Medicine and Epidemiology, Tohoku University Tohoku Medical Megabank Organization, Sendai, Japan
| | - Hisashi Ohseto
- Department of Preventive Medicine and Epidemiology, Tohoku University Tohoku Medical Megabank Organization, Sendai, Japan
| | - Ippei Takahashi
- Department of Preventive Medicine and Epidemiology, Tohoku University Tohoku Medical Megabank Organization, Sendai, Japan
| | - Tomohiro Nakamura
- Department of Health Record Informatics, Tohoku University Tohoku Medical Megabank Organization, Sendai, Japan
| | - Noriko Warita
- Department of Psychiatry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tomoka Shoji
- Department of Psychiatry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Zhiqian Yu
- Department of Psychiatry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Chiaki Ono
- Department of Psychiatry, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Regional Alliance for Promoting Liaison Psychiatry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | - Saya Kikuchi
- Department of Psychiatry, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Psychiatry, Tohoku University Hospital, Sendai, Japan
| | - Tasuku Matsuki
- Department of Psychiatry, Tohoku University Hospital, Sendai, Japan
| | - Fuji Nagami
- Department of Public Relations and Planning, Tohoku University Tohoku Medical Megabank Organization, Sendai, Japan
| | - Soichi Ogishima
- Department of Health Record Informatics, Tohoku University Tohoku Medical Megabank Organization, Sendai, Japan
| | - Junichi Sugawara
- Department of Community Medical Supports, Tohoku University Tohoku Medical Megabank Organization, Sendai, Japan
- Department of gynecology and obstetrics, Tohoku University Graduate School of Medicine, Sendai, Japan
- Suzuki Memorial Hospital, Iwanumashi, Japan
| | - Tetsuro Hoshiai
- Department of gynecology and obstetrics, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masatoshi Saito
- Department of gynecology and obstetrics, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Nobuo Fuse
- Department of Integrative Genomics, Tohoku University Tohoku Medical Megabank Organization, Sendai, Japan
| | - Kengo Kinoshita
- Department of Integrative Genomics, Tohoku University Tohoku Medical Megabank Organization, Sendai, Japan
| | - Masayuki Yamamoto
- Department of Integrative Genomics, Tohoku University Tohoku Medical Megabank Organization, Sendai, Japan
| | - Nobuo Yaegashi
- Department of Community Medical Supports, Tohoku University Tohoku Medical Megabank Organization, Sendai, Japan
- Department of gynecology and obstetrics, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Norio Ozaki
- Department of Child and Adolescent Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Pathophysiology of Mental Disorders, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Gen Tamiya
- Department of Integrative Genomics, Tohoku University Tohoku Medical Megabank Organization, Sendai, Japan
| | - Shinichi Kuriyama
- Department of Preventive Medicine and Epidemiology, Tohoku University Tohoku Medical Megabank Organization, Sendai, Japan
- Tohoku University International Research Institute of Disaster Sciences, Sendai, Japan
| | - Hiroaki Tomita
- Department of Psychiatry, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Regional Alliance for Promoting Liaison Psychiatry, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Preventive Medicine and Epidemiology, Tohoku University Tohoku Medical Megabank Organization, Sendai, Japan
- Department of Psychiatry, Tohoku University Hospital, Sendai, Japan
- Tohoku University International Research Institute of Disaster Sciences, Sendai, Japan
| |
Collapse
|
207
|
Khan H, Naseem T, Kaushik P, Narang J, Khan R, Panwar S, Parvez S. Decoding paradoxical links of cytokine markers in cognition: Cross talk between physiology, inflammaging, and Alzheimer's disease- related cognitive decline. Ageing Res Rev 2024; 101:102535. [PMID: 39374831 DOI: 10.1016/j.arr.2024.102535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 09/29/2024] [Accepted: 10/01/2024] [Indexed: 10/09/2024]
Abstract
Recent research has revolutionized our understanding of memory consolidation by emphasizing the critical role of astrocytes, microglia, and immune cells in through cytokine signaling. Cytokines, compact proteins, play pivotal roles in neuronal development, synaptic transmission, and normal aging. This review explores the cellular mechanisms contributing to cognitive decline in inflammaging and Alzheimer's disease, highlighting the paradoxical effects of most studied cytokines (IL-1, IL-6, TNF-α) in brain function, which act as a double-edged sword in brain physiology, acting both as facilitators of healthy cognitive function and as a potential contributor to cognitive decline.
Collapse
Affiliation(s)
- Hiba Khan
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Talib Naseem
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Pooja Kaushik
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Jagriti Narang
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Rehan Khan
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector 81, Knowledge City, Sahibzada Ajit Singh Nagar, Mohali, Punjab 140306, India
| | - Siddharth Panwar
- School of Computing and Electrical Engineering, Indian Institute of Technology, Mandi, Himachal Pradesh 175075, India
| | - Suhel Parvez
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
208
|
Rayan NA, Aow J, Lim MGL, Arcego DM, Ryan R, Nourbakhsh N, de Lima RMS, Craig K, Zhang TY, Goh YT, Sun AX, Tompkins T, Bronner S, Binda S, Diorio J, Parent C, Meaney MJ, Prabhakar S. Shared and unique transcriptomic signatures of antidepressant and probiotics action in the mammalian brain. Mol Psychiatry 2024; 29:3653-3668. [PMID: 38844534 DOI: 10.1038/s41380-024-02619-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 05/13/2024] [Accepted: 05/16/2024] [Indexed: 11/08/2024]
Abstract
Understanding the shared and divergent mechanisms across antidepressant (AD) classes and probiotics is critical for improving treatment for mood disorders. Here we examine the transcriptomic effects of bupropion (NDRI), desipramine (SNRI), fluoxetine (SSRI) and a probiotic formulation (Lacidofil®) on 10 regions across the mammalian brain. These treatments massively alter gene expression (on average, 2211 differentially expressed genes (DEGs) per region-treatment combination), highlighting the biological complexity of AD and probiotic action. Intersection of DEG sets against neuropsychiatric GWAS loci, sex-specific transcriptomic portraits of major depressive disorder (MDD), and mouse models of stress and depression reveals significant similarities and differences across treatments. Interestingly, molecular responses in the infralimbic cortex, basolateral amygdala and locus coeruleus are region-specific and highly similar across treatments, whilst responses in the Raphe, medial preoptic area, cingulate cortex, prelimbic cortex and ventral dentate gyrus are predominantly treatment-specific. Mechanistically, ADs concordantly downregulate immune pathways in the amygdala and ventral dentate gyrus. In contrast, protein synthesis, metabolism and synaptic signaling pathways are axes of variability among treatments. We use spatial transcriptomics to further delineate layer-specific molecular pathways and DEGs within the prefrontal cortex. Our study reveals complex AD and probiotics action on the mammalian brain and identifies treatment-specific cellular processes and gene targets associated with mood disorders.
Collapse
Affiliation(s)
- Nirmala Arul Rayan
- Genome Institute of Singapore, Agency for Science Technology and Research (A*STAR), Singapore, 138672, Singapore
| | - Jonathan Aow
- Genome Institute of Singapore, Agency for Science Technology and Research (A*STAR), Singapore, 138672, Singapore
- NUS Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Michelle Gek Liang Lim
- Genome Institute of Singapore, Agency for Science Technology and Research (A*STAR), Singapore, 138672, Singapore
| | - Danusa Mar Arcego
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, QC, H4H 1R3, Canada
| | - Richard Ryan
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, QC, H4H 1R3, Canada
| | - Nooshin Nourbakhsh
- Genome Institute of Singapore, Agency for Science Technology and Research (A*STAR), Singapore, 138672, Singapore
| | | | - Kelly Craig
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, QC, H4H 1R3, Canada
| | - Tie Yuan Zhang
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, QC, H4H 1R3, Canada
| | - Yeek Teck Goh
- Genome Institute of Singapore, Agency for Science Technology and Research (A*STAR), Singapore, 138672, Singapore
| | - Alfred Xuyang Sun
- Duke-NUS Graduate Medical School, Signature Research Program in Neuroscience and Behavioural Disorders, 8 College Road, Singapore, 169857, Singapore
| | - Thomas Tompkins
- Lallemand Bio-Ingredients, 1620 Rue Prefontaine, Montréal, QC, H1W 2N8, Canada
| | - Stéphane Bronner
- Lallemand Health Solutions, Rosell Institute for Microbiome and Probiotics, 6100 Avenue Royalmount, Montréal, QC, H4P 2R2, Canada
| | - Sylvie Binda
- Lallemand Health Solutions, Rosell Institute for Microbiome and Probiotics, 6100 Avenue Royalmount, Montréal, QC, H4P 2R2, Canada
| | - Josie Diorio
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, QC, H4H 1R3, Canada
| | - Carine Parent
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, QC, H4H 1R3, Canada
| | - Michael J Meaney
- NUS Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Montréal, QC, H4H 1R3, Canada.
- Singapore Institute for Clinical Sciences, A*STAR, Singapore, 117609, Singapore.
- Brain-Body Initiative, Institute for Cell & Molecular Biology, A*STAR, Singapore, Singapore.
| | - Shyam Prabhakar
- Genome Institute of Singapore, Agency for Science Technology and Research (A*STAR), Singapore, 138672, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore.
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore.
| |
Collapse
|
209
|
Ruhland S, Poeppl TB, Schoisswohl S, Schwitzgebel F, Osnabrügge M, Kanig C, Langguth B, Schecklmann M. Motor-evoked potentials as biomarkers for sexual arousal? J Sex Med 2024; 21:1004-1010. [PMID: 39279159 DOI: 10.1093/jsxmed/qdae122] [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: 04/10/2024] [Revised: 08/03/2024] [Accepted: 08/29/2024] [Indexed: 09/18/2024]
Abstract
BACKGROUND Motor cortex excitability may represent the neuronal endpoint of motivational processes and was shown to be modulated by both sexual arousal and deceptive behavior. AIM This is the first study to investigate the influence of lying and sex in heterosexual women and men based on motor-evoked potentials (MEPs) measured while viewing sexually arousing pictures. METHODS Sixteen heterosexual couples were shown 360 trials consisting of pictures displaying both almost naked females and males and neutral control images. In a subsequent forced-choice question about wanting to see the respective pictures fully naked, they were instructed to either answer in agreement with or opposite to their sexual preference. Participants went through 2 blocks of answering truthfully and 2 blocks of lying, with these 4 blocks being shown in a randomized alternating order. OUTCOMES To measure cortical excitability, MEPs were used, evoked by single transcranial magnetic stimulation pulses between image presentation and response. RESULTS In normalized MEPs, women and men showed higher amplitudes for preferred over non-preferred sexual stimuli, but only on a descriptive level. Planned contrasts showed higher non-normalized MEPs for lying in all picture categories. Direct comparisons to a preliminary study showed overall lower effect sizes. CLINICAL IMPLICATIONS Both sexes tend to show higher MEPs in response to their sexually preferred stimuli. MEPs are not stable markers for willful volitionally controlled deception although lying does increase cortical excitability. The present experimental design does not seem valid enough to serve as a diagnostic marker for sexual preference or paraphilia and malingering. STRENGTHS AND LIMITATIONS This is the first study investigating whether sexual motivational stimuli modulate MEPs in women, while also examining the influence of lying for both sexes. The sample was too small for some found effects to be significant. Also, the experimental setup may have been less suited for female participants in comparison to male ones. CONCLUSION The operationalization of sexual motivation via MEPs seems to highly depend on different experimental factors including the sex of the participants, induced motivation, and lying.
Collapse
Affiliation(s)
- Stefanie Ruhland
- Department of Psychiatry and Psychotherapy, University of Regensburg, Universitaetsstrasse 84, Regensburg 93053, Germany
| | - Timm B Poeppl
- Department of Psychiatry and Psychotherapy, University of Regensburg, Universitaetsstrasse 84, Regensburg 93053, Germany
- Department of Psychiatry and Psychotherapy, Faculty of Medicine, RWTH Aachen University, Templergraben 55, Aachen 52062, Germany
| | - Stefan Schoisswohl
- Department of Psychiatry and Psychotherapy, University of Regensburg, Universitaetsstrasse 84, Regensburg 93053, Germany
- Department of Human Sciences, University of the Bundeswehr Munich, Werner-Heisenberg-Weg 39, Neubiberg 85579, Germany
| | - Florian Schwitzgebel
- Department of Psychiatry and Psychotherapy, University of Regensburg, Universitaetsstrasse 84, Regensburg 93053, Germany
- Department of Electrical Engineering, University of the Bundeswehr Munich, Werner-Heisenberg-Weg 39, Neubiberg 85579, Germany
| | - Mirja Osnabrügge
- Department of Psychiatry and Psychotherapy, University of Regensburg, Universitaetsstrasse 84, Regensburg 93053, Germany
- Department of Human Sciences, University of the Bundeswehr Munich, Werner-Heisenberg-Weg 39, Neubiberg 85579, Germany
| | - Carolina Kanig
- Department of Psychiatry and Psychotherapy, University of Regensburg, Universitaetsstrasse 84, Regensburg 93053, Germany
- Department of Human Sciences, University of the Bundeswehr Munich, Werner-Heisenberg-Weg 39, Neubiberg 85579, Germany
| | - Berthold Langguth
- Department of Psychiatry and Psychotherapy, University of Regensburg, Universitaetsstrasse 84, Regensburg 93053, Germany
| | - Martin Schecklmann
- Department of Psychiatry and Psychotherapy, University of Regensburg, Universitaetsstrasse 84, Regensburg 93053, Germany
| |
Collapse
|
210
|
Chakravarti B, Rajput S, Srivastava A, Sharma LK, Sinha RA, Chattopadhyay N, Siddiqui JA. A Systematic Review and Meta-Analysis of the Effects of Dietary Isoflavones on Female Hormone-Dependent Cancers for Benefit-Risk Evaluation. Phytother Res 2024. [PMID: 39480044 DOI: 10.1002/ptr.8358] [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: 02/09/2024] [Revised: 07/22/2024] [Accepted: 09/30/2024] [Indexed: 11/02/2024]
Abstract
Female hormone-dependent cancers depend on estrogen for their growth. Numerous studies have explored the antitumor effect of dietary isoflavones on female hormone-dependent cancers. Still, few clinical evidence supports the use of isoflavones in female hormone-dependent cancer patients. This study was performed to examine the impact of dietary isoflavones on tumor growth of female hormone-dependent cancers and accelerate the transformation of research from bench to bedside. We searched PubMed Medline, Web of Science, and Google Scholar for relevant articles related to the effect of dietary isoflavone on tumor growth of experimental animal models of female hormone-dependent cancers from 1998 to 2024. The effects of dietary isoflavones on tumor growth were analyzed between the control and treatment groups using comprehensive meta-analysis software (CMA). We included 30 studies describing tumor growth focused on female hormone-dependent cancer types, including breast, ovarian, and uterine cancers. Overall, a pooled analysis revealed that dietary isoflavones reduced tumor volume (Hedge's g = -1.151, 95% CI = -1.717 to -0.585, p = 0.000) and tumor weight (Hedge's g = -2.584, 95% CI = -3.618 to -1.549, p = 0.000). On the other hand, dietary isoflavones increased tumor area (Hedge's g = 1.136, 95% CI = 0.752 to 1.520, p = 0.000). Dietary isoflavones have potential benefits and risks in female hormone-dependent cancers. Therefore, caution should be exercised when considering the intake of dietary isoflavones in female hormone-dependent cancer patients, particularly in the form of supplements.
Collapse
Affiliation(s)
- Bandana Chakravarti
- Stem Cell/Cell culture lab Unit, Center for Advance Research, King George's Medical University, Lucknow, India
| | - Swati Rajput
- Division of Endocrinology and Center for Research in Anabolic Skeletal Target in Health and Illness (ASTHI), CSIR-Central Drug Research Institute, Council of Scientific and Industrial Research, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Anubhav Srivastava
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences Lucknow, Lucknow, Uttar Pradesh, India
| | - Lokendra Kumar Sharma
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences Lucknow, Lucknow, Uttar Pradesh, India
| | - Rohit Anthony Sinha
- Stem Cell/Cell culture lab Unit, Center for Advance Research, King George's Medical University, Lucknow, India
| | - Naibedya Chattopadhyay
- Division of Endocrinology and Center for Research in Anabolic Skeletal Target in Health and Illness (ASTHI), CSIR-Central Drug Research Institute, Council of Scientific and Industrial Research, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Jawed Akhtar Siddiqui
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, USA
- Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS, USA
| |
Collapse
|
211
|
Ruiz-Cruz M, Roa J, Tena-Sempere M. Gonadotropin-releasing hormone. Trends Endocrinol Metab 2024:S1043-2760(24)00258-3. [PMID: 39487046 DOI: 10.1016/j.tem.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 10/10/2024] [Accepted: 10/11/2024] [Indexed: 11/04/2024]
Affiliation(s)
- Miguel Ruiz-Cruz
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain; Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Córdoba, Spain
| | - Juan Roa
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain; Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Córdoba, Spain
| | - Manuel Tena-Sempere
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain; Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Córdoba, Spain.
| |
Collapse
|
212
|
Anderson JC, Seitz DP, Crockford D, Addington D, Baek H, Lorenzetti DL, Barry R, Bolton JM, Taylor VH, Kurdyak P, Kirkham J. Quality indicators for schizophrenia care: A scoping review. Schizophr Res 2024; 274:406-416. [PMID: 39486104 DOI: 10.1016/j.schres.2024.10.013] [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: 06/24/2024] [Revised: 10/03/2024] [Accepted: 10/19/2024] [Indexed: 11/04/2024]
Abstract
Measuring quality of care is a critical first step towards improving the healthcare contributing to persistent poor outcomes experienced by many people living with schizophrenia. This scoping review aims to identify and characterize indicators for measuring the quality of care for people living with schizophrenia. We searched 6 academic databases, 4 grey literature databases, and 23 organization websites for documents containing quality indicators developed for or applied in a population with schizophrenia-spectrum disorders. We identified 119 unique documents, yielding 390 distinct quality indicators. Most measures were process indicators (68 %; n = 267) commonly reflecting safety (30 %; n = 118) and effectiveness (35 %; n = 136) domains of quality of care. Quality indicators included measures of primarily mental healthcare (77 %; n = 299), as well as physical healthcare (23 %; n = 91). Indicators reflected aspects of care related to service delivery, pharmacotherapy, assessments, resources and policies, psychological interventions, social and other interventions. Indicator development was notable for a lack of well-described validation and selection processes. Gaps in indicator availability for comorbid substance use, reproductive health, and healthcare equity were also identified. Results reflect a growing recognition of the importance of quality measurement in this population but highlight the need for prioritization of indicators to guide future quality measurement and improvement.
Collapse
Affiliation(s)
- Jennifer C Anderson
- Department of Psychiatry, Cumming School of Medicine, University of Calgary, 2500 University Dr. NW, Calgary, Alberta T2N 1N4, Canada; Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, 3280 Hospital Dr. NW, Calgary, Alberta T2N 4Z6, Canada.
| | - Dallas P Seitz
- Department of Psychiatry, Cumming School of Medicine, University of Calgary, 2500 University Dr. NW, Calgary, Alberta T2N 1N4, Canada; Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Dr. NW, Alberta T2N 1N4, Canada.
| | - David Crockford
- Department of Psychiatry, Cumming School of Medicine, University of Calgary, 2500 University Dr. NW, Calgary, Alberta T2N 1N4, Canada; Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Dr. NW, Alberta T2N 1N4, Canada.
| | - Donald Addington
- Department of Psychiatry, Cumming School of Medicine, University of Calgary, 2500 University Dr. NW, Calgary, Alberta T2N 1N4, Canada; Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Dr. NW, Alberta T2N 1N4, Canada.
| | - Hanji Baek
- Department of Psychiatry, Cumming School of Medicine, University of Calgary, 2500 University Dr. NW, Calgary, Alberta T2N 1N4, Canada.
| | - Diane L Lorenzetti
- Libraries and Cultural Resources, University of Calgary, 2500 University Dr. NW, Calgary, Alberta T2N 1N4, Canada; O'Brien Institute for Public Health, University of Calgary, 3280 Hospital Dr. NW, Calgary, Alberta T2N 4Z6, Canada; Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, 3280 Hospital Dr. NW, Calgary, Alberta T2N 4Z6, Canada.
| | - Rebecca Barry
- Department of Psychiatry, Cumming School of Medicine, University of Calgary, 2500 University Dr. NW, Calgary, Alberta T2N 1N4, Canada; Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Dr. NW, Alberta T2N 1N4, Canada.
| | - James M Bolton
- Department of Psychiatry, Max Rady College of Medicine, University of Manitoba, 771 Bannatyne Ave., Winnipeg, Manitoba R3E 3N4, Canada; Manitoba Centre for Health Policy, 727 McDermot Ave., Winnipeg, Manitoba R3E 3P5, Canada.
| | - Valerie H Taylor
- Department of Psychiatry, Cumming School of Medicine, University of Calgary, 2500 University Dr. NW, Calgary, Alberta T2N 1N4, Canada; Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Dr. NW, Alberta T2N 1N4, Canada; The Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, 3280 Hospital Dr. NW, Calgary, Alberta T2N 4N1, Canada; Alberta Children's Hospital Research Institute, 3330 Hospital Dr. NW, Calgary, Alberta T2N 4N1, Canada.
| | - Paul Kurdyak
- Department of Psychiatry, University of Toronto; 250 College St., Toronto, Ontario M5T 1R8, Canada; Centre for Addiction and Mental Health, 250 College St., Toronto, Ontario M5T 1R8, Canada; ICES, 2075 Bayview Ave., Toronto, Ontario M4N 3M5, Canada.
| | - Julia Kirkham
- Department of Psychiatry, Cumming School of Medicine, University of Calgary, 2500 University Dr. NW, Calgary, Alberta T2N 1N4, Canada; Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Dr. NW, Alberta T2N 1N4, Canada.
| |
Collapse
|
213
|
Klimek A, Kletkiewicz H, Siejka A, Wyszkowska J, Maliszewska J, Klimiuk M, Jankowska M, Rogalska J. The extremely low-frequency electromagnetic field (50 Hz) can establish a new "set-point" for the activity of the locus coeruleus-noradrenergic (LC-NA) system in rat. Brain Res Bull 2024; 219:111111. [PMID: 39486464 DOI: 10.1016/j.brainresbull.2024.111111] [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: 08/20/2024] [Revised: 10/25/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024]
Abstract
Exposure of organisms to extremely low-frequency electromagnetic field (ELF-EMF; 50 Hz) has been increasing in recent decades, which is connected with dynamic technological development. ELF-EMF is considered a stress factor and its effects on organisms are still being investigated. We aimed to determine its impact on the locus coeruleus-noradrenergic (LC-NA) system enabling adaptation to stressful conditions. For this purpose, we exposed rats to 50 Hz ELF-EMF of 1 and 7 mT, 1 h/day for 7 days. The procedure was repeated three times to examine the organism's adaptive capabilities. Subsequently, the concentration of adrenaline, noradrenaline and its metabolite MHPG as well as the expression of the β2-adrenergic receptor was assessed. After the end of each exposure, part of the animals were subjected to a behavioural test to assess the influence of repeated ELF-EMF exposure on stress response to subsequent stress factors. Our research proved that mechanisms underlying the effects of ELF-EMF on stress response include the LC-NA system. ELF-EMF of 1 mT induced adaptive changes in the NA-LC system. However, exposure to 7 mT caused increased activity of the stress system which resulted in sensitization to subsequent, heterotypic (different from the one previously acting) stress factor. As ELF-EMF of 7 mT caused a profound decrease in β2-AR level would strongly inhibit the potential for neuroplastic processes in the hippocampus. Moreover, rats exposed to ELF-EMF of 7 mT showed moderately increased anxiety-related behaviour. Disturbances in NA-LC transmission may underlie the development of some neurodegenerative and psychiatric diseases which indicates the possible involvement of ELF-EMF in the pathogenesis of these disorders.
Collapse
Affiliation(s)
- Angelika Klimek
- Department of Exercise Physiology and Functional Anatomy, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Bydgoszcz 85-077, Poland.
| | - Hanna Kletkiewicz
- Department of Animal Physiology and Neurobiology, Faculty of Biological and Veterinary Sciences Nicolaus Copernicus University in Torun, 87-100, Poland
| | - Agnieszka Siejka
- Department of Animal Physiology and Neurobiology, Faculty of Biological and Veterinary Sciences Nicolaus Copernicus University in Torun, 87-100, Poland
| | - Joanna Wyszkowska
- Department of Animal Physiology and Neurobiology, Faculty of Biological and Veterinary Sciences Nicolaus Copernicus University in Torun, 87-100, Poland
| | - Justyna Maliszewska
- Department of Animal Physiology and Neurobiology, Faculty of Biological and Veterinary Sciences Nicolaus Copernicus University in Torun, 87-100, Poland
| | - Maciej Klimiuk
- Department of Animal Physiology and Neurobiology, Faculty of Biological and Veterinary Sciences Nicolaus Copernicus University in Torun, 87-100, Poland
| | - Milena Jankowska
- Department of Animal Physiology and Neurobiology, Faculty of Biological and Veterinary Sciences Nicolaus Copernicus University in Torun, 87-100, Poland
| | - Justyna Rogalska
- Department of Animal Physiology and Neurobiology, Faculty of Biological and Veterinary Sciences Nicolaus Copernicus University in Torun, 87-100, Poland.
| |
Collapse
|
214
|
Morgan A, Shekhar N, Strnadová V, Pirník Z, Haasová E, Kopecký J, Pačesová A, Železná B, Kuneš J, Bardová K, Maletínská L. Deficiency of GPR10 and NPFFR2 receptors leads to sex-specific prediabetic syndrome and late-onset obesity in mice. Biosci Rep 2024; 44:BSR20241103. [PMID: 39440369 PMCID: PMC11499387 DOI: 10.1042/bsr20241103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 09/20/2024] [Accepted: 09/24/2024] [Indexed: 10/25/2024] Open
Abstract
GPR10 and neuropeptide FF receptor 2 (NPFFR2) play important role in the regulation of food intake and energy homeostasis. Understanding the interaction between these receptors and their specific ligands, such as prolactin-releasing peptide, is essential for developing stable peptide analogs with potential for treating obesity. By breeding and characterizing double knockout (dKO) mice fed standard or high-fat diet (HFD), we provide insights into the metabolic regulation associated with the GPR10 and NPFFR2 deficiency. Both WT and dKO mice were subjected to behavioral tests and an oral glucose tolerance test. Moreover, dual-energy X-ray absorptiometry (DEXA) followed by indirect calorimetry were performed to characterize dKO mice. dKO mice of both sexes, when exposed to an HFD, showed reduced glucose tolerance, hyperinsulinemia, and insulin resistance compared with controls. Moreover, they displayed increased liver weight with worsened hepatic steatosis. Mice displayed significantly increased body weight, which was more pronounced in dKO males and caused by higher caloric intake on a standard diet, while dKO females displayed obesity characterized by increased white adipose tissue and enhanced hepatic lipid accumulation on an HFD. Moreover, dKO females exhibited anxiety-like behavior in the open field test. dKO mice on a standard diet had a lower respiratory quotient, with no significant changes in energy expenditure. These results provide insights into alterations associated with disrupted GPR10 and NPFFR2 signaling, contributing to the development of potential anti-obesity treatment.
Collapse
MESH Headings
- Animals
- Male
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Receptors, G-Protein-Coupled/deficiency
- Obesity/metabolism
- Obesity/genetics
- Female
- Mice, Knockout
- Mice
- Diet, High-Fat/adverse effects
- Receptors, Neuropeptide/genetics
- Receptors, Neuropeptide/metabolism
- Receptors, Neuropeptide/deficiency
- Prediabetic State/metabolism
- Prediabetic State/genetics
- Energy Metabolism/genetics
- Insulin Resistance
- Mice, Inbred C57BL
- Sex Factors
- Adipose Tissue, White/metabolism
Collapse
Affiliation(s)
- Alena Morgan
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, 166 10 Prague, Czech Republic
| | - Nivasini Shekhar
- Institute of Physiology of the Czech Academy of Sciences, 142 00 Prague, Czech Republic
- Department of Physiology, Faculty of Science, Charles University in Prague, 128 44 Prague, Czech Republic
| | - Veronika Strnadová
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, 166 10 Prague, Czech Republic
| | - Zdenko Pirník
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, 166 10 Prague, Czech Republic
- Biomedical Research Center, Slovak Academy of Sciences, 845 05 Bratislava, Slovak Republic
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, 813 72 Bratislava, Slovak Republic
| | - Eliška Haasová
- Institute of Physiology of the Czech Academy of Sciences, 142 00 Prague, Czech Republic
- Department of Physiology, Faculty of Science, Charles University in Prague, 128 44 Prague, Czech Republic
| | - Jan Kopecký
- Institute of Physiology of the Czech Academy of Sciences, 142 00 Prague, Czech Republic
| | - Andrea Pačesová
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, 166 10 Prague, Czech Republic
| | - Blanka Železná
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, 166 10 Prague, Czech Republic
| | - Jaroslav Kuneš
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, 166 10 Prague, Czech Republic
- Institute of Physiology of the Czech Academy of Sciences, 142 00 Prague, Czech Republic
| | - Kristina Bardová
- Institute of Physiology of the Czech Academy of Sciences, 142 00 Prague, Czech Republic
| | - Lenka Maletínská
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, 166 10 Prague, Czech Republic
| |
Collapse
|
215
|
Tampé JF, Monni E, Palma-Tortosa S, Brogårdh E, Böiers C, Lindgren AG, Kokaia Z. Human monocyte subtype expression of neuroinflammation- and regeneration-related genes is linked to age and sex. PLoS One 2024; 19:e0300946. [PMID: 39475881 PMCID: PMC11524521 DOI: 10.1371/journal.pone.0300946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 09/22/2024] [Indexed: 11/02/2024] Open
Abstract
Aging profoundly affects the immune system leading to an increased propensity for inflammation. Age-related dysregulation of immune cells is implicated in the development and progression of numerous age-related diseases such as: cardiovascular diseases, neurodegenerative disorders, and metabolic syndromes. Monocytes and monocyte-derived macrophages, being important players in the inflammatory response, significantly influence the aging process and the associated increase in inflammatory disease risk. Ischemic stroke is among age-related diseases where inflammation, particularly monocyte-derived macrophages, plays an important deteriorating role but could also strongly promote post-stroke recovery. Also, biological sex influences the incidence, presentation, and outcomes of ischemic stroke, reflecting both biological differences between men and women. Here, we studied whether human peripheral blood monocyte subtype (classical, intermediate, and non-classical) expression of genes implicated in stroke-related inflammation and post-stroke tissue regeneration depends on age and sex. A flow cytometry analysis of blood samples from 44 healthy volunteers (male and female, aged 28 to 98) showed that in contrast to other immune cells, the proportion of NK-cells increased in females. The proportion of B-cells decreased in both sexes with age. Gene expression analysis by qPCR identified several genes differentially correlating with age and sex within different monocyte subtypes. Interestingly, ANXA1 and CD36 showed a consistent increase with aging in all monocytes, specifically in intermediate (CD36) and intermediate and non-classical (ANXA1) subtypes. Other genes (IL-1β, S100A8, TNFα, CD64, CD33, TGFβ1, TLR8, CD91) were differentially changed in monocyte subtypes with increasing age. Most age-dependent gene changes were differentially expressed in female monocytes. Our data shed light on the nuanced interplay of age and sex in shaping the expression of inflammation- and regeneration-related genes within distinct monocyte subtypes. Understanding these dynamics could pave the way for targeted interventions and personalized approaches in post-stroke care, particularly for the aging population and individuals of different sexes.
Collapse
Affiliation(s)
- Juliane F. Tampé
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Emanuela Monni
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Sara Palma-Tortosa
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Emil Brogårdh
- Department of Neurology, Skåne University Hospital; Department of Clinical Sciences Lund, Neurology, Lund University, Lund, Sweden
| | - Charlotta Böiers
- Division of Molecular Hematology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Arne G. Lindgren
- Department of Neurology, Skåne University Hospital; Department of Clinical Sciences Lund, Neurology, Lund University, Lund, Sweden
| | - Zaal Kokaia
- Laboratory of Stem Cells and Restorative Neurology, Lund Stem Cell Center, Lund University, Lund, Sweden
| |
Collapse
|
216
|
Giannini S, Pitino A, Sella S, Fusaro M, Arcidiacono GP, Torres MO, Zaninotto M, Gori M, Aghi A, Egan CG, Simioni P, Tripepi G, Plebani M. Sex-related differences in vitamin D testing in the Veneto Region, Italy: a retrospective analysis from 2005 to 2016. Arch Osteoporos 2024; 19:105. [PMID: 39477860 PMCID: PMC11525240 DOI: 10.1007/s11657-024-01460-w] [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: 07/03/2024] [Accepted: 10/09/2024] [Indexed: 11/02/2024]
Abstract
A retrospective analysis was performed to evaluate the frequency of vitamin D blood testing in individuals from the Padua province, Veneto, Italy from 2005 to 2016. A significant increase in the frequency of vitamin D blood tests, particularly in females was observed and in individuals with severe vitamin D deficiency (Class I). PURPOSE Vitamin D deficiency has been linked to negative health outcomes that extend beyond bone-related conditions. The frequency of vitamin D blood testing in residents from the Padua province, (Veneto, Italy) from 2005 to 2016 was evaluated. METHODS Data were retrospectively retrieved from blood test databases (Laboratory Medicine Unit, Padua University Hospital) and information on number of vitamin D blood tests performed on residents from 2005 to 2016 was collected. Data were stratified by sex and ten birth cohorts from 1901 to 2016. Blood tests were classified into five vitamin D classes: I < 50 nmol/L, II 50-74.9 nmol/L, III 75-149 nmol/L, IV 150-250 nmol/L, and V > 250-1000 nmol/L. Blood test trends were analyzed as blood test rate and vitamin D class rate/resident population. Population analysis was analyzed by incidence rates and stratified by vitamin D class. RESULTS 293,013 vitamin D blood tests were conducted between 2005 and 2016 across 10 birth cohorts. Females accounted for 75% of tests and fewer were conducted in the youngest and oldest birth cohorts. Sex differences in vitamin D blood test frequency were observed; adjusted rates ranging from 1.7 to 35.6% for males and 8 to 81% for females from 2005 to 2016. Crude incidence rates (per 1000 from 2005 to 2016) varied from 1.5 to 10.8‰ for males and 7 to 19.4‰ for females. Crude blood test rates for vitamin D deficiency (Class I) increased from 1.1 to 9.9‰ in 2016 for males and 5 to 17.3‰ for females. Crude incidence rates (from 2005 to 2016) for Class I were 9.7-57.1‰ in males and 43.6-92.4‰ in females. CONCLUSIONS These findings highlight sex-related differences in vitamin D testing, providing valuable insight for healthcare planning.
Collapse
Affiliation(s)
- Sandro Giannini
- Clinica Medica 1, Department of Medicine, University of Padova, Padua, Italy
| | - Annalisa Pitino
- Institute of Clinical Physiology (IFC), National Research Council (CNR), Rome, Italy
| | - Stefania Sella
- Clinica Medica 1, Department of Medicine, University of Padova, Padua, Italy
| | - Maria Fusaro
- Clinica Medica 1, Department of Medicine, University of Padova, Padua, Italy.
- Institute of Clinical Physiology (IFC), National Research Council (CNR), Pisa, Italy.
| | | | | | | | - Mercedes Gori
- Institute of Clinical Physiology (IFC), National Research Council (CNR), Rome, Italy
| | - Andrea Aghi
- Clinica Medica 1, Department of Medicine, University of Padova, Padua, Italy
| | | | - Paolo Simioni
- Clinica Medica 1, Department of Medicine, University of Padova, Padua, Italy
| | - Giovanni Tripepi
- Institute of Clinical Physiology (IFC), National Research Council (CNR), Reggio Calabria, Italy
| | - Mario Plebani
- QI.Lab.Med, Spin-off of the University of Padova, Padua, Italy
| |
Collapse
|
217
|
Aguilar-Delgadillo A, Cruz-Mendoza F, Luquin-de Andais teh S, Ruvalcaba-Delgadillo Y, Jáuregui-Huerta F. Stress-induced c-fos expression in the medial prefrontal cortex differentially affects the main residing cell phenotypes. Heliyon 2024; 10:e39325. [PMID: 39498004 PMCID: PMC11532284 DOI: 10.1016/j.heliyon.2024.e39325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 10/10/2024] [Accepted: 10/11/2024] [Indexed: 11/07/2024] Open
Abstract
Stress poses a challenge to the body's equilibrium and triggers a series of responses that enable organisms to adapt to stressful stimuli. The medial prefrontal cortex (mPFC), particularly in acute stress conditions, undergoes significant physiological changes to cope with the demands associated with cellular activation. The proto-oncogene c-fos and its protein product c-Fos have long been utilized to investigate the effects of external factors on the central nervous system (CNS). While c-Fos expression has traditionally been attributed to neurons, emerging evidence suggests its potential expression in glial cells. In this study, our main objective was to explore the expression of c-Fos in glial cells and examine how acute stress influences these activity patterns. We conducted our experiments on male Wistar rats, subjecting them to acute stress and sacrificing them 2 h after the stressor initiation. Using double-labelling fluorescent immunohistochemistry targeting c-Fos, along with markers such as GFAP, Iba-1, Olig2, NG2, and NeuN, we analyzed 35 μm brain slices obtained from the mPFC. Our findings compellingly demonstrate that c-Fos expression extends beyond neurons and is present in astrocytes, oligodendrocytes, microglia, and NG2 cells-the diverse population of glial cells. Moreover, we observed distinct regulation of c-Fos expression in response to stress across different subregions of the mPFC. These results emphasize the importance of considering glial cells and their perspective in studies investigating brain activity, highlighting c-Fos as a response marker in glial cells. By shedding light on the differential regulation of c-Fos expression in response to stress, our study contributes to the understanding of glial cell involvement in stress-related processes. This underscores the significance of including glial cells in investigations of brain activity and expands our knowledge of c-Fos as a potential marker for glial cell responses.
Collapse
Affiliation(s)
| | - Fernando Cruz-Mendoza
- Neurosciences Department, Health sciences center, University of Guadalajara, Guadalajara, Mexico
| | | | | | - Fernando Jáuregui-Huerta
- Neurosciences Department, Health sciences center, University of Guadalajara, Guadalajara, Mexico
- Department of Physiology, School of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| |
Collapse
|
218
|
Duncan PJ, Romanò N, Nair SV, McClafferty H, Le Tissier P, Shipston MJ. Long-term, Dynamic Remodelling of the Corticotroph Transcriptome and Excitability After a Period of Chronic Stress. Endocrinology 2024; 165:bqae139. [PMID: 39423299 DOI: 10.1210/endocr/bqae139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/23/2024] [Accepted: 10/16/2024] [Indexed: 10/21/2024]
Abstract
Chronic stress results in long-term dynamic changes at multiple levels of the hypothalamic-pituitary-adrenal (HPA) axis resulting in stress axis dysregulation with long-term impacts on human and animal health. However, the underlying mechanisms and dynamics of altered of HPA axis function, in particular at the level of pituitary corticotrophs, during a period of chronic stress and in the weeks after its cessation (defined as "recovery") are very poorly understood. Here, we address the fundamental question of how a period of chronic stress results in altered anterior pituitary corticotroph function and whether this persists in recovery, as well as the transcriptomic changes underlying this. We demonstrate that, in mice, spontaneous and corticotrophin-releasing hormone-stimulated electrical excitability of corticotrophs, essential for ACTH secretion, is suppressed for weeks to months of recovery following a period of chronic stress. Surprisingly, there are only modest changes in the corticotroph transcriptome during the period of stress, but major alterations occur in recovery. Importantly, although transcriptional changes for a large proportion of mRNAs follow the time course suppression of corticotroph excitability, many other genes display highly dynamic transcriptional changes with distinct time courses throughout recovery. Taken together, this suggests that chronic stress results in complex dynamic transcriptional and functional changes in corticotroph physiology, which are highly dynamic for weeks following cessation of chronic stress. These insights provide a fundamental new framework to further understand underlying molecular mechanisms as well approaches to both diagnosis and treatment of stress-related dysfunction of the HPA axis.
Collapse
Affiliation(s)
- Peter J Duncan
- Centre for Discovery Brain Science, Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh EH8 9AG, UK
| | - Nicola Romanò
- Centre for Discovery Brain Science, Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh EH8 9AG, UK
- Zhejiang University-University of Edinburgh Joint Institute, Zhejiang University School of Medicine, Haining 314400, PR China
| | - Sooraj V Nair
- Centre for Discovery Brain Science, Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh EH8 9AG, UK
| | - Heather McClafferty
- Centre for Discovery Brain Science, Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh EH8 9AG, UK
| | - Paul Le Tissier
- Centre for Discovery Brain Science, Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh EH8 9AG, UK
- Zhejiang University-University of Edinburgh Joint Institute, Zhejiang University School of Medicine, Haining 314400, PR China
| | - Michael J Shipston
- Centre for Discovery Brain Science, Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh EH8 9AG, UK
- Zhejiang University-University of Edinburgh Joint Institute, Zhejiang University School of Medicine, Haining 314400, PR China
| |
Collapse
|
219
|
Hu Q, Cong E, Chen J, Ma J, Li Y, Xu Y, Yue C. Genetical effects of sleep traits on postpartum depression: a bidirectional two-sample Mendelian randomization study. BMC Pregnancy Childbirth 2024; 24:711. [PMID: 39478516 PMCID: PMC11523897 DOI: 10.1186/s12884-024-06929-7] [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] [Received: 01/02/2024] [Accepted: 10/24/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Postpartum depression (PPD) is widely recognized as the most prevalent mental health crisis following childbirth and has been linked to sleep disturbances. However, the potential causal relationships between various sleep traits and PPD remain unclear. This study employs a bidirectional two-sample Mendelian randomization (MR) approach to investigate these associations. METHODS The inverse-variance-weighted method was used to evaluate the causally linked sleep traits on postpartum depression. The weighted median, weighted mode, and MR-Egger were used to estimate the robustness of the inverse-variance-weighted method. The leave-one-out method estimated the sensitivity of the result. Cochran's Q method was used for the heterogeneous test. The MR-Egger intercept and MR-PRESSO methods detected the horizontal pleiotropy. RESULTS We examined the genetic causal relationships between nine sleep traits and postpartum depression. Sleep apnea syndrome (OR: 1.122; 95%CI: 1.063-1.185; p = 0.000), sleeplessness/insomnia (OR: 1.465; 95%CI: 1.104-1.943; p = 0.008), and frequency of tiredness/lethargy in last 2 weeks (OR: 1.725; 95%CI: 1.345-2.213; p = 0.000) genetically predicted the increased risk of postpartum depression. The reverse Mendelian randomization analysis showed PPD caused sleeplessness/insomnia (β: 0.006; 95%CI: 0.001-0.010; p = 0.016) and frequency of tiredness/lethargy in last 2 weeks (β: 0.007; 95%CI: 0.002-0.011; p = 0.004). The remaining six sleep traits showed no significant association with PPD. There was no heterogeneity or horizontal pleiotropy. CONCLUSIONS Genetic evidence reveals causal relationships between specific sleep traits and PPD, including sleep apnea syndrome, sleeplessness/insomnia, and tiredness. Whether certain sleep health indicators suggest a risk of postpartum depression or sleep issues that are caused by PPD, both may offer insights into the prevention and treatment of PPD.
Collapse
Affiliation(s)
- Qianying Hu
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Enzhao Cong
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jianhua Chen
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingjing Ma
- Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuting Li
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yifeng Xu
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Chaoyan Yue
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China.
| |
Collapse
|
220
|
Lian X, Bai Y, Du P, Jing Z, Gao J, Liu F, Hu J, Xi Y. Causal influences of testosterone on brain structure change rate: A sex-stratified Mendelian randomization study. J Steroid Biochem Mol Biol 2024; 245:106629. [PMID: 39481491 DOI: 10.1016/j.jsbmb.2024.106629] [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: 07/26/2024] [Revised: 10/26/2024] [Accepted: 10/29/2024] [Indexed: 11/02/2024]
Abstract
The impact of testosterone levels on changes in brain structure has been reported. However, it is still unclear which specific brain region could be affected. This study approached Mendelian randomization method to reveal the causal relationship between testosterone levels and the rate of longitudinal structural changes in the brain. The testosterone-related GWAS data were determined from 425,097 European participants. The GWAS data on the rate of longitudinal structural changes in the brain came from the ENIGMA consortium, which included 15,640 all-age participants from 40 longitudinal cohorts. The inverse variance weighted was considered as the main estimate, MR Egger and weighted median methods were used to supplement IVW. A positive correlation was found between total testosterone levels and bioavailable testosterone levels in women and age-independent longitudinal changes in cerebral WM and surface area. The sex hormone-binding globulin levels were found a negative correlation with age-dependent longitudinal structural changes of cortical GM in men. Additionally, we also found that the bioavailable testosterone level in males was negatively associated with the quadratic age-dependent longitudinal change rate in the globus pallidum. We also found estradiol levels and sex hormone-binding globulin levels were negatively associated with the quadratic age-dependent longitudinal change rate of total brain in men. Moreover, we found a positive correlation between total testosterone levels and linear age-dependent longitudinal changes in the hippocampus in both males and females. The testosterone levels in different genders may have varying degrees of causal effects on the structural changes of brain regions. These findings provide evidence for the influence of the brain glandular axis on brain structure, particularly during female brain development.
Collapse
Affiliation(s)
- Xin Lian
- Department of Obstetrics and Gynecology, People's Hospital of Linyi County, Yuncheng Central Hospital of Shanxi Medical University, 1125 Fuxi Street, Yuncheng 044100, China
| | - Yaqi Bai
- School of Clinical Medicine, Shanxi Medical University, 56 Xinjian South Road, Taiyuan 030000, China
| | - Pengyang Du
- Department of Neurology, Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan 030001, China
| | - Zhinan Jing
- School of Clinical Medicine, Shanxi Medical University, 56 Xinjian South Road, Taiyuan 030000, China
| | - Jimi Gao
- Department of Obstetrics and Gynecology, Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan 030001, China
| | - Fan Liu
- Department of Neurology, Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan 030001, China
| | - Jingjing Hu
- Department of Obstetrics and Gynecology, Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan 030001, China.
| | - Yujia Xi
- Department of Urology, Second Hospital of Shanxi Medical University, Male Reproductive Medicine Center, 382 Wuyi Road, Taiyuan 030001, China.
| |
Collapse
|
221
|
Schmid AM, Thomas TA, Blümel S, Erdal NK, Müller SM, Merz CJ, Wolf OT, Brand M, Müller A, Steins-Loeber S. Transfer from goal-directed behavior to stimulus-response habits and its modulation by acute stress in individuals with risky gaming behavior. Sci Rep 2024; 14:26015. [PMID: 39472683 PMCID: PMC11522379 DOI: 10.1038/s41598-024-73899-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 09/23/2024] [Indexed: 11/02/2024] Open
Abstract
Habitual responses towards addiction-related cues play a relevant role in the development and maintenance of addictions. Such automatic responses may be more likely under stress, as stress has been shown to induce a shift from goal-directed to habitual behavior. The current study investigated these mechanisms in risky gaming behavior. Individuals with risky gaming behavior (n = 68), as established by a structured clinical interview, and a matched control group (n = 67) completed a Pavlovian-to-Instrumental Transfer (PIT) paradigm with gaming-related cues and rewards. After the Pavlovian training, participants underwent a stress (Trier Social Stress Test) or control condition before performing the instrumental training and the transfer phase of the PIT paradigm. To assess habitual behavior, the gaming-related rewards were devalued after half of the transfer phase. In both groups, gaming-related cues enhanced the choice of the gaming-related reward and this gaming PIT effect was reduced, however, not eliminated by the devaluation. Unexpectedly, stress did not significantly increase responding for the gaming-related reward in participants aware of the stimulus-outcome associations, however seemed to enhance habitual responding in unaware participants. Our findings underline the relevance of gaming-related cues in triggering habitual responses, which may undermine attempts to change a problematic gaming behavior.
Collapse
Affiliation(s)
- Anna M Schmid
- Department of Clinical Psychology and Psychotherapy, Otto-Friedrich-University of Bamberg, Bamberg, Germany.
| | - Tobias A Thomas
- Department of Psychosomatic Medicine and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Stefan Blümel
- Department of Clinical Psychology and Psychotherapy, Otto-Friedrich-University of Bamberg, Bamberg, Germany
| | - Nicolas K Erdal
- Department of Psychosomatic Medicine and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Silke M Müller
- General Psychology: Cognition, Faculty of Computer Science, University of Duisburg-Essen, Duisburg, Germany
- Center for Behavioral Addiction Research (CeBAR), Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Erwin L. Hahn Institute for Magnetic Resonance Imaging, Essen, Germany
| | - Christian J Merz
- Department of Cognitive Psychology, Institute of Cognitive Neuroscience, Faculty of Psychology, Ruhr University Bochum, Bochum, Germany
| | - Oliver T Wolf
- Department of Cognitive Psychology, Institute of Cognitive Neuroscience, Faculty of Psychology, Ruhr University Bochum, Bochum, Germany
| | - Matthias Brand
- General Psychology: Cognition, Faculty of Computer Science, University of Duisburg-Essen, Duisburg, Germany
- Center for Behavioral Addiction Research (CeBAR), Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Erwin L. Hahn Institute for Magnetic Resonance Imaging, Essen, Germany
| | - Astrid Müller
- Department of Psychosomatic Medicine and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Sabine Steins-Loeber
- Department of Clinical Psychology and Psychotherapy, Otto-Friedrich-University of Bamberg, Bamberg, Germany
| |
Collapse
|
222
|
Sreeya Devarakonda S, Basha S, Pithakumar A, Thoshna, Mukunda DC, Rodrigues J, Ameera K, Biswas S, Pai AR, Belurkar S, Mahato KK. Molecular Mechanisms of Neurofilament Alterations and its Application in Assessing Neurodegenerative Disorders. Ageing Res Rev 2024:102566. [PMID: 39481763 DOI: 10.1016/j.arr.2024.102566] [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: 08/09/2024] [Revised: 10/04/2024] [Accepted: 10/24/2024] [Indexed: 11/02/2024]
Abstract
Neurofilaments are intermediate filaments present in neurons. These provide structural support and maintain the size and shape of the neurons. Dysregulation, mutation, and aggregation of neurofilaments raise the levels of these proteins in the blood and cerebrospinal fluid, which are characteristic features of axonal damage and certain rare neurological diseases, such as Giant axonal neuropathy and Charcot-Mare-Tooth disease. Understanding the structure, dynamics, and function of neurofilaments has been greatly enhanced by a diverse range of biochemical and preclinical investigations conducted over more than four decades. Recently, there has been a resurgence of interest in post-translational modifications of neurofilaments, such as phosphorylation, aggregation, mutation, oxidation, etc. Over the past twenty years, several rare disorders have been studied from structural alterations of neurofilaments. These disorders are monitored by fluid biomarkers such as neurofilament light chains. Currently, there are many tools, such as Enzyme-Linked Immunosorbent Assay, Electrochemiluminescence Assay, Single-Molecule Array, Western/immunoblotting, etc., in use to assess the neurofilament proteins in Blood and CSF. However, all these techniques utilize expensive, non-specific, or antibody-based methods, which make them unsuitable for routine screening of neurodegenerative disorders. This provides room to search for newer sensitive, cost-effective, point-of-care tools for rapid screening of the disease. For a long time, the molecular mechanisms of neurofilaments have been poorly understood due to insufficient research attempts, and a deeper understanding of them remains elusive. Therefore, this review aims to highlight the available literature on molecular mechanisms of neurofilaments and the function of neurofilaments in axonal transport, axonal conduction, axonal growth, and neurofilament aggregation, respectively. Further, this review discusses the role of neurofilaments as potential biomarkers for the identification of several neurodegenerative diseases in clinical laboratory practice.
Collapse
Affiliation(s)
| | - Shaik Basha
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Karnataka, Manipal, 576 104, India
| | - Anjana Pithakumar
- Manipal School of Life Sciences, Manipal Academy of Higher Education, Karnataka, Manipal, 576 104, India
| | - Thoshna
- Manipal School of Life Sciences, Manipal Academy of Higher Education, Karnataka, Manipal, 576 104, India
| | | | - Jackson Rodrigues
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Karnataka, Manipal, 576 104, India
| | - K Ameera
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Karnataka, Manipal, 576 104, India
| | - Shimul Biswas
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Karnataka, Manipal, 576 104, India
| | - Aparna Ramakrishna Pai
- Department of Neurology, Kasturba Medical College-Manipal, Manipal Academy of Higher Education, Karnataka, Manipal, 576 104, India
| | - Sushma Belurkar
- Department of Pathology, Kasturba Medical College-Manipal, Manipal Academy of Higher Education, Karnataka, Manipal, 576 104, India
| | - Krishna Kishore Mahato
- Department of Biophysics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Karnataka, Manipal, 576 104, India.
| |
Collapse
|
223
|
Drexhage HA, Bergink V, Poletti S, Benedetti F, Osborne LM. Conventional and new immunotherapies for immune system dysregulation in postpartum mood disorders: comparisons to immune system dysregulations in bipolar disorder, major depression, and postpartum autoimmune thyroid disease. Expert Rev Clin Immunol 2024:1-23. [PMID: 39441185 DOI: 10.1080/1744666x.2024.2420053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 09/17/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
INTRODUCTION Postpartum mood disorders are heterogenous disorders and comprise postpartum psychosis and postpartum depression. Evidence is accumulating that systemic monocyte/macrophage activation, low-grade inflammation and (premature senescence related) T cell defects increase the risk for mood disorders outside pregnancy by affecting the function of microglia and T cells in the emotional brain (the cortico-limbic system) leading to inadequate mood regulation upon stress. AREAS COVERED The evidence in the literature that similar immune dysregulations are present in postpartum mood disorders. RESULTS The physiological postpartum period is characterized by a rapid T cell surge and a mild activation of the monocyte/macrophage system. Postpartum mood disorder patients show a diminished T cell surge (including that of T regulatory cells) and an increase in low grade inflammation, that is, an increased inflammatory state of monocytes/macrophages and higher levels of serum pro-inflammatory cytokines. EXPERT OPINION Anti-inflammatory agents (e.g. COX-2 inhibitors) and T cell boosting agents (e.g. low-dose IL-2 therapy) should be further investigated as treatment. The hypothesis should be investigated that postpartum mood disorders are active episodes (triggered by changes in the postpartum immuno-endocrine milieu) in ongoing, dynamically fluctuating aberrant neuro-immune-endocrine trajectories leading to mood disorders in women (inheritably) vulnerable to these disorders.
Collapse
Affiliation(s)
- Hemmo A Drexhage
- Department of Immunology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Veerle Bergink
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Sara Poletti
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milano, Italy
- Università Vita-Salute San Raffaele, Milano, Italy
| | - Francesco Benedetti
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milano, Italy
- Università Vita-Salute San Raffaele, Milano, Italy
| | - Lauren M Osborne
- Departments of Obstetrics and Gynecology and of Psychiatry, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
224
|
Reimert I, Bolhuis JE. Possible relations between emotional contagion and social buffering. Sci Rep 2024; 14:25944. [PMID: 39472724 PMCID: PMC11522516 DOI: 10.1038/s41598-024-77394-7] [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: 04/07/2024] [Accepted: 10/22/2024] [Indexed: 11/02/2024] Open
Abstract
Emotional contagion can be defined as the transfer of an emotional state from the demonstrator of that state towards an observer. Social buffering is a process by which the demonstrator has a reduced stress response due to the presence of one or more other individuals. While both processes are well studied separately, it is unknown whether and how both processes are related. Therefore, the aim of this study was to investigate the relation between emotional contagion and social buffering in pigs. Hereto correlations were performed between measures of emotional contagion (i.e., the difference in behaviour of observer pigs between a situation with and without demonstrator pigs present) and measures of social buffering (i.e., the difference in behaviour of demonstrator pigs in a negative situation with and without observer pigs present). The results did not point towards a clear and consistent relation as only few and contrasting correlations between measures of emotional contagion and social buffering were found, and after correcting for chance no significant correlations remained. To conclude, more research is needed on the relation between emotional contagion and social buffering to shed light on how and when emotions will spread through and/or are buffered in a group of animals.
Collapse
Affiliation(s)
- Inonge Reimert
- Adaptation Physiology Group, Department of Animal Sciences, Wageningen University and Research, PO Box 338, Wageningen, 6700 AH, The Netherlands.
| | - J Elizabeth Bolhuis
- Adaptation Physiology Group, Department of Animal Sciences, Wageningen University and Research, PO Box 338, Wageningen, 6700 AH, The Netherlands
| |
Collapse
|
225
|
Hohl CH, Zilcha-Mano S, Delgadillo J. Is the "social hormone" oxytocin relevant to psychotherapy treatment outcomes? A systematic review of observational and experimental studies. Neurosci Biobehav Rev 2024:105935. [PMID: 39481670 DOI: 10.1016/j.neubiorev.2024.105935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 10/27/2024] [Accepted: 10/29/2024] [Indexed: 11/02/2024]
Abstract
BACKGROUND Oxytocin, popularly known as the "social hormone", has wide implications for the regulation of socially relevant cognitions, emotions and behaviors. Individual differences in oxytocin may be relevant to mental health treatment outcomes, given the centrality of the therapeutic relationship in psychotherapy. METHODS This systematic review aimed to synthesize findings from psychotherapy studies that examined oxytocin measurement and augmentation methods and their association with treatment outcomes. The methodology was preregistered in the Open Science Framework ((https://osf.io/xtyvc/?view_only=2bc37dc0b2cd41f8939e2964bd8b884f). Five databases were searched on 30th of March 2023 (PubMed, SCOPUS, Web of Science, Medline, PsycINFO). Eligible studies were assessed for risk of bias and findings were summarized using narrative synthesis and vote counting methods. RESULTS Overall, 24 studies (n=881 participants) including experimental and observational designs and covering various diagnostic groups were reviewed. Findings from 9 studies (n=406) indicate that oxytocin measures were associated with psychotherapy treatment outcomes for depression, and oxytocin-augmentation improved depression outcomes. Results regarding other mental disorders were mixed and inconclusive. DISCUSSION Current evidence indicates that oxytocin-augmented psychotherapy for depression warrants further research. Currently there is not sufficient evidence to draw firm conclusions regarding the clinical relevance of oxytocin in the context of other disorders. Key limitations are the lack of meta-analytic synthesis and small sample sizes for primary studies.
Collapse
Affiliation(s)
- Caio Hummel Hohl
- Department of Psychology, University of Sheffield, United Kingdom
| | | | - Jaime Delgadillo
- Department of Psychology, University of Sheffield, United Kingdom
| |
Collapse
|
226
|
Stefanakis K, Upadhyay J, Cisneros AR, Patel N, Sahai A, Mantzoros CS. Leptin physiology and pathophysiology in energy homeostasis, immune function, neuroendocrine regulation and bone health. Metabolism 2024:156056. [PMID: 39481533 DOI: 10.1016/j.metabol.2024.156056] [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: 10/04/2024] [Revised: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 11/02/2024]
Abstract
Since its discovery and over the past thirty years, extensive research has significantly expanded our understanding of leptin and its diverse roles in human physiology, pathophysiology and therapeutics. A prototypical adipokine initially identified for its critical function in appetite regulation and energy homeostasis, leptin has been revealed to also exert profound effects on the hypothalamic-pituitary-gonadal, thyroid, adrenal and growth hormone axis, differentially between animals and humans, as well as in regulating immune function. Beyond these roles, leptin plays a pivotal role in significantly affecting bone health by promoting bone formation and regulating bone metabolism both directly and indirectly through its neuroendocrine actions. The diverse actions of leptin are particularly notable in leptin-deficient animal models and in conditions characterized by low circulating leptin levels, such as lipodystrophies and relative energy deficiency. Conversely, the effectiveness of leptin is attenuated in leptin-sufficient states, such as obesity and other high-adiposity conditions associated with hyperleptinemia and leptin tolerance. This review attempts to consolidate 30 years of leptin research with an emphasis on its physiology and pathophysiology in humans, including its promising therapeutic potential. We discuss preclinical and human studies describing the pathophysiology of energy deficiency across organ systems and the significant role of leptin in regulating neuroendocrine, immune, reproductive and bone health. We finally present past proof of concept clinical trials of leptin administration in leptin-deficient subjects that have demonstrated positive neuroendocrine, reproductive, and bone health outcomes, setting the stage for future phase IIb and III randomized clinical trials in these conditions.
Collapse
Affiliation(s)
- Konstantinos Stefanakis
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jagriti Upadhyay
- Department of Medicine, Lahey Hospital and Medical Center, Burlington, MA, USA
| | - Arantxa Ramirez Cisneros
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Nihar Patel
- Department of Medicine, Lahey Hospital and Medical Center, Burlington, MA, USA
| | - Akshat Sahai
- Vassar Brothers Medical Center, Poughkeepsie, NY, USA
| | - Christos S Mantzoros
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Department of Medicine, Boston VA Healthcare System, Boston, MA, USA.
| |
Collapse
|
227
|
Compagno MK, Silver CR, Cox-Holmes A, Basso KB, Bishop C, Bernstein AM, Carley A, Cazorla J, Claydon J, Crane A, Crespi C, Curley E, Dolezel T, Franck E, Heiden K, Huffstetler CM, Loeven AM, May CA, Maykut N, Narvarez A, Pacheco FA, Turner O, Fadool DA. Maternal ingestion of cannabidiol (CBD) in mice leads to sex-dependent changes in memory, anxiety, and metabolism in the adult offspring, and causes a decrease in survival to weaning age. Pharmacol Biochem Behav 2024:173902. [PMID: 39481653 DOI: 10.1016/j.pbb.2024.173902] [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: 07/09/2024] [Revised: 09/30/2024] [Accepted: 10/24/2024] [Indexed: 11/02/2024]
Abstract
RATIONALE The consequences of perinatal cannabidiol (CBD) exposure are severely understudied, but are important, given its widespread use and believed safety as a natural supplement. OBJECTIVE The objective of this study was to test the health, metabolic, and behavioral consequences of perinatal CBD exposure on dams and their offspring raised to adult. METHODS Primiparous female C57BL/6 J mice were orally administered 100 mg/kg CBD in strawberry jam to expose offspring during gestation, lactation, or both using a cross-fostering design. Adult offspring were metabolically profiled using indirect calorimetry and intraperitoneal glucose tolerance testing. Adults were behaviorally phenotyped, video recorded, and mouse position tracked using DeepLabCut. RESULTS CBD was detected in maternal plasma using LC-MS 10-min post consumption (34.2 ± 1.7 ng/ul) and peaked within 30 min (371.0 ± 34.0 ng/ul). Fetal exposure to CBD significantly decreased survival of the pups, and decreased male postnatal development, but did not alter litter size, maternal body weight or pup birth weight. We observed many sex-dependent effects of perinatal CBD exposure. Exposure to CBD during gestation and lactation increased meal size, caloric intake, and respiratory exchange ratio for adult male offspring, while exposure during lactation decreased fasting glucose, but had no effect on clearance. Adult female offspring exposed to CBD during lactation showed increased drink size. Perinatal CBD exposure increased obsessive compulsive- and decreased anxiety-like behaviors (marble burying, light-dark box, elevated-plus maze) in female mice, decreased long-term object memory in male mice, and had no effect on attention tasks for either sex. CONCLUSIONS We conclude that orally-administered CBD during pregnancy affects behavior and metabolism in a sex-dependent manner, and mice are differentially sensitive to exposure during gestation vs. lactation, or both. Because long-term changes are observed following perinatal exposure to the drug, and exposure significantly decreases survival to weaning, more research during development is warranted.
Collapse
Affiliation(s)
| | - Claudia Rose Silver
- Interdisciplinary Medical Sciences, Florida State University, Tallahassee, FL 32306, USA.
| | - Alexis Cox-Holmes
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA.
| | - Kari B Basso
- Department of Chemistry, University of Florida, Gainesville, FL 32611, USA.
| | - Caroline Bishop
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA.
| | | | - Aidan Carley
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA.
| | - Joshua Cazorla
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA.
| | - Jenna Claydon
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA.
| | - Ashleigh Crane
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA.
| | - Chloe Crespi
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA.
| | - Emma Curley
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA.
| | - Tyla Dolezel
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA; Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA.
| | - Ezabelle Franck
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA; Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA.
| | - Katie Heiden
- Department of Chemistry, University of Florida, Gainesville, FL 32611, USA.
| | | | - Ashley M Loeven
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA.
| | - Camilla Ann May
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA.
| | - Nicholas Maykut
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA.
| | - Alejandro Narvarez
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA; Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA.
| | - Franklin A Pacheco
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA.
| | - Olivia Turner
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA.
| | - Debra Ann Fadool
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL 32306, USA; Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA; Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA.
| |
Collapse
|
228
|
Ramirez G, Okpara C, Arnett M, Segvich DM, Deosthale P, González PO, Kritikos AE, Melo JB, Sanz N, Pin F, Wallace JM, Plotkin LI. Independent contribution of gonads and sex chromosomes to sex differences in bone mass and strength in the four-core genotypes mouse model. J Bone Miner Res 2024; 39:1659-1672. [PMID: 39255371 PMCID: PMC11523188 DOI: 10.1093/jbmr/zjae147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/19/2024] [Accepted: 08/07/2024] [Indexed: 09/12/2024]
Abstract
Vertebrate sexual dimorphism is ascribed to the presence of testes or ovaries, and, hence, to the secretion of gonad-specific hormones. However, mounting evidence indicates that sex differences in tissues and organs also stem from the presence of sex chromosomes (XX or XY). To tease out the contribution of gonads from sex chromosomes to the musculoskeletal system, we used the Four-Core Genotypes (FCG) mouse model, in which the Sry gene, which dictates testis formation, was either deleted from the Y chromosome, resulting in XY mice with ovaries (XY-SryO), or overexpressed in XX mice, resulting in XX mice with testes (XXT), together with gonadal males with XY-SryT (Sry deletion and overexpression of the Sry transgene in chromosome 3) and females with XXO. The FCG mice are generated by crossing XXO with XY-SryT mice, all of C57BL/6 J background. We now show that the musculoskeletal phenotype of 2- to 4-mo-old FCG mice varies based on both gonads and sex chromosomes, depending on the age and the organ/tissue/cell analyzed. The effect of sex chromosomes on body weight, fat and lean/skeletal muscle mass, and bone mass and structure is minor in 2-/3-mo-old mice, soon after sexual maturation. The contribution of sex chromosomes (XX vs XY-Sry in mice with the same gonads and sex hormones) to several of our measurements becomes apparent in adult 4-mo-old mice. The contribution of 1X and 1Y-Sry vs 2X chromosomes varies among different measurements in gonadal males or females, and mice with XY-Sry chromosomes might have higher or lower values that XX mice. Our study shows XX vs XY-Sry chromosome contribution to the musculoskeletal phenotype, which becomes more evident as the animals reach peak bone mass, suggesting that although gonadal sex has a major role, sex chromosomes are also an unrecognized contributor to musculoskeletal mass and bone strength.
Collapse
Affiliation(s)
- Gabriel Ramirez
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Chiebuka Okpara
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Matthew Arnett
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Dyann M Segvich
- Department of Biomedical Engineering, Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, United States
| | - Padmini Deosthale
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Paola Ortiz González
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Alexander E Kritikos
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Julian Balanta Melo
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
- Dentistry Unit, Universidad del Valle School of Dentistry, Cali, Valle de Cauca 760042, Colombia
| | - Natasha Sanz
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
- Bone Biology Laboratory. School of Medicine, Rosario National University, Rosario, Santa Fe 2000, Argentina
| | - Fabrizio Pin
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Joseph M Wallace
- Department of Biomedical Engineering, Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, United States
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN 46202, United States
- Roudebush Veterans Administration Medical Center, Indianapolis, IN 46202, United States
| | - Lilian I Plotkin
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN 46202, United States
- Roudebush Veterans Administration Medical Center, Indianapolis, IN 46202, United States
| |
Collapse
|
229
|
Arnold ME, Harber CE, Beugelsdyk LA, Decker Ramirez EB, Phillips GB, Schank JR. Corticotropin releasing hormone receptor 1 in the medial prefrontal cortex mediates aversion resistant alcohol intake. Psychopharmacology (Berl) 2024:10.1007/s00213-024-06707-5. [PMID: 39466414 DOI: 10.1007/s00213-024-06707-5] [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: 07/01/2024] [Accepted: 10/18/2024] [Indexed: 10/30/2024]
Abstract
RATIONALE Alcohol consumption despite negative consequences is a core symptom of Alcohol Use Disorder. In animal models, this is studied by pairing aversive stimuli with alcohol access, and continuation of drinking under these conditions is known as aversion resistance. Previously, we found that female mice are more aversion resistant than males. Corticotropin releasing hormone (Crh) and the Crh receptor 1 (Crhr1) regulate stress-induced reinstatement, alcohol dependence, and binge-like drinking. However, the role of the Crh system in aversion resistance has not been assessed. OBJECTIVES We aimed to identify sex differences in the Crh system during quinine-adulterated alcohol intake. METHODS We used two-bottle choice and adulterated the alcohol solution with quinine. Next, we measured Crh and Crhr1 levels in brain tissue using real-time polymerase chain reaction (RT-qPCR) and RNAscope in situ hybridization. We then infused a Crhr1 antagonist into the medial prefrontal cortex (mPFC) prior to quinine-alcohol intake. RESULTS After quinine-alcohol consumption, females exhibited increased mPFC Crhr1 mRNA levels as measured by RT-qPCR. This was confirmed with greater anatomical specificity using RNAscope, with females exhibiting an increased number of Crhr1 + cells in the dorsomedial PFC and the ventromedial PFC. mPFC Crhr1 antagonist treatment reduced quinine-alcohol consumption in females but did not impact consumption in males. Quinine-free alcohol intake was unaffected by Crhr1 antagonist treatment. CONCLUSIONS Our findings suggest that Crhr1 in mPFC plays a role in aversion resistant alcohol intake in females. Future experiments will examine the sources of Crh innervation to the mPFC and their distinct roles in alcohol seeking.
Collapse
Affiliation(s)
- Miranda E Arnold
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, 501 DW Brooks Drive, Athens, GA, 30602, USA
| | - Cecelia E Harber
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, 501 DW Brooks Drive, Athens, GA, 30602, USA
| | - Lauren A Beugelsdyk
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, 501 DW Brooks Drive, Athens, GA, 30602, USA
| | - Ellie B Decker Ramirez
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, 501 DW Brooks Drive, Athens, GA, 30602, USA
| | - Grace B Phillips
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, 501 DW Brooks Drive, Athens, GA, 30602, USA
| | - Jesse R Schank
- Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, 501 DW Brooks Drive, Athens, GA, 30602, USA.
| |
Collapse
|
230
|
Buyukeren M, Ozcan B, Can U, Kenar A, Kececi R, Bayman MG, Gunenc O. Comparison of cord blood alarin levels of full-term infants according to birth weight. J Perinat Med 2024:jpm-2024-0236. [PMID: 39455426 DOI: 10.1515/jpm-2024-0236] [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: 05/27/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024]
Abstract
OBJECTIVES To compare the cord blood alarin levels of infants in different birth weight groups with those of infants born to mothers diagnosed with gestational diabetes mellitus (GDM) who were not subgrouped according to birth weight. METHODS This prospective study was conducted between September 2023 and January 2024. Healthy term babies whose families agreed to participate in the study were divided into four groups according to their birth weight (small for gestational age (SGA), appropriate for gestational age (AGA) and large for gestational age (LGA)) and whether their mothers had GDM. RESULTS There was a significant difference between the cord blood alarin levels of the AGA and SGA groups (p=0.014). There was also a significant difference between the cord blood alarin levels of the AGA and GDM groups (p=0.012). However, the cord blood alarin levels of the LGA group (whose mothers did not have GDM) were similar to those of the AGA group (p=0.394). CONCLUSIONS We found evidence that alarin levels in umbilical cord blood are associated with low birth weight and GDM.
Collapse
Affiliation(s)
- Melek Buyukeren
- Department of Pediatrics, Division of Neonatology, Konya City Hospital, Konya, Türkiye
| | - Beyza Ozcan
- Department of Pediatrics, Division of Neonatology, Konya City Hospital, Konya, Türkiye
| | - Ummugulsum Can
- Department of Biochemistry, Konya City Hospital, Konya, Türkiye
| | - Aytac Kenar
- Department of Pediatrics, Division of Neonatology, Konya City Hospital, Konya, Türkiye
| | - Ramazan Kececi
- Department of Pediatrics, Division of Neonatology, Konya City Hospital, Konya, Türkiye
| | | | - Oguzhan Gunenc
- Clinic of Obstetrics and Gynecology, Konya City Hospital, Konya, Türkiye
| |
Collapse
|
231
|
Joshi A, Kathuria D, Paul M, Singh N. An overview on the potential application of nanotechnology in enhancing the therapeutic efficacy of phytoestrogens. Food Chem 2024; 464:141779. [PMID: 39481307 DOI: 10.1016/j.foodchem.2024.141779] [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: 05/23/2024] [Revised: 09/20/2024] [Accepted: 10/23/2024] [Indexed: 11/02/2024]
Abstract
Phytoestrogens, derived from plants possesses structural similarity with 17 β-estradiol found in mammals. It is abundantly present in soybean along with red clove, alfalfa as well as other legumes, nuts, vegetables and seeds. It is used as hormone replacement therapy and exhibits both anti-estrogenic and estrogenic properties that linked to therapeutic benefits as well as plays active role in sports nutrition. Despite the potential benefits of phytoestrogens, their low solubility, bioavailability, and stability make it challenging to target them effectively. Recent advancements in nanotechnology have paved in facilitating target delivery. Scaling at nano level offered greater surface area, improved solubility, and bioavailability of phytoestrogens which has ultimately reduced the required medication dosage, and enhanced cost-effectiveness, particularly for expensive bioactive substances where precise dosages are recommended. The present article discussed about the potential application of nanotechnology in enhancing therapeutic benefits of phytoestrogens while minimizing their potential side effects.
Collapse
Affiliation(s)
- Aroma Joshi
- Department of Food Science and Technology, Graphic Era (Deemed to be University), Dehradun, Uttarakhand 248002, India
| | - Deepika Kathuria
- Department of Food Science and Technology, Graphic Era (Deemed to be University), Dehradun, Uttarakhand 248002, India
| | - Maman Paul
- Department of Physiotherapy, Guru Nanak Dev University, Amritsar, Punjab 143005, India
| | - Narpinder Singh
- Department of Food Science and Technology, Graphic Era (Deemed to be University), Dehradun, Uttarakhand 248002, India.
| |
Collapse
|
232
|
Wang S, Xu J, Wang X, Wang M, Xue H, Wu M, Fan C, Chen L, Xu L. cba-miR-222-3p involved in photoperiod-induced apoptosis in testes of striped hamsters by targeting TRAF7. Integr Zool 2024. [PMID: 39466916 DOI: 10.1111/1749-4877.12918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
The role of miRNAs in the regulation of seasonal reproduction in rodents, particularly in relation to photoperiod changes, is still poorly understood. Previous studies on miRNA transcriptomes of striped hamster (Cricetulus barabensis) testes have indicated that the photoperiodism of testes, especially apoptosis, may be influenced by miRNAs. As a functional miRNA, cba-miR-222-3p in striped hamster testes exhibits suppression under a short photoperiod. To elucidate the potential role of testicular cba-miR-222-3p in the seasonal reproduction of striped hamsters, we exposed male striped hamsters to different photoperiods or injected miRNA agomir into the testes and observed the effects of these treatments, particularly some indicators related to apoptosis. The results showed that the levels of apoptosis in the testes increased in short daylength, accompanied by a significant decrease in cba-miR-222-3p expression and an increase in TRAF7 expression. Dual luciferase reporter assays verified the targeting relationship between cba-miR-222-3p and TRAF7 predicted by bioinformatics. In addition, the expression of TRAF7 decreased in the testes, which injected miRNA agomir, leading to inhibition of apoptosis, and the expression of key genes (MEKK3, p38, p53) in the downstream MAPK signaling pathway of TRAF7 was suppressed. These results suggest that short daylength induces testicular apoptosis in striped hamsters, and one possible mechanism is that the decreased expression of miR-222-3p in testes reduces the repression of TRAF7 translation, thereby activating the MAPK pathway and affecting the level of testicular apoptosis. These findings reveal the potential role of miR-222-3p in animal reproduction and provide new insights into the regulation of rodent populations.
Collapse
Affiliation(s)
- Shuo Wang
- School of Life Sciences, Qufu Normal University, Qufu, Shandong, China
| | - Jinhui Xu
- School of Life Sciences, Qufu Normal University, Qufu, Shandong, China
| | - Xingchen Wang
- School of Life Sciences, Qufu Normal University, Qufu, Shandong, China
| | - Mingdi Wang
- School of Life Sciences, Qufu Normal University, Qufu, Shandong, China
| | - Huiliang Xue
- School of Life Sciences, Qufu Normal University, Qufu, Shandong, China
| | - Ming Wu
- School of Life Sciences, Qufu Normal University, Qufu, Shandong, China
| | - Chao Fan
- School of Life Sciences, Qufu Normal University, Qufu, Shandong, China
| | - Lei Chen
- School of Life Sciences, Qufu Normal University, Qufu, Shandong, China
| | - Laixiang Xu
- School of Life Sciences, Qufu Normal University, Qufu, Shandong, China
| |
Collapse
|
233
|
Salia S, Burke FF, Hinks ME, Randell AM, Matheson MA, Walling SG, Swift-Gallant A. Gut microbiota transfer from the preclinical maternal immune activation model of autism is sufficient to induce sex-specific alterations in immune response and behavioural outcomes. Brain Behav Immun 2024; 123:813-823. [PMID: 39471905 DOI: 10.1016/j.bbi.2024.10.030] [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: 07/16/2024] [Revised: 10/22/2024] [Accepted: 10/26/2024] [Indexed: 11/01/2024] Open
Abstract
The gut microbiome plays a vital role in health and disease, including neurodevelopmental disorders like autism spectrum disorder (ASD). ASD affects 4:1 males-to-females, and sex differences are apparent in gut microbiota composition among ASD individuals and in animal models of this condition, such as the maternal immune activation (MIA) mouse model. However, few studies have included sex as a biological variable when assessing the role of gut microbiota in mediating ASD symptoms. Using the MIA model of ASD, we assessed whether gut microbiota contributes to the sex differences in the presentation of ASD-like behaviors. Gut microbiota transplantation from MIA or vehicle/control male and female mice into healthy, otherwise unmanipulated, 4-week-old C57Bl/6 mice was performed for 6 treatments over 12 days. Colonization with male, but not female, MIA microbiota was sufficient to reduce sociability, decrease microbiota diversity and increase neuroinflammation with more pronounced deficits in male recipients. Colonization with both male and female donor microbiota altered juvenile ultrasonic vocalizations and anxiety-like behavior in recipients of both sexes, and there was an accompanied change in the gut microbiota and serum cytokine IL-4 and IL-7 levels of all recipients of MIA gut microbiota. In addition to the increases in gut microbes associated with pathological states, the female donor microbiota profile also had increases in gut microbes with known neural protective effects (e.g., Lactobacillus and Rikenella). These results suggest that gut reactivity to environmental insults, such as in the MIA model, may play a role in shaping the sex disparity in ASD development.
Collapse
Affiliation(s)
- Stephanie Salia
- Department of Psychology, Memorial University of Newfoundland, 232 Elizabeth Avenue, St. John's, NL A1B 3X9, Canada.
| | - Francine F Burke
- Department of Psychology, Memorial University of Newfoundland, 232 Elizabeth Avenue, St. John's, NL A1B 3X9, Canada
| | - Meagan E Hinks
- Department of Psychology, Memorial University of Newfoundland, 232 Elizabeth Avenue, St. John's, NL A1B 3X9, Canada
| | - Alison M Randell
- Department of Psychology, Memorial University of Newfoundland, 232 Elizabeth Avenue, St. John's, NL A1B 3X9, Canada
| | - Mairead Anna Matheson
- Department of Psychology, Memorial University of Newfoundland, 232 Elizabeth Avenue, St. John's, NL A1B 3X9, Canada
| | - Susan G Walling
- Department of Psychology, Memorial University of Newfoundland, 232 Elizabeth Avenue, St. John's, NL A1B 3X9, Canada
| | - Ashlyn Swift-Gallant
- Department of Psychology, Memorial University of Newfoundland, 232 Elizabeth Avenue, St. John's, NL A1B 3X9, Canada.
| |
Collapse
|
234
|
Sharma R, Berendzen KM, Everitt A, Wang B, Williams G, Wang S, Quine K, Larios RD, Long KLP, Hoglen N, Sulaman BA, Heath MC, Sherman M, Klinkel R, Cai A, Galo D, Caamal LC, Goodwin NL, Beery A, Bales KL, Pollard KS, Willsey AJ, Manoli DS. Oxytocin receptor controls distinct components of pair bonding and development in prairie voles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.25.613753. [PMID: 39399774 PMCID: PMC11468833 DOI: 10.1101/2024.09.25.613753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Oxytocin receptor (Oxtr) signaling influences complex social behaviors in diverse species, including social monogamy in prairie voles. How Oxtr regulates specific components of social attachment behaviors and the neural mechanisms mediating them remains unknown. Here, we examine prairie voles lacking Oxtr and demonstrate that pair bonding comprises distinct behavioral modules: the preference for a bonded partner, and the rejection of novel potential mates. Our longitudinal study of social attachment shows that Oxtr sex-specifically influences early interactions between novel partners facilitating the formation of partner preference. Additionally, Oxtr suppresses promiscuity towards novel potential mates following pair bonding, contributing to rejection. Oxtr function regulates coordinated patterns of gene expression in regions implicated in attachment behaviors and regulates the expression of oxytocin in the paraventricular nucleus of the hypothalamus, a principal source of oxytocin. Thus, Oxtr controls genetically separable components of pair bonding behaviors and coordinates development of the neural substrates of attachment.
Collapse
|
235
|
Wang G, Cao L, Li S, Zhang M, Li Y, Duan J, Li Y, Hu Z, Wu J, Ni J, Lan D, Li T, Lu J. Gut microbiota dysbiosis-mediated ceramides elevation contributes to corticosterone-induced depression by impairing mitochondrial function. NPJ Biofilms Microbiomes 2024; 10:111. [PMID: 39468065 PMCID: PMC11519513 DOI: 10.1038/s41522-024-00582-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: 10/16/2023] [Accepted: 10/10/2024] [Indexed: 10/30/2024] Open
Abstract
The role of gut microbiota (GM) dysbiosis in the pathogenesis of depression has received widespread attention, but the mechanism remains elusive. Corticosterone (CORT)-treated mice showed depression-like behaviors, reduced hippocampal neurogenesis, and altered composition of the GM. Fecal microbial transplantation from CORT-treated mice transferred depression-like phenotypes and their dominant GM to the recipients. Fecal metabolic profiling exposed remarkable increase of gut ceramides in CORT-treated and recipient mice. Oral gavage with Bifidobacterium pseudolongum and Lactobacillus reuteri could induce elevations of gut ceramides in mice. Ceramides-treated mice showed depressive-like phenotypes, significant downregulation of oxidative phosphorylation-associated genes, and hippocampal mitochondrial dysfunction. Our study demonstrated a link between chronic exposure to CORT and its impact on GM composition, which induces ceramides accumulation, ultimately leading to hippocampal mitochondrial dysfunction. This cascade of events plays a critical role in reducing adult hippocampal neurogenesis and is strongly associated with the development of depression-like behaviors.
Collapse
Affiliation(s)
- Guanhao Wang
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Lining Cao
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Shuanqing Li
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Meihui Zhang
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yingqi Li
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Jinjin Duan
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - You Li
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Zhangsen Hu
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Jiaan Wu
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Jianbo Ni
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Danmei Lan
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Tianming Li
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Jianfeng Lu
- Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China.
- Suzhou Institute of Tongji University, Suzhou, China.
| |
Collapse
|
236
|
Campbell I, Sharifpour R, Balda Aizpurua JF, Beckers E, Paparella I, Berger A, Koshmanova E, Mortazavi N, Read J, Zubkov M, Talwar P, Collette F, Sherif S, Phillips C, Lamalle L, Vandewalle G. Regional response to light illuminance across the human hypothalamus. eLife 2024; 13:RP96576. [PMID: 39466317 PMCID: PMC11517251 DOI: 10.7554/elife.96576] [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: 10/29/2024] Open
Abstract
Light exerts multiple non-image-forming biological effects on physiology including the stimulation of alertness and cognition. However, the subcortical circuitry underlying the stimulating impact of light is not established in humans. We used 7 Tesla functional magnetic resonance imaging to assess the impact of variations in light illuminance on the regional activity of the hypothalamus while healthy young adults (N=26; 16 women; 24.3±2.9 y) were completing two auditory cognitive tasks. We find that, during both the executive and emotional tasks, higher illuminance triggered an activity increase over the posterior part of the hypothalamus, which includes part of the tuberomamillary nucleus and the posterior part of the lateral hypothalamus. In contrast, increasing illuminance evoked a decrease in activity over the anterior and ventral parts of the hypothalamus, encompassing notably the suprachiasmatic nucleus and another part of the tuberomammillary nucleus. Critically, the performance of the executive task was improved under higher illuminance and was negatively correlated with the activity of the posterior hypothalamus area. These findings reveal the distinct local dynamics of different hypothalamus regions that underlie the impact of light on cognition.
Collapse
Affiliation(s)
| | | | | | - Elise Beckers
- GIGA-CRC Human Imaging, University of LiègeLiègeBelgium
- Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre Limburg, Maastricht UniversityMaastrichtNetherlands
| | | | - Alexandre Berger
- GIGA-CRC Human Imaging, University of LiègeLiègeBelgium
- Synergia Medical SAMont-Saint-GuibertBelgium
- Institute of Neuroscience (IoNS), Department of Clinical Neuroscience, Université Catholique de Louvain (UCLouvain)Woluwe-Saint-LambertBelgium
| | | | | | - John Read
- GIGA-CRC Human Imaging, University of LiègeLiègeBelgium
| | | | - Puneet Talwar
- GIGA-CRC Human Imaging, University of LiègeLiègeBelgium
| | | | - Siya Sherif
- GIGA-CRC Human Imaging, University of LiègeLiègeBelgium
| | | | | | | |
Collapse
|
237
|
Moran KM, Enstrom AE, Jarrell L, Khashchuluun M, Tran A, Delville Y. Adolescent social stress alters the role of orexin innervation in the hindbrain in male hamsters. J Neuroendocrinol 2024:e13457. [PMID: 39462511 DOI: 10.1111/jne.13457] [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: 05/06/2024] [Revised: 09/05/2024] [Accepted: 10/09/2024] [Indexed: 10/29/2024]
Abstract
Juvenile male hamsters exposed to chronic social stress eat more, gain weight, and have larger fat pads. The purpose of the present study was to address possible changes in food hoarding and orexin/hypocretin innervation in response to social stress. Male hamsters in early adolescence were exposed to a resident-intruder social stress paradigm or control condition daily for 2 weeks. Metabolism-related physiological measures and behaviors were tracked, and brains were immunocytochemically labeled for orexin-A. Our data confirm our previous observations on appetite, weight gain, and obesity, and showed a strong trend toward enhanced food hoarding as in prior studies. In addition, there were no statistically significant differences in orexin innervation in any brain area analyzed. However, unique correlation patterns were observed between orexin innervation and appetite or metabolic outcome. In particular, opposite correlations were observed between groups within the dorsal raphe nucleus, lateral parabrachial nucleus, and nucleus of the solitary tract. These opposite patterns of correlations suggest chronic social stress causes site-specific alterations in synaptic activity in relation with these behaviors.
Collapse
Affiliation(s)
- Kevin M Moran
- Psychology Department, The University of Texas at Austin, Austin, Texas, USA
| | - Ava Elana Enstrom
- Psychology Department, The University of Texas at Austin, Austin, Texas, USA
| | - Leah Jarrell
- Psychology Department, The University of Texas at Austin, Austin, Texas, USA
| | | | - Anna Tran
- Psychology Department, The University of Texas at Austin, Austin, Texas, USA
| | - Yvon Delville
- Psychology Department, The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
238
|
Lewis AR, Goolsby BC, Juarez BH, Lacey MP, O’Connell LA. Infanticide is driven by unfamiliarity with offspring location and associated with androgenic shifts in mimic poison frogs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.11.589025. [PMID: 39484385 PMCID: PMC11526867 DOI: 10.1101/2024.04.11.589025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Infanticide is widespread across the animal kingdom, but the physiological drivers of infanticide versus care or neglect are relatively unexplored. Here, we identified salient environmental and physiological antecedents of infanticide in the mimic poison frog (Ranitomeya imitator), a biparental amphibian in which female parents feed their tadpoles unfertilized eggs. Specifically, we explored potential environmental cues influencing infant-directed behavior by evaluating changes in the frequency of food provisioning and tadpole mortality after either cross-fostering tadpoles between family units or displacing tadpoles within the terraria of their parents. We found that changes in offspring location reduce care and increase infanticide. Specifically, parents fed their displaced offspring less and, in some instances, tadpole mortality increased. We also investigated whether care and infanticide were related to changes in steroid hormone concentrations in an unfamiliar setting. Infanticide of fertilized eggs and hatchlings in the new territory included cannibalism and was associated with lower testosterone concentrations, but not with changes in corticosterone. Overall, our results support earlier findings that familiarity with offspring location drives parental investment in poison frogs, while indicating an association between low androgen levels and infanticidal behavior in an amphibian.
Collapse
Affiliation(s)
- Amaris R. Lewis
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | | | - Bryan H. Juarez
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Madison P. Lacey
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | | |
Collapse
|
239
|
Sweet SR, Biddinger JE, Zimmermann JB, Yu GL, Simerly RB. Early perturbations to fluid homeostasis alter development of hypothalamic feeding circuits with context-specific changes in ingestive behavior. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.25.620307. [PMID: 39484367 PMCID: PMC11527132 DOI: 10.1101/2024.10.25.620307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Drinking and feeding are tightly coordinated homeostatic events and the paraventricular nucleus of the hypothalamus (PVH) represents a possible node of neural integration for signals related to energy and fluid homeostasis. We used TRAP2;Ai14 and Fos labeling to visualize neurons in the PVH and median preoptic nucleus (MEPO) responding to both water deprivation and hunger. Moreover, we determined that structural and functional development of dehydration-sensitive inputs to the PVH from the MEPO precedes those of agouti-related peptide (AgRP) neurons, which convey hunger signals and are known to be developmentally programmed by nutrition. We also determined that osmotic hyperstimulation of neonatal mice led to enhanced AgRP inputs to the PVH in adulthood, as well as disruptions to ingestive behaviors during high-fat diet feeding and dehydration-anorexia. Thus, development of feeding circuits is impacted not only by nutritional signals, but also by early perturbations to fluid homeostasis with context-specific consequences for coordination of ingestive behavior.
Collapse
|
240
|
Kozłowska-Tomczyk K, Borski N, Głód P, Gogola-Mruk J, Ptak A. PGRMC1 and PAQR4 are promising molecular targets for a rare subtype of ovarian cancer. Open Life Sci 2024; 19:20220982. [PMID: 39464509 PMCID: PMC11512499 DOI: 10.1515/biol-2022-0982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 10/29/2024] Open
Abstract
The heterogeneity of ovarian cancer (OC) has made developing effective treatments difficult. Nowadays, hormone therapy plays a growing role in the treatment of OC; however, hormone modulators have had only limited success so far. To provide a more rigorous foundation for hormonal therapy for different OC subtypes, the current study used a series of bioinformatics approaches to analyse the expression profiles of genes encoding membrane progesterone (PGRMC1, progestins and the adipoQ receptor [PAQR] family), and androgen (zinc transporter member 9 [ZIP9], OXER1) receptors. Our work investigated also their prognostic value in the context of OC. We found differences in expression of ZIP9 and OXER1 between different OC subtypes, as well as between patient tumour and normal tissues. Expression of mRNA encoding PAQR7 and PAQR8 in a panel of OC cell lines was below the qPCR detection limit and was downregulated in tumour tissue samples, whereas high expression of PGRMC1 and PAQR4 mRNA was observed in rare subtypes of OC cell lines. In addition, chemical inhibition of PGRMC1 reduced the viability of rare OCs represented by COV434 cells. In conclusion, PGRMC1 and PAQR4 are promising targets for anticancer therapy, particularly for rare subtypes of OC. These findings may reflect differences in the observed responses of various OC subtypes to hormone therapy.
Collapse
Affiliation(s)
- Kamila Kozłowska-Tomczyk
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387, Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Faculty of Biology, Institute of Zoology and Biomedical Sciences, Jagiellonian University, Gronostajowa 9, 30-387, Krakow, Poland
| | - Norbert Borski
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387, Krakow, Poland
| | - Paulina Głód
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387, Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Faculty of Biology, Institute of Zoology and Biomedical Sciences, Jagiellonian University, Gronostajowa 9, 30-387, Krakow, Poland
| | - Justyna Gogola-Mruk
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387, Krakow, Poland
| | - Anna Ptak
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa 9, 30-387, Krakow, Poland
| |
Collapse
|
241
|
Edwards MM, Nguyen HK, Dodson AD, Herbertson AJ, Honeycutt MK, Slattery JD, Rambousek JR, Tsui E, Wolden-Hanson T, Wietecha TA, Graham JL, Tapia GP, Sikkema CL, O'Brien KD, Mundinger TO, Peskind ER, Ryu V, Havel PJ, Khan AM, Taborsky GJ, Blevins JE. Sympathetic innervation of interscapular brown adipose tissue is not a predominant mediator of OT-elicited reductions of body weight gain and adiposity in male diet-induced obese rats. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.12.612710. [PMID: 39345420 PMCID: PMC11430106 DOI: 10.1101/2024.09.12.612710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Recent studies indicate that central administration of oxytocin (OT) reduces body weight (BW) in high fat diet-induced obese (DIO) rodents by reducing energy intake and increasing energy expenditure (EE). Previous studies in our lab have shown that administration of OT into the fourth ventricle (4V; hindbrain) elicits weight loss and stimulates interscapular brown adipose tissue temperature (TIBAT) in DIO rats. We hypothesized that OT-elicited stimulation of sympathetic nervous system (SNS) activation of IBAT contributes to its ability to activate BAT and reduce BW in DIO rats. To test this, we determined the effect of disrupting SNS activation of IBAT on OT-elicited stimulation of TIBAT and reduction of BW in DIO rats. We first confirmed that bilateral surgical SNS denervation to IBAT was successful based on having achieved ≥ 60% reduction in IBAT norepinephrine (NE) content from DIO rats. NE content was selectively reduced in IBAT by 94.7 ± 2.7, 96.8 ± 1.8 and 85.9 ± 6.1% (P<0.05) at 1, 6 and 7-weeks post-denervation, respectively, and was unchanged in liver or inguinal white adipose tissue. We then measured the impact of bilateral surgical SNS denervation to IBAT on the ability of acute 4V OT (1, 5 μg) to stimulate TIBAT in DIO rats. We found that the high dose of 4V OT (5 μg) stimulated TIBAT similarly between sham and denervated rats (P=NS) and that the effects of 4V OT to stimulate TIBAT did not require beta-3 adrenergic receptor signaling. We subsequently measured the effect of bilateral surgical denervation of IBAT on the effect of chronic 4V OT (16 nmol/day) or vehicle infusion to reduce BW, adiposity, and energy intake in DIO rats. Chronic 4V OT reduced BW gain by -7.2 ± 9.6 g and -14.1 ± 8.8 g in sham and denervated rats (P<0.05 vs vehicle treatment), respectively, and this effect was similar between groups (P=NS). These effects were associated with reductions in adiposity and energy intake (P<0.05). Collectively, these findings support the hypothesis that sympathetic innervation of IBAT is not required for central OT to increase BAT thermogenesis and reduce BW gain and adiposity in male DIO rats.
Collapse
Affiliation(s)
- Melise M Edwards
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA
| | - Ha K Nguyen
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA
| | - Andrew D Dodson
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA
| | - Adam J Herbertson
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA
| | - Mackenzie K Honeycutt
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA
| | - Jared D Slattery
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA
| | - June R Rambousek
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA
| | - Edison Tsui
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA
| | - Tami Wolden-Hanson
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA
| | - Tomasz A Wietecha
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
- UW Medicine Diabetes Institute, University of Washington School of Medicine, Seattle, WA 98109
| | - James L Graham
- Department of Nutrition, University of California, Davis, CA 95616, USA
| | - Geronimo P Tapia
- UTEP Systems Neuroscience Laboratory, Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX 79968, USA
- Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA
| | - Carl L Sikkema
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, Washington
| | - Kevin D O'Brien
- Division of Cardiology, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
- UW Medicine Diabetes Institute, University of Washington School of Medicine, Seattle, WA 98109
| | - Thomas O Mundinger
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Elaine R Peskind
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, Washington
| | - Vitaly Ryu
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Peter J Havel
- Department of Nutrition, University of California, Davis, CA 95616, USA
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA
| | - Arshad M Khan
- UTEP Systems Neuroscience Laboratory, Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX 79968, USA
- Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA
| | - Gerald J Taborsky
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - James E Blevins
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| |
Collapse
|
242
|
Schneider E, Balasubramanian R, Ferri A, Cotter PD, Clarke G, Cryan JF. Fibre & fermented foods: differential effects on the microbiota-gut-brain axis. Proc Nutr Soc 2024:1-16. [PMID: 39449646 DOI: 10.1017/s0029665124004907] [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: 10/26/2024]
Abstract
The ability to manipulate brain function through the communication between the microorganisms in the gastrointestinal tract and the brain along the gut-brain axis has emerged as a potential option to improve cognitive and emotional health. Dietary composition and patterns have demonstrated a robust capacity to modulate the microbiota-gut-brain axis. With their potential to possess pre-, pro-, post-, and synbiotic properties, dietary fibre and fermented foods stand out as potent shapers of the gut microbiota and subsequent signalling to the brain. Despite this potential, few studies have directly examined the mechanisms that might explain the beneficial action of dietary fibre and fermented foods on the microbiota-gut-brain axis, thus limiting insight and treatments for brain dysfunction. Herein, we evaluate the differential effects of dietary fibre and fermented foods from whole food sources on cognitive and emotional functioning. Potential mediating effects of dietary fibre and fermented foods on brain health via the microbiota-gut-brain axis are described. Although more multimodal research that combines psychological assessments and biological sampling to compare each food type is needed, the evidence accumulated to date suggests that dietary fibre, fermented foods, and/or their combination within a psychobiotic diet can be a cost-effective and convenient approach to improve cognitive and emotional functioning across the lifespan.
Collapse
Affiliation(s)
| | - Ramya Balasubramanian
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- Food Biosciences Department, Teagasc Food Research Centre, Moorepark, Cork, Ireland
| | - Aimone Ferri
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Paul D Cotter
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Food Biosciences Department, Teagasc Food Research Centre, Moorepark, Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| |
Collapse
|
243
|
Corbett CM, Bozarth SL, West EA. Effects of sex and estrous cycle on action-outcome contingencies. Behav Brain Res 2024; 477:115317. [PMID: 39490537 DOI: 10.1016/j.bbr.2024.115317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/10/2024] [Accepted: 10/25/2024] [Indexed: 11/05/2024]
Abstract
Goal-directed and habitual-like behaviors are both necessary to efficiently and effectively navigate the environment. A dysregulation between these behaviors can lead to an overreliance on habitual-like behaviors and may contribute to symptoms experienced in some neuropsychiatric disorders such as substance use disorder. One behavioral task used to evaluate goal-directed and habitual-like behavior is an action-outcome task, contingency degradation, where an action (i.e., lever press) is degraded by decoupling the receipt of a reward from the action. However, little is known about how male and female rats and females across the estrous cycle respond during contingency degradation training and extinction testing. Here, we investigated how the variable of sex and estrous cycle influences contingency degradation training and extinction testing and the correlation between baseline anxiety-like behaviors and performance on contingency degradation extinction testing in adult male and female Long-Evans rats. We found that both males and females learned the contingency degradation task. However, during extinction testing, males respond more to the contingent lever than the non-contingent lever while females do not differ in their responses on the non-contingent and contingent levers. Lower baseline anxiety-like behavior predicted better performance on the contingency degradation test in males, but not females. Next, when we examined performance during extinction testing in females based on their estrous cycle stage on test day, we found that females in the proestrus and estrus stages of the estrous cycle do not differ in their responses on the non-contingent and contingent levers, while females in the metestrus and diestrus stages of the estrous cycle respond more on the contingent lever than the non-contingent lever on the extinction test day, similar to male rats. Our findings indicate that the estrous cycle influences how female rats respond during contingency degradation extinction testing that is dependent on their estrous cycle stage.
Collapse
Affiliation(s)
- Claire M Corbett
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Virtua Health College of Medicine and Life Sciences of Rowan University, Stratford, NJ; Rowan-Virtua School of Osteopathic Medicine, Virtua Health College of Medicine and Life Sciences of Rowan University, Stratford, NJ
| | - Samantha L Bozarth
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Virtua Health College of Medicine and Life Sciences of Rowan University, Stratford, NJ; Rowan-Virtua School of Osteopathic Medicine, Virtua Health College of Medicine and Life Sciences of Rowan University, Stratford, NJ
| | - Elizabeth A West
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Virtua Health College of Medicine and Life Sciences of Rowan University, Stratford, NJ; Rowan-Virtua School of Osteopathic Medicine, Virtua Health College of Medicine and Life Sciences of Rowan University, Stratford, NJ.
| |
Collapse
|
244
|
Dong W, Xu H, Wei W, Ning R, Chang Y. Advances in the study of ferroptosis and its relationship to autoimmune diseases. Int Immunopharmacol 2024; 140:112819. [PMID: 39096870 DOI: 10.1016/j.intimp.2024.112819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/10/2024] [Accepted: 07/25/2024] [Indexed: 08/05/2024]
Abstract
Ferroptosis represents a novel mode of programmed cell death characterized by the intracellular accumulation of iron and lipid peroxidation, culminating in oxidative stress and subsequent cell demise. Mounting evidence demonstrates that ferroptosis contributes significantly to the onset and progression of diverse pathological conditions and diseases, including infections, neurodegenerative disorders, tissue ischemia-reperfusion injury, and immune dysregulation. Recent investigations have underscored the pivotal role of ferroptosis in the pathogenesis of rheumatoid arthritis, ulcerative colitis, systemic lupus erythematosus, and asthma. This review provides a comprehensive overview of the current understanding of the regulatory mechanisms governing ferroptosis, particularly its interplay with iron, lipid, and amino acid metabolism. Furthermore, we explore the implications of ferroptosis in autoimmune diseases and deliberate on its potential as a promising therapeutic target for diverse autoimmune disorders.
Collapse
Affiliation(s)
- Weibo Dong
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Hepeng Xu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Rende Ning
- The Third Affiliated Hospital of Anhui Medical University (The First People's Hospital of Hefei), 390 Huaihe Road, Hefei 230061, Anhui, China.
| | - Yan Chang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China; Laboratory Animal Center, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
245
|
Zhang W, Qin P, Li M, Pan Z, Wu Z, Zhu Y, Liu Y, Li Y, Fang F. NAGK regulates the onset of puberty in female mice. Theriogenology 2024; 231:228-239. [PMID: 39488153 DOI: 10.1016/j.theriogenology.2024.10.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/23/2024] [Accepted: 10/23/2024] [Indexed: 11/04/2024]
Abstract
This study examines the role of N-acetylglucosamine kinase (NAGK) in initiating puberty in female mice. We employed real-time quantitative reverse transcription polymerase chain reaction (RT-qPCR) and immunofluorescence to measure NAGK expression in the hypothalamic-pituitary-ovarian axis across various developmental stages: infant, prepuberty, puberty, and adult. We further investigated the impact of Nagk gene knockdown on puberty in female mice. This included assessing the expression of puberty-related genes both in vivo and in vitro, GT1-7 cells proliferation and apoptosis, concentrations of GnRH and Kisspeptin, puberty onset timing, serum levels of progesterone (P4) and estradiol (E2), and ovarian morphology. Results revealed that Nagk mRNA is present in the hypothalamus, pituitary, and ovaries throughout different developmental stages in female mice. In the hypothalamus, Nagk mRNA levels were comparable during infant and prepuberty, lowest during puberty, and highest in adult. In the pituitary, Nagk mRNA peaked in adult, with no significant variation between infant, prepuberty, and puberty. In the ovaries, Nagk mRNA levels increased during puberty and peaked in adult. NAGK is predominantly located in the arcuate nucleus (ARC), periventricular nucleus (PeN), dorsomedial hypothalamic nucleus (DMH), paraventricular nucleus (PVN), adenohypophysis, and in the ovarian oocytes, interstitium, and granulosa cells across all developmental stages in female mice. Nagk knockdown in GT1-7 cells decreased the transcriptional level of Gnrh, Kiss1, Gpr54, Igf1 and Mapk14 mRNA and cell proliferation but increased the level of β-catenin mRNA and cell apoptosis, while reducing GnRH secretion. Following ICV injection, Nagk gene knockdown mice exhibited delayed the timing of vaginal opening (VO) and reduced hypothalamic levels of Gnrh, Kiss1, Gpr54, Igf1, Mapk14, and β-catenin mRNA. Additionally, serum concentrations of E2 in Nagk gene knockdown mice were significantly lower compared to the control group. These findings indicate that Nagk regulates the expression of Gnrh and Kiss1 mRNA in GT1-7 cells, affects hypothalamus Gnrh mRNA levels and serum E2 concentration, and that its knockdown can delay puberty onset in female mice.
Collapse
Affiliation(s)
- Wei Zhang
- Anhui Provincial Laboratory of Animal Genetic Resources Protection and Breeding, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui, 230036, China; Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui, 230036, China
| | - Ping Qin
- Anhui Provincial Laboratory of Animal Genetic Resources Protection and Breeding, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui, 230036, China; Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui, 230036, China
| | - Mengxian Li
- Anhui Provincial Laboratory of Animal Genetic Resources Protection and Breeding, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui, 230036, China; Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui, 230036, China
| | - Zhihao Pan
- Anhui Provincial Laboratory of Animal Genetic Resources Protection and Breeding, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui, 230036, China; Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui, 230036, China
| | - Zhuoya Wu
- Anhui Provincial Laboratory of Animal Genetic Resources Protection and Breeding, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui, 230036, China; Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui, 230036, China
| | - Yanyun Zhu
- Anhui Provincial Laboratory of Animal Genetic Resources Protection and Breeding, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui, 230036, China; Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui, 230036, China
| | - Ya Liu
- Anhui Provincial Laboratory of Animal Genetic Resources Protection and Breeding, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui, 230036, China; Anhui Provincial Key Laboratory for Local Livestock and Poultry Genetic Resource Conservation and Bio-Breeding, 130 Changjiang West Road, Hefei, Anhui, 230036, China; Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui, 230036, China
| | - Yunsheng Li
- Anhui Provincial Laboratory of Animal Genetic Resources Protection and Breeding, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui, 230036, China; Anhui Provincial Key Laboratory for Local Livestock and Poultry Genetic Resource Conservation and Bio-Breeding, 130 Changjiang West Road, Hefei, Anhui, 230036, China; Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui, 230036, China
| | - Fugui Fang
- Anhui Provincial Laboratory of Animal Genetic Resources Protection and Breeding, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui, 230036, China; Anhui Provincial Key Laboratory for Local Livestock and Poultry Genetic Resource Conservation and Bio-Breeding, 130 Changjiang West Road, Hefei, Anhui, 230036, China; Department of Animal Veterinary Science, College of Animal Science and Technology, Anhui Agricultural University, 130 Changjiang West Road, Hefei, Anhui, 230036, China.
| |
Collapse
|
246
|
Pirník Z, Szadvári I, Borbélyová V, Tomova A. Altered sex differences related to food intake, hedonic preference, and FosB/deltaFosB expression within central neural circuit involved in homeostatic and hedonic food intake regulation in Shank3B mouse model of autism spectrum disorder. Neurochem Int 2024; 181:105895. [PMID: 39461669 DOI: 10.1016/j.neuint.2024.105895] [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: 05/27/2024] [Revised: 10/09/2024] [Accepted: 10/24/2024] [Indexed: 10/29/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder accompanied by narrow interests, difficulties in communication and social interaction, and repetitive behavior. In addition, ASD is frequently associated with eating and feeding problems. Although the symptoms of ASD are more likely to be observed in boys, the prevalence of eating disorders is more common in females. The ingestive behavior is regulated by the integrative system of the brain, which involves both homeostatic and hedonic neural circuits. Sex differences in the physiology of food intake depend on sex hormones regulating the expression of the ASD-associated Shank genes. Shank3 mutation leads to ASD-like traits and Shank3B -/- mice have been established as an animal model to study the neurobiology of ASD. Therefore, the long-lasting neuronal activity in the central neural circuit related to the homeostatic and hedonic regulation of food intake was evaluated in both sexes of Shank3B mice, followed by the evaluation of the food intake and preference. In the Shank3B +/+ genotype, well-preserved relationships in the tonic activity within the homeostatic neural network together with the relationships between ingestion and hedonic preference were observed in males but were reduced in females. These interrelations were partially or completely lost in the mice with the Shank3B -/- genotype. A decreased hedonic preference for the sweet taste but increased total food intake was found in the Shank3B -/- mice. In the Shank3B -/- group, there were altered sex differences related to the amount of tonic cell activity in the hedonic and homeostatic neural networks, together with altered sex differences in sweet and sweet-fat solution intake. Furthermore, the Shank3B -/- females exhibited an increased intake and preference for cheese compared to the Shank3B +/+ ones. The obtained data indicate altered functional crosstalk between the central homeostatic and hedonic neural circuits involved in the regulation of food intake in ASD.
Collapse
Affiliation(s)
- Zdenko Pirník
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia; Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia; Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic.
| | - Ivan Szadvári
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Veronika Borbélyová
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Aleksandra Tomova
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| |
Collapse
|
247
|
Khati WH, Al Mutery AF, Moudilou EN, Exbrayat JM, Hammouche S. WITHDRAWN: Distribution of the Novel RFRP-3/receptors system in the epididymis of the seasonal desert rodent, Gerbillus tarabuli, during sexual activity. Morphologie 2024:S1286-0115(21)00233-2. [PMID: 34774455 DOI: 10.1016/j.morpho.2021.10.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/27/2021] [Accepted: 10/10/2021] [Indexed: 12/06/2022]
Abstract
This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at: https://www.elsevier.com/about/policies/article-withdrawal.
Collapse
Affiliation(s)
- W H Khati
- USTHB, Arid Area Research Laboratory, Biological Sciences Faculty, University of Sciences and Technology of Houari-Boumediene, Algiers, Algeria.
| | - A F Al Mutery
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah, United Arab Emirates; Human Genetics & Stem Cells Research Group, Research Institute of Sciences & Engineering, University of Sharjah, Sharjah, United Arab Emirates; Molecular Genetics Research Laboratory, University of Sharjah, Sharjah, United Arab Emirates
| | - E N Moudilou
- UMRS 449 - General Biology - Reproduction and Comparative Development, UDL; École Pratique des Hautes Études, PSL, Lyon Catholic University, Lyon, France
| | - J-M Exbrayat
- UMRS 449 - General Biology - Reproduction and Comparative Development, UDL; École Pratique des Hautes Études, PSL, Lyon Catholic University, Lyon, France
| | - S Hammouche
- USTHB, Arid Area Research Laboratory, Biological Sciences Faculty, University of Sciences and Technology of Houari-Boumediene, Algiers, Algeria
| |
Collapse
|
248
|
Sokolowski DJ, Hou H, Yuki KE, Roy A, Chan C, Choi W, Faykoo-Martinez M, Hudson M, Corre C, Uusküla-Reimand L, Goldenberg A, Palmert MR, Wilson MD. Age, sex, and cell type-resolved hypothalamic gene expression across the pubertal transition in mice. Biol Sex Differ 2024; 15:83. [PMID: 39449090 PMCID: PMC11515584 DOI: 10.1186/s13293-024-00661-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 10/07/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND The hypothalamus plays a central role in regulating puberty. However, our knowledge of the postnatal gene regulatory networks that control the pubertal transition in males and females is incomplete. Here, we investigate the age-, sex- and cell-type-specific gene regulation in the hypothalamus across the pubertal transition. METHODS We used RNA-seq to profile hypothalamic gene expression in male and female mice at five time points spanning the onset of puberty (postnatal days (PD) 12, 22, 27, 32, and 37). By combining this data with hypothalamic single nuclei RNA-seq data from pre- and postpubertal mice, we assigned gene expression changes to their most likely cell types of origin. In our colony, pubertal onset occurs earlier in male mice, allowing us to focus on genes whose expression is dynamic across ages and offset between sexes, and to explore the bases of sex effects. RESULTS Our age-by-sex pattern of expression enriched for biological pathways involved hormone production, neuronal activation, and glial maturation. Additionally, we inferred a robust expansion of oligodendrocytes precursor cells into mature oligodendrocytes spanning the prepubertal (PD12) to peri-pubertal (PD27) timepoints. Using spatial transcriptomic data from postpubertal mice, we observed the lateral hypothalamic area and zona incerta were the most oligodendrocyte-rich regions and that these cells expressed genes known to be involved in pubertal regulation. CONCLUSION Together, by incorporating multiple biological timepoints and using sex as a variable, we identified gene and cell-type changes that may participate in orchestrating the pubertal transition and provided a resource for future studies of postnatal hypothalamic gene regulation.
Collapse
Affiliation(s)
- Dustin J Sokolowski
- Genetics and Genome Biology, SickKids Research Institute, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Huayun Hou
- Genetics and Genome Biology, SickKids Research Institute, Toronto, ON, Canada
| | - Kyoko E Yuki
- Genetics and Genome Biology, SickKids Research Institute, Toronto, ON, Canada
| | - Anna Roy
- Genetics and Genome Biology, SickKids Research Institute, Toronto, ON, Canada
| | - Cadia Chan
- Genetics and Genome Biology, SickKids Research Institute, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Donnelly Centre for Cellular & Biomolecular Research, Toronto, ON, Canada
| | - Wendy Choi
- Genetics and Genome Biology, SickKids Research Institute, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Mariela Faykoo-Martinez
- Genetics and Genome Biology, SickKids Research Institute, Toronto, ON, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Matt Hudson
- Genetics and Genome Biology, SickKids Research Institute, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Christina Corre
- Genetics and Genome Biology, SickKids Research Institute, Toronto, ON, Canada
| | | | - Anna Goldenberg
- Genetics and Genome Biology, SickKids Research Institute, Toronto, ON, Canada
- Department of Computer Science, University of Toronto, Toronto, ON, Canada
- Vector Institute, Toronto, ON, Canada
- CIFAR, Toronto, ON, Canada
| | - Mark R Palmert
- Genetics and Genome Biology, SickKids Research Institute, Toronto, ON, Canada
- Division of Endocrinology, The Hospital for Sick Children, Toronto, ON, Canada
- Departments of Pediatrics and Physiology, University of Toronto, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Michael D Wilson
- Genetics and Genome Biology, SickKids Research Institute, Toronto, ON, Canada.
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
249
|
Hu W, Jiang L, Wang Q, Hu Q, Zhong T, Wu J, Chen X, Liu T. Chronic unpredictable stress during adolescence exerts sex-specific effects on depressive-like behavior and neural activation triggered by tail suspension test. Behav Brain Res 2024; 477:115314. [PMID: 39461371 DOI: 10.1016/j.bbr.2024.115314] [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: 05/01/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 10/29/2024]
Abstract
During adolescence, acute stress can modify neuronal excitability in various brain regions, leading to negative behavioral outcomes. However, the impact of chronic stress during adolescence on neuronal responses to acute stimuli remains unclear. To address this, we subjected adolescent mice to 12 days of chronic unpredictable stress (CUS). Anxiety and depressive behaviors were evaluated, along with changes in c-Fos expression, which is one of the most widely used markers of neuronal activation. By comparing c-Fos immunoreactivity between the CUS and control groups both before and after the tail suspension test (TST), we found that adolescent CUS induced depressive behaviors in male mice, but not in female mice. Adolescent CUS primarily affected the excitability of neurons in the infralimbic cortex (IL), the dorsomedial and dorsolateral area of the bed nucleus of the stria terminalis (BNST), and the ventral hippocampus CA3. TST exerted a significant main effect on the density of c-Fos+ neurons in the prelimbic cortex (PL), infralimbic cortex (IL), cingulate areas 1 and 2 (Cg1, Cg2), the lateral septum (LS), BNST, and lateral habenular (LHb). Furthermore, the excitability of neurons in the paraventricular thalamic nucleus (PVT) was impacted by sex. These data suggest that adolescent CUS elicits region- and sex-specific modifications in TST-induced c-Fos expression, establishing a theoretical basis for understanding the pathophysiological alterations in mood disorders following adolescent stress.
Collapse
Affiliation(s)
- Wenjing Hu
- Department of Pediatrics, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Lifang Jiang
- Department of Pediatrics, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Qiyuan Wang
- Department of Pediatrics, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Qijiang Hu
- Department of Pediatrics, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Tianfeng Zhong
- Department of Pediatrics, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Jian Wu
- Department of Pediatrics, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Xiao Chen
- Department of Pediatrics, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Tao Liu
- Department of Pediatrics, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China.
| |
Collapse
|
250
|
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] [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
|