1
|
Duan J, Sun J, Ma X, Du P, Dong P, Xue J, Lu Y, Jiang T. Association of escitalopram-induced shifts in gut microbiota and sphingolipid metabolism with depression-like behavior in wistar-kyoto rats. Transl Psychiatry 2025; 15:54. [PMID: 39962083 PMCID: PMC11833111 DOI: 10.1038/s41398-025-03277-8] [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: 07/06/2024] [Revised: 01/14/2025] [Accepted: 02/07/2025] [Indexed: 02/20/2025] Open
Abstract
The microbiota-gut-brain axis plays a pivotal role in neuropsychiatric disorders, particularly in depression. Escitalopram (ESC) is a first-line antidepressant, however, its regulatory mechanisms on the microbiota-gut-brain axis in the treatment of depression remain unclear. The antidepressant effects of ESC were evaluated using the forced swim test in Wistar-Kyoto (WKY) rats, while damage in the gut and brain regions was assessed through H&E staining and immunohistochemistry. The therapeutic mechanisms in WKY rats with depression-like behavior were investigated through 16S rRNA sequencing of the gut microbiota, serum untargeted metabolomics, and hippocampal proteomics. Results indicated that ESC intervention improved depressive-like behaviors, as evidenced by increased swimming times in WKY rats, and also restored intestinal permeability and brain tissue integrity. Significant changes in the gut microbiota composition, particularly an increase in Bacteroides barnesiae, as well as increases in serum sphingolipid metabolites (Sphinganine 1-phosphate, Sphingosine, and Sphingosine-1-phosphate) and hippocampal proteins (Sptlc1, Enpp5, Enpp2), were strongly correlated. These robust correlations suggest that ESC may exert its antidepressant effects by modulating sphingolipid metabolism through the influence of gut microbiota. Accordingly, this research elucidates novel mechanisms underlying the antidepressant efficacy of ESC and highlights the pivotal importance of the microbiota-gut-brain axis in mediating these effects.
Collapse
Affiliation(s)
- Jiajia Duan
- Department of Clinical Laboratory, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Jiaxing Sun
- Department of Clinical Laboratory, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Xiao Ma
- Department of Clinical Laboratory, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Peipei Du
- Department of Clinical Laboratory, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Pengfei Dong
- Department of Clinical Laboratory, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Juan Xue
- Department of Clinical Laboratory, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Yanli Lu
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, China
| | - Tao Jiang
- Department of Clinical Laboratory, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China.
| |
Collapse
|
2
|
Tian X, Russo SJ, Li L. Behavioral Animal Models and Neural-Circuit Framework of Depressive Disorder. Neurosci Bull 2025; 41:272-288. [PMID: 39120643 PMCID: PMC11794861 DOI: 10.1007/s12264-024-01270-7] [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: 02/22/2024] [Accepted: 04/26/2024] [Indexed: 08/10/2024] Open
Abstract
Depressive disorder is a chronic, recurring, and potentially life-endangering neuropsychiatric disease. According to a report by the World Health Organization, the global population suffering from depression is experiencing a significant annual increase. Despite its prevalence and considerable impact on people, little is known about its pathogenesis. One major reason is the scarcity of reliable animal models due to the absence of consensus on the pathology and etiology of depression. Furthermore, the neural circuit mechanism of depression induced by various factors is particularly complex. Considering the variability in depressive behavior patterns and neurobiological mechanisms among different animal models of depression, a comparison between the neural circuits of depression induced by various factors is essential for its treatment. In this review, we mainly summarize the most widely used behavioral animal models and neural circuits under different triggers of depression, aiming to provide a theoretical basis for depression prevention.
Collapse
Affiliation(s)
- Xiangyun Tian
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Scott J Russo
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| | - Long Li
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- University of the Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
3
|
Kronman H, Singh A, Azam S, Guzman AS, Zelli D, Lau T, Dobbin J, Bigio B, Nasca C. Multidimensional Effects of Stress on Neuronal Exosome Levels and Simultaneous Transcriptomic Profiles. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2025; 5:100401. [PMID: 39720402 PMCID: PMC11667124 DOI: 10.1016/j.bpsgos.2024.100401] [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: 02/13/2024] [Revised: 09/04/2024] [Accepted: 09/08/2024] [Indexed: 12/26/2024] Open
Abstract
Background An excess of exosomes, nanovesicles released from all cells and key regulators of brain plasticity, is an emerging therapeutic target for stress-related mental illnesses. The effects of chronic stress on exosome levels are unknown; even less is known about molecular drivers of exosome levels in the stress response. Methods We used our state-of-the-art protocol with 2 complementary strategies to isolate neuronal exosomes from plasma, ventral dentate gyrus, basolateral amygdala, and olfactory bulbs of male mice to determine the effects of chronic restraint stress (CRS) on exosome levels. Next, we used RNA sequencing and bioinformatic analyses to identify molecular drivers of exosome levels. Results We found that CRS leads to an increase in the levels of neuronal exosomes but not total (i.e., not neuronally enriched) exosome levels assayed in plasma and the ventral dentate gyrus, whereas CRS leads to a decrease in neuronal exosome levels but not total exosome levels in the basolateral amygdala. There was a further specificity of effects as shown by a lack of changes in the levels of neuronal exosomes assayed in the olfactory bulbs. In pursuit of advancing translational applications, we showed that acetyl-L-carnitine administration restores the CRS-induced increase in neuronal exosome levels assayed in plasma (the most accessible specimen). Furthermore, the CRS-induced changes in neuronal exosome levels in the ventral dentate gyrus and basolateral amygdala mirrored the opposite pattern of CRS-induced transcriptional changes in these key brain areas, with β-estradiol signaling as a potential upstream driver of neuronal exosome levels. Conclusions This study provides a foundation for future studies of new forms of local and distant communication in stress neurobiology by demonstrating specific relationships between neuronal exosome levels assayed in plasma and the brain and providing new candidate targets for the normalization of exosome levels.
Collapse
Affiliation(s)
- Hope Kronman
- Department of Psychiatry, New York University Grossman School of Medicine, New York, New York
- Nathan Kline Institute for Psychiatric Research, Orangeburg, New York
| | - Amarjyot Singh
- Nathan Kline Institute for Psychiatric Research, Orangeburg, New York
| | - Shofiul Azam
- Department of Psychiatry, New York University Grossman School of Medicine, New York, New York
- Nathan Kline Institute for Psychiatric Research, Orangeburg, New York
| | - Andrea S. Guzman
- Department of Psychiatry, New York University Grossman School of Medicine, New York, New York
- Nathan Kline Institute for Psychiatric Research, Orangeburg, New York
| | - Danielle Zelli
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, the Rockefeller University, New York, New York
| | - Timothy Lau
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, the Rockefeller University, New York, New York
| | - Josh Dobbin
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, the Rockefeller University, New York, New York
| | - Benedetta Bigio
- Department of Psychiatry, New York University Grossman School of Medicine, New York, New York
- Nathan Kline Institute for Psychiatric Research, Orangeburg, New York
| | - Carla Nasca
- Department of Psychiatry, New York University Grossman School of Medicine, New York, New York
- Nathan Kline Institute for Psychiatric Research, Orangeburg, New York
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, the Rockefeller University, New York, New York
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, New York
- Neuroscience Institute, New York University Grossman School of Medicine, New York, New York
| |
Collapse
|
4
|
Schwartz A, Macalli M, Navarro MC, Jean FAM, Crivello F, Galera C, Tzourio C. Adverse childhood experiences and left hippocampal volumetric reductions: A structural magnetic resonance imaging study. J Psychiatr Res 2024; 180:183-189. [PMID: 39427447 DOI: 10.1016/j.jpsychires.2024.09.039] [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: 03/21/2024] [Revised: 09/10/2024] [Accepted: 09/29/2024] [Indexed: 10/22/2024]
Abstract
BACKGROUND Adverse childhood experiences (ACEs) have been associated with volume alterations of stress-related brain structures among aging and clinical populations, however, existing studies have predominantly assessed only one type of ACE, with small sample sizes, and it is less clear if these associations exist among a general population of young adults. OBJECTIVE The aims were to describe structural hippocampal volumetric differences by ACEs exposure and investigate the association between ACEs exposure and left and right hippocampal volume in a student sample of young adults. METHODS 959 young adult students (18-24 years old) completed an online questionnaire on ACEs, mental health conditions, and sociodemographic characteristics. Magnetic resonance imaging (MRI) was used to measure left and right hippocampal volume (mm3). We used linear regression to explore the differences of hippocampal volumes in university students with and without ACEs. RESULTS Two thirds of students (65.9%) reported ACEs exposure. As ACEs exposure increased there were significant volumetric reductions in left (p < 0.0001) and right hippocampal volume (p = 0.001) and left (p = 0.0023) and right (p = 0.0013) amygdala volume. After adjusting for intracranial brain volume, sex, age, and depression diagnosis there was a negative association between ACEs exposure and left (β = -22.6, CI = -44.5, -0.7, p = 0.0412) but not right hippocampal volume (β = -18.3, CI = -39.2, 2.6, p = 0.0792). After adjusting for intracranial volume there were no associations between ACEs exposure and left (β = -9.2, CI = -26.2, 7.9 p = 0.2926) or right (β = -5.6, CI = -19.9,8.8 p = 0.4466) amygdala volume. CONCLUSIONS Hippocampal volume varied by ACEs exposure in young adult students. ACEs appear to contribute to neuroanatomic differences in young adults from the general population.
Collapse
Affiliation(s)
- Ashlyn Schwartz
- Trinity College, Department of Public Health & Primary Care, D24 DH74, Dublin, Ireland.
| | - Mélissa Macalli
- Inserm, Bordeaux Population Health Research Center, U1219, CHU Bordeaux, F-33000, Bordeaux, France.
| | - Marie C Navarro
- Inserm, Bordeaux Population Health Research Center, U1219, CHU Bordeaux, F-33000, Bordeaux, France.
| | - François A M Jean
- Dr Jean Eric Techer Hospital, Department of Psychiatry, Calais, France.
| | - Fabrice Crivello
- Univ. Bordeaux, CEA, CNRS, IMN UMR 5293, Bordeaux, F-33000, France.
| | | | - Christophe Tzourio
- Inserm, Bordeaux Population Health Research Center, U1219, CHU Bordeaux, F-33000, Bordeaux, France.
| |
Collapse
|
5
|
Kim B, Weibel M, McDaniel J. Loneliness Gets Under the Skin: A Scoping Review Exploring the Link Between Loneliness and Biological Measures of Inflammation. West J Nurs Res 2024; 46:989-1001. [PMID: 39451131 DOI: 10.1177/01939459241292037] [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: 10/26/2024]
Abstract
BACKGROUND Research suggests that systemic inflammation may link loneliness to adverse health outcomes, yet there is a gap in comprehensively reviewing recent evidence on the relationship between loneliness and biological measures of inflammation in adults. This scoping review synthesizes current research to address the question: Is there a definitive link between loneliness and biological markers of inflammation in adults? METHODS Following the methods outlined by Arksey and O'Malley, we developed a protocol, defined our research question, and systematically searched PubMed, CINAHL, Embase, and Scopus for English-language studies conducted from 2018 to 2023 exploring the relationship between loneliness and biomarkers of inflammation in adults. RESULTS Twelve studies meeting the inclusion criteria displayed heterogeneity in terms of sample characteristics, loneliness scales, and inflammatory biomarkers. The UCLA Loneliness Scale, in various forms, emerged as the predominant tool for measuring loneliness, while C-reactive protein and interleukin-6 were the most frequently evaluated inflammatory biomarkers. Notably, all 12 studies reported an association between loneliness and at least 1 biological marker of inflammation. CONCLUSION Research consistently associates loneliness with poor health outcomes in aging adults, but the underlying mechanisms remain unclear. This scoping review suggests that inflammation may serve as a pathway linking loneliness to adverse health outcomes. However, the variability across studies highlights the need for standardized measurement methods and a consideration of both the duration and extent of loneliness. Enhancing our understanding of how loneliness affects systemic inflammation may help clarify why loneliness is associated with negative health outcomes.
Collapse
Affiliation(s)
- Bohyun Kim
- College of Nursing, The Ohio State University, Columbus, OH, USA
| | - Maria Weibel
- College of Nursing, The Ohio State University, Columbus, OH, USA
| | - Jodi McDaniel
- College of Nursing, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
6
|
Díaz-Carballo D, Safoor A, Saka S, Noa-Bolaño A, D'Souza F, Klein J, Acikelli AH, Malak S, Rahner U, Turki AT, Höppner A, Kamitz A, Song W, Chen YG, Kamada L, Tannapfel A, Brinkmann S, Ochsenfarth C, Strumberg D. The neuroepithelial origin of ovarian carcinomas explained through an epithelial-mesenchymal-ectodermal transition enhanced by cisplatin. Sci Rep 2024; 14:29286. [PMID: 39592661 PMCID: PMC11599565 DOI: 10.1038/s41598-024-76984-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 10/18/2024] [Indexed: 11/28/2024] Open
Abstract
Acquired resistance to platinum-derived cytostatics poses major challenges in ovarian carcinoma therapy. In this work, we show a shift in the epithelial-mesenchymal transition (EMT) process towards an "ectodermal" conversion of ovarian carcinoma cells in response to cisplatin treatment, a progression we have termed epithelial-mesenchymal-ectodermal transition (EMET). EMET appears to occur via the classical EMT as judged by a) the downregulation of several epithelial markers and b) upregulation of Vimentin, accompanied by various embryonal transcription factors and, importantly, a plethora of neuronal markers, consistent with ectodermal differentiation. Moreover, we isolated cells from ovarian carcinoma cultures exhibiting a dual neural/stemness signature and multidrug resistance (MDR) phenotype. We also found that the epithelial cells differentiate from these neural/stem populations, indicating that the cell of origin in this tumor must in fact be a neural cell type with stemness features. Notably, some transcription factors like PAX6 and PAX9 were not localized in the nucleoplasm of these cells, hinting at altered nuclear permeability. In addition, the neuronal morphology was rapidly established when commercially available and primary ovarian carcinoma cells were cultured in the form of organoids. Importantly, we also identified a cell type in regular ovarian tissues, which possess similar neural/stemness features as observed in 2D or 3D cultures. The signature of this cell type is amplified in ovarian carcinoma tumors, suggesting a neuroepithelial origin of this tumor type. In conclusion, we propose that ovarian carcinomas harbor a small population of cells with an intrinsic neuronal/stemness/MDR phenotype, serving as the cradle from which ovarian carcinoma evolves.
Collapse
Affiliation(s)
- David Díaz-Carballo
- Institute of Molecular Oncology and Experimental Therapeutics, Division of Hematology and Oncology, Ruhr University Bochum Medical School, Marien Hospital Herne, Düngelstr. 33, 44623, Herne, Germany.
| | - Ayesha Safoor
- Institute of Molecular Oncology and Experimental Therapeutics, Division of Hematology and Oncology, Ruhr University Bochum Medical School, Marien Hospital Herne, Düngelstr. 33, 44623, Herne, Germany
| | - Sahitya Saka
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, USA
| | - Adrien Noa-Bolaño
- Institute of Molecular Oncology and Experimental Therapeutics, Division of Hematology and Oncology, Ruhr University Bochum Medical School, Marien Hospital Herne, Düngelstr. 33, 44623, Herne, Germany
| | - Flevy D'Souza
- Institute of Molecular Oncology and Experimental Therapeutics, Division of Hematology and Oncology, Ruhr University Bochum Medical School, Marien Hospital Herne, Düngelstr. 33, 44623, Herne, Germany
| | - Jacqueline Klein
- Institute of Molecular Oncology and Experimental Therapeutics, Division of Hematology and Oncology, Ruhr University Bochum Medical School, Marien Hospital Herne, Düngelstr. 33, 44623, Herne, Germany
| | - Ali H Acikelli
- Institute of Molecular Oncology and Experimental Therapeutics, Division of Hematology and Oncology, Ruhr University Bochum Medical School, Marien Hospital Herne, Düngelstr. 33, 44623, Herne, Germany
| | - Sascha Malak
- Institute of Molecular Oncology and Experimental Therapeutics, Division of Hematology and Oncology, Ruhr University Bochum Medical School, Marien Hospital Herne, Düngelstr. 33, 44623, Herne, Germany
| | - Udo Rahner
- Institute of Molecular Oncology and Experimental Therapeutics, Division of Hematology and Oncology, Ruhr University Bochum Medical School, Marien Hospital Herne, Düngelstr. 33, 44623, Herne, Germany
| | - Amin T Turki
- Institute of Molecular Oncology and Experimental Therapeutics, Division of Hematology and Oncology, Ruhr University Bochum Medical School, Marien Hospital Herne, Düngelstr. 33, 44623, Herne, Germany
| | - Anne Höppner
- Institute of Molecular Oncology and Experimental Therapeutics, Division of Hematology and Oncology, Ruhr University Bochum Medical School, Marien Hospital Herne, Düngelstr. 33, 44623, Herne, Germany
| | - Annabelle Kamitz
- Institute of Molecular Oncology and Experimental Therapeutics, Division of Hematology and Oncology, Ruhr University Bochum Medical School, Marien Hospital Herne, Düngelstr. 33, 44623, Herne, Germany
| | - Wanlu Song
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Ye-Guang Chen
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Lalitha Kamada
- Clinic of Pediatric Oncology, Hematology and Immunology, Düsseldorf University Hospital , 40225, Düsseldorf, Germany
| | - Andrea Tannapfel
- Institute of Pathology, Ruhr University Bochum, Medical School, Bürkle-de-La-Camp-Platz 1, 44789, Bochum, Germany
| | - Sebastian Brinkmann
- Department of General and Visceral Surgery, St. Josef-Hospital, Ruhr University Bochum, Medical School, Bürkle-de-La-Camp-Platz 1, 44789, Bochum, Germany
| | - Crista Ochsenfarth
- Department of Anesthesia, Intensive Care, Pain and Palliative Medicine, Ruhr-University Bochum Medical School, Marien Hospital Herne, 44625, Herne, Germany
| | - Dirk Strumberg
- Institute of Molecular Oncology and Experimental Therapeutics, Division of Hematology and Oncology, Ruhr University Bochum Medical School, Marien Hospital Herne, Düngelstr. 33, 44623, Herne, Germany
| |
Collapse
|
7
|
Shansky RM. Behavioral neuroscience's inevitable SABV growing pains. Trends Neurosci 2024; 47:669-676. [PMID: 39034262 DOI: 10.1016/j.tins.2024.06.007] [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: 05/06/2024] [Revised: 06/17/2024] [Accepted: 06/26/2024] [Indexed: 07/23/2024]
Abstract
The field of rodent behavioral neuroscience is undergoing two major sea changes: an ever-growing technological revolution, and worldwide calls to consider sex as a biological variable (SABV) in experimental design. Both have enormous potential to improve the precision and rigor with which the brain can be studied, but the convergence of these shifts in scientific practice has exposed critical limitations in classic and widely used behavioral paradigms. While our tools have advanced, our behavioral metrics - mostly developed in males and often allowing for only binary outcomes - have not. This opinion article explores how this disconnect has presented challenges for the accurate depiction and interpretation of sex differences in brain function, arguing for the expansion of current behavioral constructs to better account for behavioral diversity.
Collapse
|
8
|
Ferro MD, Proctor CM, Gonzalez A, Jayabal S, Zhao E, Gagnon M, Slézia A, Pas J, Dijk G, Donahue MJ, Williamson A, Raymond J, Malliaras GG, Giocomo L, Melosh NA. NeuroRoots, a bio-inspired, seamless brain machine interface for long-term recording in delicate brain regions. AIP ADVANCES 2024; 14:085109. [PMID: 39130131 PMCID: PMC11309783 DOI: 10.1063/5.0216979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 07/09/2024] [Indexed: 08/13/2024]
Abstract
Scalable electronic brain implants with long-term stability and low biological perturbation are crucial technologies for high-quality brain-machine interfaces that can seamlessly access delicate and hard-to-reach regions of the brain. Here, we created "NeuroRoots," a biomimetic multi-channel implant with similar dimensions (7 μm wide and 1.5 μm thick), mechanical compliance, and spatial distribution as axons in the brain. Unlike planar shank implants, these devices consist of a number of individual electrode "roots," each tendril independent from the other. A simple microscale delivery approach based on commercially available apparatus minimally perturbs existing neural architectures during surgery. NeuroRoots enables high density single unit recording from the cerebellum in vitro and in vivo. NeuroRoots also reliably recorded action potentials in various brain regions for at least 7 weeks during behavioral experiments in freely-moving rats, without adjustment of electrode position. This minimally invasive axon-like implant design is an important step toward improving the integration and stability of brain-machine interfacing.
Collapse
Affiliation(s)
- Marc D. Ferro
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, USA
| | - Christopher M. Proctor
- Electrical Engineering Division, Department of Engineering, University of Cambridge, Cambridge CB3 0FA, United Kingdom
| | - Alexander Gonzalez
- Department of Neurobiology, Stanford University School of Medicine, Stanford, California 94304, USA
| | - Sriram Jayabal
- Department of Neurobiology, Stanford University School of Medicine, Stanford, California 94304, USA
| | - Eric Zhao
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, USA
| | - Maxwell Gagnon
- Department of Neurobiology, Stanford University School of Medicine, Stanford, California 94304, USA
| | - Andrea Slézia
- Multimodal Neurotechnology Group, Institute of Cognitive Neuroscience and Psychology, HUN-REN Research Centre for Natural Sciences, Hungarian Research Network, 1117 Budapest, Magyar tudósok körútja 2., Hungary
| | - Jolien Pas
- Department of Bioelectronics, Ecole Nationale Supérieure des Mines, CMP-EMSE, 13541 Gardanne, France
| | - Gerwin Dijk
- Department of Bioelectronics, Ecole Nationale Supérieure des Mines, CMP-EMSE, 13541 Gardanne, France
| | - Mary J. Donahue
- Laboratory of Organic Electronics, Department of Science and Technology, Linköping University, Norrköping, 60221, Sweden
| | | | - Jennifer Raymond
- Department of Neurobiology, Stanford University School of Medicine, Stanford, California 94304, USA
| | - George G. Malliaras
- Electrical Engineering Division, Department of Engineering, University of Cambridge, Cambridge CB3 0FA, United Kingdom
| | - Lisa Giocomo
- Department of Neurobiology, Stanford University School of Medicine, Stanford, California 94304, USA
| | - Nicholas A. Melosh
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, USA
| |
Collapse
|
9
|
Copenhaver AE, LeGates TA. Sex-Specific Mechanisms Underlie Long-Term Potentiation at Hippocampus→Medium Spiny Neuron Synapses in the Medial Shell of the Nucleus Accumbens. J Neurosci 2024; 44:e0100242024. [PMID: 38806250 PMCID: PMC11223474 DOI: 10.1523/jneurosci.0100-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 05/16/2024] [Accepted: 05/21/2024] [Indexed: 05/30/2024] Open
Abstract
Sex differences have complicated our understanding of the neurobiological basis of many behaviors that are key for survival. As such, continued elucidation of the similarities and differences between sexes is necessary to gain insight into brain function and vulnerability. The connection between the hippocampus (Hipp) and nucleus accumbens (NAc) is a crucial site where modulation of neuronal activity mediates reward-related behavior. Our previous work demonstrated that long-term potentiation (LTP) of Hipp→NAc synapses is rewarding, and mice can establish learned associations between LTP of these synapses and the contextual environment in which LTP occurred. Here, we investigated sex differences in the mechanisms underlying Hipp→NAc LTP using whole-cell electrophysiology and pharmacology. We observed similarities in basal synaptic strength between males and females and found that LTP occurs postsynaptically with similar magnitudes in both sexes. However, key sex differences emerged as LTP in males required NMDA receptors (NMDAR), whereas LTP in females utilized an NMDAR-independent mechanism involving L-type voltage-gated Ca2+ channels (VGCCs) and estrogen receptor α (ERα). We also uncovered sex-similar features as LTP in both sexes depended on CaMKII activity and occurred independently of dopamine-1 receptor (D1R) activation. Our results have elucidated sex-specific molecular mechanisms for LTP in an integral pathway that mediates reward-related behaviors, emphasizing the importance of considering sex as a variable in mechanistic studies. Continued characterization of sex-specific mechanisms underlying plasticity will offer novel insight into the neurophysiological basis of behavior, with significant implications for understanding how diverse processes mediate behavior and contribute to vulnerability to developing psychiatric disorders.
Collapse
Affiliation(s)
- Ashley E Copenhaver
- Department of Biological Sciences, University of Maryland, Baltimore County (UMBC), Baltimore, Maryland 21250
| | - Tara A LeGates
- Department of Biological Sciences, University of Maryland, Baltimore County (UMBC), Baltimore, Maryland 21250
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, Maryland 21201
| |
Collapse
|
10
|
Schwandt ML, Cullins E, Ramchandani VA. The role of resilience in the relationship between stress and alcohol. Neurobiol Stress 2024; 31:100644. [PMID: 38827175 PMCID: PMC11140813 DOI: 10.1016/j.ynstr.2024.100644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 04/12/2024] [Accepted: 05/15/2024] [Indexed: 06/04/2024] Open
Abstract
Stress plays a well-documented role in alcohol consumption and the risk for developing alcohol use disorder. The concept of resilience - coping with and successfully adapting to stressful life experiences - has received increasing attention in the field of addiction research in recent decades, and there has been an accumulation of evidence for resilience as a protective factor against problematic alcohol consumption, risk for alcohol use disorder, disorder severity, and relapse. The conceptual and methodological approaches used in the generation of this evidence vary considerably across investigations, however. In light of this, we carried out this review in order to provide a more thorough understanding of the meaning and scope of resilience, what factors contribute to resilience, how it is measured, and how it relates to alcohol-associated phenotypes. Implications for treatment through the use of resilience-building interventions are likewise discussed, as well as implications for future research on the role of resilience in the etiology and clinical outcomes of alcohol use disorder.
Collapse
Affiliation(s)
- Melanie L. Schwandt
- Office of the Clinical Director, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - Eva Cullins
- Human Psychopharmacology Laboratory, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | - Vijay A. Ramchandani
- Human Psychopharmacology Laboratory, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| |
Collapse
|
11
|
Laine MA, Greiner EM, Shansky RM. Sex differences in the rodent medial prefrontal cortex - What Do and Don't we know? Neuropharmacology 2024; 248:109867. [PMID: 38387553 DOI: 10.1016/j.neuropharm.2024.109867] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/22/2024] [Accepted: 02/08/2024] [Indexed: 02/24/2024]
Abstract
The prefrontal cortex, particularly its medial subregions (mPFC), mediates critical functions such as executive control, behavioral inhibition, and memory formation, with relevance for everyday functioning and psychopathology. Despite broad characterization of the mPFC in multiple model organisms, the extent to which mPFC structure and function vary according to an individual's sex is unclear - a knowledge gap that can be attributed to a historical bias for male subjects in neuroscience research. Recent efforts to consider sex as a biological variable in basic science highlight the great need to close this gap. Here we review the knowns and unknowns about how rodents categorized as male or female compare in mPFC neuroanatomy, pharmacology, as well as in aversive, appetitive, and goal- or habit-directed behaviors that recruit the mPFC. We propose that long-standing dogmatic concepts of mPFC structure and function may not remain supported when we move beyond male-only studies, and that empirical challenges to these dogmas are warranted. Additionally, we note some common pitfalls in this work. Most preclinical studies operationalize sex as a binary categorization, and while this approach has furthered the inclusion of non-male rodents it is not as such generalizable to what we know of sex as a multidimensional, dynamic variable. Exploration of sex variability may uncover both sex differences and sex similarities, but care must be taken in their interpretation. Including females in preclinical research needs to go beyond the investigation of sex differences, improving our knowledge of how this brain region and its subregions mediate behavior and health. This article is part of the Special Issue on "PFC circuit function in psychiatric disease and relevant models".
Collapse
Affiliation(s)
- M A Laine
- Department of Psychology, Northeastern University, Boston, MA, USA
| | - E M Greiner
- Department of Psychology, Northeastern University, Boston, MA, USA.
| | - R M Shansky
- Department of Psychology, Northeastern University, Boston, MA, USA
| |
Collapse
|
12
|
Schneider JM, Behboudi MH, Maguire MJ. The Necessity of Taking Culture and Context into Account When Studying the Relationship between Socioeconomic Status and Brain Development. Brain Sci 2024; 14:392. [PMID: 38672041 PMCID: PMC11048655 DOI: 10.3390/brainsci14040392] [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: 03/25/2024] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Decades of research has revealed a relationship between childhood socioeconomic status (SES) and brain development at the structural and functional levels. Of particular note is the distinction between income and maternal education, two highly correlated factors which seem to influence brain development through distinct pathways. Specifically, while a families' income-to-needs ratio is linked with physiological stress and household chaos, caregiver education influences the day-to-day language environment a child is exposed to. Variability in either one of these environmental experiences is related to subsequent brain development. While this work has the potential to inform public policies in a way that benefits children, it can also oversimplify complex factors, unjustly blame low-SES parents, and perpetuate a harmful deficit perspective. To counteract these shortcomings, researchers must consider sociodemographic differences in the broader cultural context that underlie SES-based differences in brain development. This review aims to address these issues by (a) identifying how sociodemographic mechanisms associated with SES influence the day-to-day experiences of children, in turn, impacting brain development, while (b) considering the broader cultural contexts that may differentially impact this relationship.
Collapse
Affiliation(s)
- Julie M. Schneider
- Department of Communication Sciences and Disorders, Louisiana State University, 72 Hatcher Hall, Field House Drive, Baton Rouge, LA 70803, USA;
| | - Mohammad Hossein Behboudi
- Callier Center for Communication Disorders, The University of Texas at Dallas, 1966 Inwood Road, Dallas, TX 75235, USA;
| | - Mandy J. Maguire
- Callier Center for Communication Disorders, The University of Texas at Dallas, 1966 Inwood Road, Dallas, TX 75235, USA;
- Center for Children and Families, The University of Texas at Dallas, 800 W Campbell Road, Dallas, TX 75080, USA
| |
Collapse
|
13
|
Bo Y, Yu Q, Gao W. Progress of depression mechanism based on Omics method. J Pharm Biomed Anal 2024; 240:115884. [PMID: 38183729 DOI: 10.1016/j.jpba.2023.115884] [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: 06/10/2023] [Revised: 11/24/2023] [Accepted: 11/26/2023] [Indexed: 01/08/2024]
Abstract
Depression is a very common disabling mental disorder, which is typically characterized by high rates of disability and mortality. Although research into the various mechanisms of depression was still underway, its physiopathology remains uncertain. The rapid developments in new technologies and the combined use of a variety of techniques will help to understand the pathogenesis of depression and explore effective treatment methods. In this review, we focus on the combination of proteomic and metabolomic approaches to analyze metabolites and proteins in animal models of depression induced by different modeling approaches, with the aim of broadening the understanding of the physiopathological mechanisms of depression using complementary "omics" strategy.
Collapse
Affiliation(s)
- Yaping Bo
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, PR China
| | - Qing Yu
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, PR China
| | - Wenyuan Gao
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, PR China.
| |
Collapse
|
14
|
Guidolin D, Tortorella C, De Caro R, Agnati LF. A Self-Similarity Logic May Shape the Organization of the Nervous System. ADVANCES IN NEUROBIOLOGY 2024; 36:203-225. [PMID: 38468034 DOI: 10.1007/978-3-031-47606-8_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
From the morphological point of view, the nervous system exhibits a fractal, self-similar geometry at various levels of observations, from single cells up to cell networks. From the functional point of view, it is characterized by a hierarchical organization in which self-similar structures (networks) of different miniaturizations are nested within each other. In particular, neuronal networks, interconnected to form neuronal systems, are formed by neurons, which operate thanks to their molecular networks, mainly having proteins as components that via protein-protein interactions can be assembled in multimeric complexes working as micro-devices. On this basis, the term "self-similarity logic" was introduced to describe a nested organization where, at the various levels, almost the same rules (logic) to perform operations are used. Self-similarity and self-similarity logic both appear to be intimately linked to the biophysical evidence for the nervous system being a pattern-forming system that can flexibly switch from one coherent state to another. Thus, they can represent the key concepts to describe its complexity and its concerted, holistic behavior.
Collapse
Affiliation(s)
- Diego Guidolin
- Department of Neuroscience, University of Padova, Padova, Italy.
| | | | | | - Luigi F Agnati
- Department of Biomedical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
15
|
Mickle AM, Tanner JJ, Olowofela B, Wu S, Garvan C, Lai S, Addison A, Przkora R, Edberg JC, Staud R, Redden D, Goodin BR, Price CC, Fillingim RB, Sibille KT. Elucidating individual differences in chronic pain and whole person health with allostatic load biomarkers. Brain Behav Immun Health 2023; 33:100682. [PMID: 37701788 PMCID: PMC10493889 DOI: 10.1016/j.bbih.2023.100682] [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: 06/23/2023] [Revised: 08/12/2023] [Accepted: 08/26/2023] [Indexed: 09/14/2023] Open
Abstract
Chronic pain is a stressor that affects whole person functioning. Persistent and prolonged activation of the body's stress systems without adequate recovery can result in measurable physiological and neurobiological dysregulation recognized as allostatic load. We and others have shown chronic pain is associated with measures of allostatic load including clinical biomarker composites, telomere length, and brain structures. Less is known regarding how different measures of allostatic load align. The purpose of the study was to evaluate relationships among two measures of allostatic load: a clinical composite and pain-related brain structures, pain, function, and socioenvironmental measures. Participants were non-Hispanic black and non-Hispanic white community-dwelling adults between 45 and 85 years old with knee pain. Data were from a brain MRI, questionnaires specific to pain, physical and psychosocial function, and a blood draw. Individuals with all measures for the clinical composite were included in the analysis (n = 175). Indicating higher allostatic load, higher levels of the clinical composite were associated with thinner insula cortices with trends for thinner inferior temporal lobes and dorsolateral prefrontal cortices (DLPFC). Higher allostatic load as measured by the clinical composite was associated with greater knee osteoarthritis pathology, pain disability, and lower physical function. Lower allostatic load as indicated by thicker insula cortices was associated with higher income and education, and greater physical functioning. Thicker insula and DLPFC were associated with a lower chronic pain stage. Multiple linear regression models with pain and socioenvironmental measures as the predictors were significant for the clinical composite, insular, and inferior temporal lobes. We replicate our previously reported bilateral temporal lobe group difference pattern and show that individuals with high chronic pain stage and greater socioenvironmental risk have a higher allostatic load as measured by the clinical composite compared to those individuals with high chronic pain stage and greater socioenvironmental buffers. Although brain structure differences are shown in individuals with chronic pain, brain MRIs are not yet clinically applicable. Our findings suggest that a clinical composite measure of allostatic load may help identify individuals with chronic pain who have biological vulnerabilities which increase the risk for poor health outcomes.
Collapse
Affiliation(s)
- Angela M. Mickle
- Department of Physical Medicine & Rehabilitation, University of Florida, 101 Newell Dr, Gainesville, FL 32603, USA
| | - Jared J. Tanner
- Department of Clinical and Health Psychology, University of Florida, 1225 Center Dr, Gainesville, FL 32603, USA
| | - Bankole Olowofela
- Department of Anesthesiology, Division of Pain Medicine, University of Florida, 1600 SW Archer Road, Gainesville, FL. 32610, USA
| | - Stanley Wu
- Department of Physical Medicine & Rehabilitation, University of Florida, 101 Newell Dr, Gainesville, FL 32603, USA
| | - Cynthia Garvan
- Department of Anesthesiology, Division of Pain Medicine, University of Florida, 1600 SW Archer Road, Gainesville, FL. 32610, USA
| | - Song Lai
- Department of Radiation Oncology & CTSI Human Imaging Core, University of Florida, 2004 Mowry Rd Gainesville, FL 32610, USA
| | - Adriana Addison
- Department of Psychology, University of Alabama at Birmingham, Campbell Hall 415, 1300 University Blvd, Birmingham, AL, 35223, USA
| | - Rene Przkora
- Department of Anesthesiology, Division of Pain Medicine, University of Florida, 1600 SW Archer Road, Gainesville, FL. 32610, USA
| | - Jeffrey C. Edberg
- Department of Medicine, Division of Clinical Immunology & Rheumatology, University of Alabama at Birmingham, 1825 University Blvd, Birmingham, AL 35294, USA
| | - Roland Staud
- Department of Medicine, University of Florida, PO Box 100277, Gainesville, FL, USA
| | - David Redden
- Department of Biostatistics, The University of Alabama at Birmingham, 1665 University Boulevard, Birmingham, AL, USA
| | - Burel R. Goodin
- Department of Psychology, University of Alabama at Birmingham, Campbell Hall 415, 1300 University Blvd, Birmingham, AL, 35223, USA
- Department of Anesthesiology, Washington University, 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | - Catherine C. Price
- Department of Clinical and Health Psychology, University of Florida, 1225 Center Dr, Gainesville, FL 32603, USA
| | - Roger B. Fillingim
- Department of Community of Dentistry, University of Florida, 1329 SW 16th St, Room 5180, Gainesville, FL 32610, USA
| | - Kimberly T. Sibille
- Department of Physical Medicine & Rehabilitation, University of Florida, 101 Newell Dr, Gainesville, FL 32603, USA
- Department of Anesthesiology, Division of Pain Medicine, University of Florida, 1600 SW Archer Road, Gainesville, FL. 32610, USA
| |
Collapse
|
16
|
Ma Q, Wonnacott S, Bailey SJ, Bailey CP. Sex Differences in Brain Region-Specific Activation of c-Fos following Kappa Opioid Receptor Stimulation or Acute Stress in Mice. Int J Mol Sci 2023; 24:15098. [PMID: 37894779 PMCID: PMC10606335 DOI: 10.3390/ijms242015098] [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: 08/29/2023] [Revised: 09/29/2023] [Accepted: 10/02/2023] [Indexed: 10/29/2023] Open
Abstract
Kappa opioid receptors (KOPr) are involved in the response to stress. KOPr are also targets for the treatment of stress-related psychiatric disorders including depression, anxiety, and addiction although effects of KOPr are often sex-dependent. Here we investigated c-Fos expression in a range of brain regions in male and female mice following an acute stressor, and a single injection of KOPr agonist. Using adult C57BL/6 c-Fos-GFP transgenic mice and quantitative fluorescence microscopy, we identified brain regions activated in response to a challenge with the KOPr agonist U50,488 (20 mg/kg) or an acute stress (15 min forced swim stress, FSS). In male mice, U50,488 increased expression of c-Fos in the prelimbic area of the prefrontal cortex (PFCx), nucleus accumbens (NAcc), and basolateral nuclei of the amygdala (BLA). In contrast, in female mice U50,488 only activated the BLA but not the PFCx or the NAcc. FSS increased activation of PFCx, NAcc, and BLA in males while there was no activation of the PFCx in female mice. In both sexes, the KOPr antagonist norBNI significantly blocked U50,488-induced, but not stress-induced activation of brain regions. In separate experiments, activated cells were confirmed as non-GABAergic neurons in the PFCx and NAcc. Together these data demonstrate sex differences in activation of brain regions that are key components of the 'reward' circuitry. These differential responses may contribute to sex differences in stress-related psychiatric disorders and in the treatment of depression, anxiety, and addiction.
Collapse
Affiliation(s)
| | | | - Sarah J. Bailey
- Correspondence: (S.J.B.); (C.P.B.); Tel.: +44-(0)1225-383-935 (C.P.B.)
| | | |
Collapse
|
17
|
Zugman A, Alliende L, Medel V, Bethlehem RA, Seidlitz J, Ringlein G, Arango C, Arnatkevičiūtė A, Asmal L, Bellgrove M, Benegal V, Bernardo M, Billeke P, Bosch-Bayard J, Bressan R, Busatto G, Castro M, Chaim-Avancini T, Compte A, Costanzi M, Czepielewski L, Dazzan P, de la Fuente-Sandoval C, Di Forti M, Díaz-Caneja C, María Díaz-Zuluaga A, Du Plessis S, Duran F, Fittipaldi S, Fornito A, Freimer N, Gadelha A, Gama C, Garani R, Garcia-Rizo C, Gonzalez Campo C, Gonzalez-Valderrama A, Guinjoan S, Holla B, Ibañez A, Ivanovic D, Jackowski A, Leon-Ortiz P, Lochner C, López-Jaramillo C, Luckhoff H, Massuda R, McGuire P, Miyata J, Mizrahi R, Murray R, Ozerdem A, Pan P, Parellada M, Phahladira L, Ramirez-Mahaluf J, Reckziegel R, Reis Marques T, Reyes-Madrigal F, Roos A, Rosa P, Salum G, Scheffler F, Schumann G, Serpa M, Stein D, Tepper A, Tiego J, Ueno T, Undurraga J, Undurraga E, Valdes-Sosa P, Valli I, Villarreal M, Winton-Brown T, Yalin N, Zamorano F, Zanetti M, Winkler A, Pine D, Evans-Lacko S, Crossley N. Country-level gender inequality is associated with structural differences in the brains of women and men. Proc Natl Acad Sci U S A 2023; 120:e2218782120. [PMID: 37155867 PMCID: PMC10193926 DOI: 10.1073/pnas.2218782120] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 03/15/2023] [Indexed: 05/10/2023] Open
Abstract
Gender inequality across the world has been associated with a higher risk to mental health problems and lower academic achievement in women compared to men. We also know that the brain is shaped by nurturing and adverse socio-environmental experiences. Therefore, unequal exposure to harsher conditions for women compared to men in gender-unequal countries might be reflected in differences in their brain structure, and this could be the neural mechanism partly explaining women's worse outcomes in gender-unequal countries. We examined this through a random-effects meta-analysis on cortical thickness and surface area differences between adult healthy men and women, including a meta-regression in which country-level gender inequality acted as an explanatory variable for the observed differences. A total of 139 samples from 29 different countries, totaling 7,876 MRI scans, were included. Thickness of the right hemisphere, and particularly the right caudal anterior cingulate, right medial orbitofrontal, and left lateral occipital cortex, presented no differences or even thicker regional cortices in women compared to men in gender-equal countries, reversing to thinner cortices in countries with greater gender inequality. These results point to the potentially hazardous effect of gender inequality on women's brains and provide initial evidence for neuroscience-informed policies for gender equality.
Collapse
Affiliation(s)
- André Zugman
- Section on Development and Affective Neuroscience (SDAN), Emotion and Development Branch (E & D), National Institute of Mental Health, National Institutes of Health, BethesdaMD20894
| | - Luz María Alliende
- Department of Psychiatry, School of Medicine, Pontificia Universidad Católica de Chile, Santiago8330077, Chile
- Department of Psychology, Northwestern University, Evanston, IL60208
| | - Vicente Medel
- Latin American Brain Health Institute (BrainLat), Universidad Adolfo Ibáñez, Santiago7941169, Chile
| | - Richard A.I. Bethlehem
- Autism Research Centre, Department of Psychiatry, University of Cambridge, CambridgeCB2 8AH, United Kingdom
- Department of Psychology, University of Cambridge, CambridgeCB2 3EB, United Kingdom
| | - Jakob Seidlitz
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA19104
- Department of Child and Adolescent Psychiatry and Behavioral Science, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
- Penn-Children’s Hospital of Philadelphia Lifespan Brain Institute, University of Pennsylvania, Philadelphia, PA19104
| | - Grace Ringlein
- Section on Development and Affective Neuroscience (SDAN), Emotion and Development Branch (E & D), National Institute of Mental Health, National Institutes of Health, BethesdaMD20894
| | - Celso Arango
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry and Mental Health, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Centro de Investigación Biomédica en Red Salud Mental (CIBERSAM), School of Medicine, Universidad Complutense, Madrid28009, Spain
| | - Aurina Arnatkevičiūtė
- The Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Melbourne, VIC3168, Australia
- Monash Biomedical Imaging, Monash University, Melbourne, VIC3168, Australia
| | - Laila Asmal
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town7602, South Africa
| | - Mark Bellgrove
- The Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Melbourne, VIC3168, Australia
| | - Vivek Benegal
- Centre for Addiction Medicine, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, Karnataka560029, India
| | - Miquel Bernardo
- Barcelona Clinic Schizophrenia Unit, Hospital Clínic de Barcelona, Departament de Medicina, Institut de Neurociències (UBNeuro), Universitat de Barcelona (UB), Institut d’Investigacions Biomèdiques, August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red Salud Mental (CIBERSAM), Instituto de Salud Carlos III (ISCIII), Barcelona08036, Spain
| | - Pablo Billeke
- Laboratorio de Neurociencia Social y Neuromodulación, Centro de Investigación en Complejidad Social (neuroCICS), Facultad de Gobierno, Universidad del Desarrollo, Santiago7610658, Chile
| | - Jorge Bosch-Bayard
- McGill Centre for Integrative Neuroscience, Ludmer Centre for Neuroinformatics and Mental Health, Montreal Neurological Institute, Montreal, QCH3A 2B4, Canada
- McGill University, Montreal, QCH3A 2B4, Canada
| | - Rodrigo Bressan
- Interdisciplinary Laboratory in Clinical Neuroscience (LiNC), Department of Psychiatry, Federal University of São Paulo, São Paulo04039-032, Brazil
| | - Geraldo F. Busatto
- Departamento e Instituto de Psiquiatria, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo05403-903, Brazil
| | - Mariana N. Castro
- Grupo de Investigación en Neurociencias Aplicadas a las Alteraciones de la Conducta (INAAC), Fleni-Consejo Nacional de Investigaciones Científicas y Técnicas Neurosciences Institute (INEU), Ciudad Autónoma de Buenos AiresC1428, Argentina
- Department of Psychiatry and Mental Health, School of Medicine, University of Buenos Aires, Ciudad Autónoma de Buenos AiresC1114AAD, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos AiresC1033AAJ, Argentina
| | - Tiffany Chaim-Avancini
- Laboratory of Psychiatric Neuroimaging (LIM-21), Departamento e Instituto de Psiquiatria, Hospital das Clinicas, Faculdade de Medicina Universidade de São Paulo (HCFMUSP), Faculdade de Medicina Universidade de São Paulo, São PauloSP05403-903, Brazil
| | - Albert Compte
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona08036, Spain
| | - Monise Costanzi
- Laboratory of Molecular Psychiatry, Centro de Pesquisa Clínica, Hospital de Clínicas de Porto Alegre, Porto AlegreRS90035-007, Brazil
| | - Leticia Czepielewski
- Laboratory of Molecular Psychiatry, Centro de Pesquisa Clínica, Hospital de Clínicas de Porto Alegre, Porto AlegreRS90035-007, Brazil
- Programa de Pós-Graduação em Psicologia, Instituto Psicologia, Universidade Federal do Rio Grande do Sul, Porto AlegreRS90040-060, Brazil
| | - Paola Dazzan
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, LondonSE5 8AF, United Kingdom
| | - Camilo de la Fuente-Sandoval
- Laboratory of Experimental Psychiatry, Direction of Research, Instituto Nacional de Neurología y Neurocirugía, Mexico City14269, Mexico
| | - Marta Di Forti
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, LondonSE5 8AF, United Kingdom
- National Institute for Health Research (NIHR), Maudsley Biomedical Research Centre, South London and Maudsley NHS Foundation Trust, King’s College London, LondonSE5 8AZ, United Kingdom
| | - Covadonga M. Díaz-Caneja
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry and Mental Health, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Centro de Investigación Biomédica en Red Salud Mental (CIBERSAM), School of Medicine, Universidad Complutense, Madrid28009, Spain
| | - Ana María Díaz-Zuluaga
- Department of Psychiatry, Faculty of Medicine, University of Antioquia, Medellín050011, Colombia
- Center for Neurobehavioral Genetics, Jane and Terry Semel Institute for Neuroscience and Human Behavior Los Angeles, University of California Los Angeles (UCLA), Los Angeles, CA90024
| | - Stefan Du Plessis
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town7602, South Africa
- South African Medical Research Council (SA MRC), Genomics of Brain Disorders Unit, Cape Town7505, South Africa
| | - Fabio L. S. Duran
- Laboratory of Psychiatric Neuroimaging (LIM-21), Departamento e Instituto de Psiquiatria, Hospital das Clinicas, Faculdade de Medicina Universidade de São Paulo (HCFMUSP), Faculdade de Medicina Universidade de São Paulo, São PauloSP05403-903, Brazil
| | - Sol Fittipaldi
- Latin American Brain Health Institute (BrainLat), Universidad Adolfo Ibáñez, Santiago7941169, Chile
- Cognitive Neuroscience Center (CNC), Universidad de San Andres, Victoria, Ciudad Autónoma de Buenos AiresB1644BID, Argentina
- Global Brain Health Institute (GBHI), Trinity College Dublin (TCD), DublinDO2 PN40, Ireland
- Global Brain Health Institute (GBHI), University of California San Francisco (UCSF), San Francisco, CA94158
| | - Alex Fornito
- The Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Melbourne, VIC3168, Australia
- Monash Biomedical Imaging, Monash University, Melbourne, VIC3168, Australia
| | - Nelson B. Freimer
- Center for Neurobehavioral Genetics, Jane and Terry Semel Institute for Neuroscience and Human Behavior Los Angeles, University of California Los Angeles (UCLA), Los Angeles, CA90024
| | - Ary Gadelha
- Interdisciplinary Laboratory in Clinical Neuroscience (LiNC), Department of Psychiatry, Federal University of São Paulo, São Paulo04039-032, Brazil
| | - Clarissa S. Gama
- Laboratory of Molecular Psychiatry, Centro de Pesquisa Clínica, Hospital de Clínicas de Porto Alegre, Porto AlegreRS90035-007, Brazil
- Department of Psychiatry, Universidade Federal do Rio Grande do Sul (UFRGS), Hospital de Clinicas de Porto Alegre, Porto Alegre, RS90035903, Brazil
| | - Ranjini Garani
- Integrated Program in Neuroscience, McGill University, Montreal, QuebecH3A 1A12B4Canada
| | - Clemente Garcia-Rizo
- Barcelona Clinic Schizophrenia Unit, Hospital Clínic de Barcelona, Departament de Medicina, Institut de Neurociències (UBNeuro), Universitat de Barcelona (UB), Institut d’Investigacions Biomèdiques, August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red Salud Mental (CIBERSAM), Instituto de Salud Carlos III (ISCIII), Barcelona08036, Spain
| | - Cecilia Gonzalez Campo
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos AiresC1033AAJ, Argentina
- Cognitive Neuroscience Center (CNC), Universidad de San Andres, Victoria, Ciudad Autónoma de Buenos AiresB1644BID, Argentina
| | - Alfonso Gonzalez-Valderrama
- Early Intervention Program, Instituto Psiquiátrico Dr. J. Horwitz Barak, Santiago8431621, Chile
- School of Medicine, Universidad Finis Terrae, Santiago7501015, Chile
| | - Salvador Guinjoan
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos AiresC1033AAJ, Argentina
- Laureate Institute for Brain Research, Tulsa, OK74136
| | - Bharath Holla
- Department of Integrative Medicine, NIMHANS, Bengaluru, Karnataka560029, India
- Accelerator Program for Discovery in Brain disorders using Stem cells, Department of Psychiatry, NIMHANS, Bengaluru, Karnataka560029, India
| | - Agustín Ibañez
- Latin American Brain Health Institute (BrainLat), Universidad Adolfo Ibáñez, Santiago7941169, Chile
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos AiresC1033AAJ, Argentina
- Cognitive Neuroscience Center (CNC), Universidad de San Andres, Victoria, Ciudad Autónoma de Buenos AiresB1644BID, Argentina
- Global Brain Health Institute (GBHI), Trinity College Dublin (TCD), DublinDO2 PN40, Ireland
- Global Brain Health Institute (GBHI), University of California San Francisco (UCSF), San Francisco, CA94158
| | - Daniza Ivanovic
- Laboratorio de Neurociencia Social y Neuromodulación, Centro de Investigación en Complejidad Social (neuroCICS), Facultad de Gobierno, Universidad del Desarrollo, Santiago7610658, Chile
| | - Andrea Jackowski
- Department of Psychiatry, Universidade Federal de São Paulo, São Paulo04038-000, Brazil
- Department of Education, Information and Communications Technology (ICT) and Learning, Østfold University College, Halden1757, Norway
| | - Pablo Leon-Ortiz
- Laboratory of Experimental Psychiatry, Direction of Research, Instituto Nacional de Neurología y Neurocirugía, Mexico City14269, Mexico
| | - Christine Lochner
- South African Medical Research Council (SA MRC) Unit on Risk and Resilience in Mental Disorders, Department of Psychiatry, Stellenbosch University, Stellenbosch7505, South Africa
| | - Carlos López-Jaramillo
- Department of Psychiatry, Faculty of Medicine, University of Antioquia, Medellín050011, Colombia
| | - Hilmar Luckhoff
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town7602, South Africa
| | - Raffael Massuda
- Department of Psychiatry, Universidade Federal do Paraná (UFPR), CuritibaPR 80060-000, Brazil
| | - Philip McGuire
- Department of Psychiatry, University of Oxford, OxfordOX3 7JX, United Kingdom
- Wellcome Centre for Integrative Neuroimaging (WIN), University of Oxford, OxfordOX3 9DU, United Kingdom
- NIHR Oxford Health Biomedical Research Centre, OxfordOX3 7JX, United Kingdom
- Oxford HealthNational Health Service (NHS), Foundation Trust, OxfordOX4 4XN, United Kingdom
| | - Jun Miyata
- Department of Psychiatry, Graduate School of Medicine, Kyoto University, Kyoto606-8507, Japan
| | - Romina Mizrahi
- Integrated Program in Neuroscience, McGill University, Montreal, QuebecH3A 1A12B4Canada
- Clinical and Translational Sciences Lab, McGill University, Douglas Mental Health University Institute, Montreal, QCH4A 1R3, Canada
- Department of Psychiatry, McGill University,Montreal, QCH3A 1A1, Canada
| | - Robin Murray
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, LondonSE5 8AF, United Kingdom
| | - Aysegul Ozerdem
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MinnesotaMN55905
| | - Pedro M. Pan
- Interdisciplinary Laboratory in Clinical Neuroscience (LiNC), Department of Psychiatry, Federal University of São Paulo, São Paulo04039-032, Brazil
- National Institute of Developmental Psychiatry for Children and Adolescents, São Paulo04038-000, Brazil
| | - Mara Parellada
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry and Mental Health, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Centro de Investigación Biomédica en Red Salud Mental (CIBERSAM), School of Medicine, Universidad Complutense, Madrid28009, Spain
| | - Lebogan Phahladira
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town7602, South Africa
| | - Juan P. Ramirez-Mahaluf
- Department of Psychiatry, School of Medicine, Pontificia Universidad Católica de Chile, Santiago8330077, Chile
| | - Ramiro Reckziegel
- Laboratory of Molecular Psychiatry, Centro de Pesquisa Clínica, Hospital de Clínicas de Porto Alegre, Porto AlegreRS90035-007, Brazil
| | - Tiago Reis Marques
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, LondonSE5 8AF, United Kingdom
| | - Francisco Reyes-Madrigal
- Laboratory of Experimental Psychiatry, Direction of Research, Instituto Nacional de Neurología y Neurocirugía, Mexico City14269, Mexico
| | - Annerine Roos
- South African Medical Research Council (SA MRC) Unit on Risk and Resilience in Mental Disorders, Department of Psychiatry and Neuroscience Institute, University of Cape Town, Cape Town7925, South Africa
| | - Pedro Rosa
- Laboratory of Psychiatric Neuroimaging (LIM-21), Departamento e Instituto de Psiquiatria, Hospital das Clinicas, Faculdade de Medicina Universidade de São Paulo (HCFMUSP), Faculdade de Medicina Universidade de São Paulo, São PauloSP05403-903, Brazil
| | - Giovanni Salum
- Department of Psychiatry, Universidade Federal do Rio Grande do Sul (UFRGS), Hospital de Clinicas de Porto Alegre, Porto Alegre, RS90035903, Brazil
- National Institute of Developmental Psychiatry for Children and Adolescents, São Paulo04038-000, Brazil
| | - Freda Scheffler
- South African Medical Research Council (SA MRC) Unit on Risk and Resilience in Mental Disorders, Department of Psychiatry and Neuroscience Institute, University of Cape Town, Cape Town7925, South Africa
| | - Gunter Schumann
- Centre for Population Neuroscience and Stratified Medicine (PONS), Institute for Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai200433, China
- PONS-Centre, Charité Mental Health, Dept of Psychiatry and Psychotherapy, Charité Campus Mitte, Berlin10117, Germany
| | - Mauricio Serpa
- Departamento e Instituto de Psiquiatria, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo05403-903, Brazil
| | - Dan J. Stein
- South African Medical Research Council (SA MRC) Unit on Risk and Resilience in Mental Disorders, Department of Psychiatry and Neuroscience Institute, University of Cape Town, Cape Town7925, South Africa
| | - Angeles Tepper
- Department of Psychiatry, School of Medicine, Pontificia Universidad Católica de Chile, Santiago8330077, Chile
| | - Jeggan Tiego
- The Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Melbourne, VIC3168, Australia
- Monash Biomedical Imaging, Monash University, Melbourne, VIC3168, Australia
| | - Tsukasa Ueno
- Department of Psychiatry, Graduate School of Medicine, Kyoto University, Kyoto606-8507, Japan
- Integrated Clinical Education Center, Kyoto University Hospital, Kyoto606-8397, Japan
| | - Juan Undurraga
- Early Intervention Program, Instituto Psiquiátrico Dr. J. Horwitz Barak, Santiago8431621, Chile
- Department of Neurology and Psychiatry, Faculty of Medicine, Clínica Alemana Universidad del DesarrolloVitacura, Santiago7650568, Chile
| | - Eduardo A. Undurraga
- Escuela de Gobierno, Pontificia Universidad Católica de Chile, Santiago7820436, Chile
- Research Center for Integrated Disaster Risk Management (CIGIDEN), Santiago7820436, Chile
- Canadian Institute for Advanced Research (CIFAR), Azrieli Global Scholars Program, CIFAR, Toronto, ONM5G 1M1, Canada
| | - Pedro Valdes-Sosa
- The Clinical Hospital of Chengdu Brain Science Institute, University of Electronic Science and Technology of China, Chengdu610054, China
- Centro de Neurociencias de Cuba, La Habana11600, Cuba
| | - Isabel Valli
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona08036, Spain
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, LondonSE5 8AF, United Kingdom
| | - Mirta Villarreal
- Grupo de Investigación en Neurociencias Aplicadas a las Alteraciones de la Conducta (INAAC), Fleni-Consejo Nacional de Investigaciones Científicas y Técnicas Neurosciences Institute (INEU), Ciudad Autónoma de Buenos AiresC1428, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos AiresC1033AAJ, Argentina
- Department of Physics, Universidad de Buenos Aires, Ciudad Autónoma deBuenos AiresC1428EGA, Argentina
| | - Toby T. Winton-Brown
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC3004, Australia
- Department of Psychiatry, Alfred Health, Melbourne, VIC3004, Australia
| | - Nefize Yalin
- Centre for Affective Disorders, Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, LondonSE5 8AF, United Kingdom
- South London and Maudsley National Health Service (NHS), Foundation Trust, LondonSE5 8AZ, United Kingdom
| | - Francisco Zamorano
- Unidad de Imágenes Cuantitativas Avanzadas, Departamento de Imágenes, Clínica Alemana de Santiago, Universidad del Desarrollo, Santiago7650568, Chile
- Facultad de Ciencias para el Cuidado de la Salud, Universidad San Sebastián, Santiago7510602, Chile
| | - Marcus V. Zanetti
- Laboratory of Psychiatric Neuroimaging (LIM-21), Departamento e Instituto de Psiquiatria, Hospital das Clinicas, Faculdade de Medicina Universidade de São Paulo (HCFMUSP), Faculdade de Medicina Universidade de São Paulo, São PauloSP05403-903, Brazil
- Hospital Sírio-Libanês, São Paulo01308-050, Brazil
| | | | - Anderson M. Winkler
- Department of Human Genetics, University of Texas Rio Grande Valley, Brownsville, Texas TX78520
| | - Daniel S. Pine
- Section on Development and Affective Neuroscience (SDAN), Emotion and Development Branch (E & D), National Institute of Mental Health, National Institutes of Health, BethesdaMD20894
| | - Sara Evans-Lacko
- Care Policy and Evaluation Centre, School of Economics and Political Science, LondonWC2A 2AE, United Kingdom
| | - Nicolas A. Crossley
- Department of Psychiatry, School of Medicine, Pontificia Universidad Católica de Chile, Santiago8330077, Chile
- Department of Psychiatry, University of Oxford, OxfordOX3 7JX, United Kingdom
| |
Collapse
|
18
|
Kanes SJ, Dennie L, Perera P. Targeting the Arginine Vasopressin V 1b Receptor System and Stress Response in Depression and Other Neuropsychiatric Disorders. Neuropsychiatr Dis Treat 2023; 19:811-828. [PMID: 37077711 PMCID: PMC10106826 DOI: 10.2147/ndt.s402831] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 03/23/2023] [Indexed: 04/21/2023] Open
Abstract
A healthy stress response is critical for good mental and overall health and promotes neuronal growth and adaptation, but the intricately balanced biological mechanisms that facilitate a stress response can also result in predisposition to disease when that equilibrium is disrupted. The hypothalamic-pituitary-adrenal (HPA) axis neuroendocrine system plays a critical role in the body's response and adaptation to stress, and vasopressinergic regulation of the HPA axis is critical to maintaining system responsiveness during chronic stress. However, exposure to repeated or excessive physical or emotional stress or trauma can shift the body's stress response equilibrium to a "new normal" underpinned by enduring changes in HPA axis function. Exposure to early life stress due to adverse childhood experiences can also lead to lasting neurobiological changes, including in HPA axis function. HPA axis impairment in patients with depression is considered among the most reliable findings in biological psychiatry, and chronic stress has been shown to play a major role in the pathogenesis and onset of depression and other neuropsychiatric disorders. Modulating HPA axis activity, for example via targeted antagonism of the vasopressin V1b receptor, is a promising approach for patients with depression and other neuropsychiatric disorders associated with HPA axis impairment. Despite favorable preclinical indications in animal models, demonstration of clinical efficacy for the treatment of depressive disorders by targeting HPA axis dysfunction has been challenging, possibly due to the heterogeneity and syndromal nature of depressive disorders. Measures of HPA axis function, such as elevated cortisol levels, may be useful biomarkers for identifying patients who may benefit from treatments that modulate HPA axis activity. Utilizing clinical biomarkers to identify subsets of patients with impaired HPA axis function who may benefit is a promising next step in fine-tuning HPA axis activity via targeted antagonism of the V1b receptor.
Collapse
Affiliation(s)
- Stephen J Kanes
- EmbarkNeuro, Oakland, CA, USA
- Correspondence: Stephen J Kanes, EmbarkNeuro, Inc, 1111 Broadway, Suite 1300, Oakland, CA, 94607, USA, Tel +1 610 757 7821, Email
| | | | | |
Collapse
|
19
|
Marcolongo-Pereira C, Castro FCDAQ, Barcelos RM, Chiepe KCMB, Rossoni Junior JV, Ambrosio RP, Chiarelli-Neto O, Pesarico AP. Neurobiological mechanisms of mood disorders: Stress vulnerability and resilience. Front Behav Neurosci 2022; 16:1006836. [PMID: 36386785 PMCID: PMC9650072 DOI: 10.3389/fnbeh.2022.1006836] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 10/06/2022] [Indexed: 09/05/2023] Open
Abstract
Stress is an important factor in the development of several human pathologies. The response of rodents and humans to stress depends on many factors; some people and rodents develop stress-related mood disorders, such as depression and anxiety in humans, depression-like and anxiety-like behavior in mice and rats, while others report no new psychological symptoms in response to chronic or acute stress, and are considered susceptible and resilient to stress, respectively. Resilience is defined as the ability to thrive in the face of adversity and is a learned process that can help protect against occupational stressors and mental illnesses. There is growing interest in the underlying mechanisms involved in resilience and vulnerability to depression caused by stress, and some studies have demonstrated that individual variability in the way animals and humans respond to stress depends on several mechanisms, such as oxidative stress, neuronal plasticity, immunology and genetic factors, among others not discussed in this review, this review provides a general overview about this mechanism.
Collapse
Affiliation(s)
- Clairton Marcolongo-Pereira
- Coordenadoria de Pesquisa, Pós-Graduação e Extensão (CEPEG), Centro Universitário do Espírito Santo (UNESC), Colatina, Brazil
| | | | - Rafael Mazioli Barcelos
- Coordenadoria de Pesquisa, Pós-Graduação e Extensão (CEPEG), Centro Universitário do Espírito Santo (UNESC), Colatina, Brazil
| | | | - Joamyr Victor Rossoni Junior
- Coordenadoria de Pesquisa, Pós-Graduação e Extensão (CEPEG), Centro Universitário do Espírito Santo (UNESC), Colatina, Brazil
| | - Roberta Passamani Ambrosio
- Coordenadoria de Pesquisa, Pós-Graduação e Extensão (CEPEG), Centro Universitário do Espírito Santo (UNESC), Colatina, Brazil
| | - Orlando Chiarelli-Neto
- Coordenadoria de Pesquisa, Pós-Graduação e Extensão (CEPEG), Centro Universitário do Espírito Santo (UNESC), Colatina, Brazil
| | - Ana Paula Pesarico
- Curso de Medicina, Universidade Federal do Pampa (Unipampa), Bagé, Brazil
| |
Collapse
|
20
|
Salhab O, Khayat L, Alaaeddine N. Stem cell secretome as a mechanism for restoring hair loss due to stress, particularly alopecia areata: narrative review. J Biomed Sci 2022; 29:77. [PMID: 36199062 PMCID: PMC9533579 DOI: 10.1186/s12929-022-00863-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 09/28/2022] [Indexed: 12/03/2022] Open
Abstract
Background Living organisms are continuously exposed to multiple internal and external stimuli which may influence their emotional, psychological, and physical behaviors. Stress can modify brain structures, reduces functional memory and results in many diseases such as skin disorders like acne, psoriasis, telogen effluvium, and alopecia areata. In this review, we aim to discuss the effect of secretome on treating alopecia, especially alopecia areata. We will shed the light on the mechanism of action of the secretome in the recovery of hair loss and this by reviewing all reported in vitro and in vivo literature. Main body Hair loss has been widely known to be enhanced by stressful events. Alopecia areata is one of the skin disorders which can be highly induced by neurogenic stress especially if the patient has a predisposed genetic background. This condition is an autoimmune disease where stress in this case activates the immune response to attack the body itself leading to hair cycle destruction. The currently available treatments include medicines, laser therapy, phototherapy, and alternative medicine therapies with little or no satisfactory results. Regenerative medicine is a new era in medicine showing promising results in treating many medical conditions including Alopecia. The therapeutic effects of stem cells are due to their paracrine and trophic effects which are due to their secretions (secretome). Conclusion Stem cells should be more used as an alternative to conventional therapies due to their positive outcomes. More clinical trials on humans should be done to maximize the dose needed and type of stem cells that must be used to treat alopecia areata.
Collapse
Affiliation(s)
- Ola Salhab
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Luna Khayat
- University of Ottawa, Faculty of Science, Ottawa, ON, Canada
| | - Nada Alaaeddine
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon.
| |
Collapse
|
21
|
Eskandari F, Salimi M, Binayi F, Abdollahifar MA, Eftekhary M, Hedayati M, Ghanbarian H, Zardooz H. Investigating the Effects of Maternal Separation on Hypothalamic-Pituitary-Adrenal Axis and Glucose Homeostasis under Chronic Social Defeat Stress in Young Adult Male Rat Offspring. Neuroendocrinology 2022; 113:361-380. [PMID: 36088912 DOI: 10.1159/000526989] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 09/05/2022] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Given the suggested metabolic regulatory effects of stress-responsive genes and based on the impacts of early-life stress on HPA axis development, this study aimed to characterize the maternal separation (MS) impact on the communication between glucose metabolism and HPA axis dysregulations under chronic social defeat stress (CSDS). METHODS During the first 2 weeks of life, male Wistar rats were either exposed to MS or left undisturbed with their mothers (Std). Starting on postnatal day 50, the animals of each group were either left undisturbed in the standard group housing (Con) or underwent CSDS for 3 weeks. There were four groups (n = 10/group): Std-Con, MS-Con, Std-CSDS, and MS-CSDS. RESULTS Early and/or adult life adversity reduced β-cell number, muscular FK506-binding protein 51 (FKBP51) content, and BMI in adulthood. The reduction of β-cell number and BMI in the MS-CSDS rats were more profound than MS-Con group. CSDS either alone or in combination with MS reduced locomotor activity and increased and decreased corticotropin-releasing factor type 1 receptor (CRFR1) content, respectively, in hypothalamus and pancreas. Although, under CSDS, MS intensified HPA axis overactivity and reduced isolated islets' insulin secretion, it could promote resilience to depression symptoms. No differences were observed in hypothalamic Fkbp5 gene DNA methylation and glucose tolerance among groups. CONCLUSION MS exacerbated HPA axis overactivity and the endocrine pancreas dysfunctions under CSDS. The intensified corticosterone secretion and the diminished content of pancreatic CRFR1 protein could be involved in the reduced β-cell number and islets' insulin secretion under CSDS. The decreased muscular FKBP51 content might be a homeostatic response to slow down insulin resistance development under chronic stress.
Collapse
Affiliation(s)
- Farzaneh Eskandari
- Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mina Salimi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fateme Binayi
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad-Amin Abdollahifar
- Department of Biology and Anatomical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohamad Eftekhary
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Hedayati
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Ghanbarian
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Homeira Zardooz
- Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
22
|
Hahad O, Bayo Jimenez MT, Kuntic M, Frenis K, Steven S, Daiber A, Münzel T. Cerebral consequences of environmental noise exposure. ENVIRONMENT INTERNATIONAL 2022; 165:107306. [PMID: 35635962 DOI: 10.1016/j.envint.2022.107306] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/09/2022] [Accepted: 05/15/2022] [Indexed: 06/15/2023]
Abstract
The importance of noise exposure as a major environmental determinant of public health is being increasingly recognized. While in recent years a large body evidence has emerged linking environmental noise exposure mainly to cardiovascular disease, much less is known concerning the adverse health effects of noise on the brain and associated neuropsychiatric outcomes. Despite being a relatively new area of investigation, indeed, mounting research and conclusive evidence demonstrate that exposure to noise, primarily from traffic sources, may affect the central nervous system and brain, thereby contributing to an increased risk of neuropsychiatric disorders such as stroke, dementia and cognitive decline, neurodevelopmental disorders, depression, and anxiety disorder. On a mechanistic level, a significant number of studies suggest the involvement of reactive oxygen species/oxidative stress and inflammatory pathways, among others, to fundamentally drive the adverse brain health effects of noise exposure. This in-depth review on the cerebral consequences of environmental noise exposure aims to contribute to the associated research needs by evaluating current findings from human and animal studies. From a public health perspective, these findings may also help to reinforce efforts promoting adequate mitigation strategies and preventive measures to lower the societal consequences of unhealthy environments.
Collapse
Affiliation(s)
- Omar Hahad
- Department of Cardiology - Cardiology I, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), partner site Rhine-Main, Mainz, Germany; Leibniz Institute for Resilience Research (LIR), Mainz, Germany.
| | - Maria Teresa Bayo Jimenez
- Department of Cardiology - Cardiology I, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Marin Kuntic
- Department of Cardiology - Cardiology I, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Katie Frenis
- Boston Children's Hospital and Harvard Medical School, Department of Hematology/Oncology, Boston, MA, USA
| | - Sebastian Steven
- Department of Cardiology - Cardiology I, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), partner site Rhine-Main, Mainz, Germany
| | - Andreas Daiber
- Department of Cardiology - Cardiology I, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), partner site Rhine-Main, Mainz, Germany
| | - Thomas Münzel
- Department of Cardiology - Cardiology I, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), partner site Rhine-Main, Mainz, Germany
| |
Collapse
|
23
|
Szafoni S, Piegza M. Progress in Personalized Psychiatric Therapy with the Example of Using Intranasal Oxytocin in PTSD Treatment. J Pers Med 2022; 12:1067. [PMID: 35887564 PMCID: PMC9317706 DOI: 10.3390/jpm12071067] [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: 05/20/2022] [Revised: 06/21/2022] [Accepted: 06/28/2022] [Indexed: 11/22/2022] Open
Abstract
Post-traumatic stress disorder (PTSD) is a severe mental disorder that results in the frequent coexistence of other diseases, lowers patients' quality of life, and has a high annual cost of treatment. However, despite the variety of therapeutic approaches that exist, some patients still do not achieve the desired results. In addition, we may soon face an increase in the number of new PTSD cases because of the current global situation-both the COVID-19 pandemic and the ongoing armed conflicts. Hence, in recent years, many publications have sought a new, more personalized treatment approach. One such approach is the administration of intranasal oxytocin (INOXT), which, due to its pleiotropic effects, seems to be a promising therapeutic option. However, the current findings suggest that it might only be helpful for a limited, strictly selected group of patients.
Collapse
Affiliation(s)
- Sandra Szafoni
- Department of Psychiatry, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 42-612 Tarnowskie Góry, Poland;
| | | |
Collapse
|
24
|
Lv X, Zhang X, Zhao Q, Li C, Zhang T, Yang X. Acute stress promotes brain oscillations and hippocampal-cortical dialog in emotional processing. Biochem Biophys Res Commun 2022; 598:55-61. [PMID: 35151204 DOI: 10.1016/j.bbrc.2022.01.116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 01/28/2022] [Indexed: 12/30/2022]
Abstract
Hippocampal-cortical circuit oscillations in local field potential (LFP) represent network-level signals which promotes behavior. Investigating these signals promote our understanding on how the brain process cognition and emotion, and provide further perspectives into electroencephalogram endophenotypes, especially under the pathological state. The physiological adaptive stress responses to threatening stimuli are critical for individuals. The disturbance of stress response may lead to psychiatric disorders such as major depressive disorder (MDD). To quantitatively examine the effects of acute stress on hippocampal-cortical circuit, we recorded LFPs in the hippocampus (HC) and the medial prefrontal cortex (mPFC). We analyzed three major LFP oscillations with their temporal coupling. Consistent with our hypothesis that strengthened communication of hippocampal-cortical circuit may occur in stress adaption, we found that intensive acute stress induced enhanced ripple-delta-spindle coupling. The LFP coupling may facilitate the recruitment of relevant structures in hippocampal-cortical circuit, in response to acute stress, and play a role in emotional encoding migration.
Collapse
Affiliation(s)
- Xin Lv
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Xiaolin Zhang
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Qian Zhao
- Institute of Psychology and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai, 200030, China; Laboratory of Molecular Neurodegeneration, Graduate School of Biomedical Systems and Technologies, Institute of Biomedical Systems and Biotechnology, Peter the Great St. Petersburg Polytechnic University, Saint Petersburg, 195251, Russia
| | - Chunbo Li
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| | - Tianhong Zhang
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| | - Xiangyu Yang
- Institute of Psychology and Behavioral Sciences, Shanghai Jiao Tong University, Shanghai, 200030, China; Laboratory of Molecular Neurodegeneration, Graduate School of Biomedical Systems and Technologies, Institute of Biomedical Systems and Biotechnology, Peter the Great St. Petersburg Polytechnic University, Saint Petersburg, 195251, Russia.
| |
Collapse
|
25
|
Caradonna SG, Zhang TY, O’Toole N, Shen MJ, Khalil H, Einhorn NR, Wen X, Parent C, Lee FS, Akil H, Meaney MJ, McEwen BS, Marrocco J. Genomic modules and intramodular network concordance in susceptible and resilient male mice across models of stress. Neuropsychopharmacology 2022; 47:987-999. [PMID: 34848858 PMCID: PMC8938529 DOI: 10.1038/s41386-021-01219-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 09/28/2021] [Accepted: 10/18/2021] [Indexed: 12/24/2022]
Abstract
The multifactorial etiology of stress-related disorders necessitates a constant interrogation of the molecular convergences in preclinical models of stress that use disparate paradigms as stressors spanning from environmental challenges to genetic predisposition to hormonal signaling. Using RNA-sequencing, we investigated the genomic signatures in the ventral hippocampus common to mouse models of stress. Chronic oral corticosterone (CORT) induced increased anxiety- and depression-like behavior in wild-type male mice and male mice heterozygous for the gene coding for brain-derived neurotrophic factor Val66Met, a variant associated with genetic susceptibility to stress. In a separate set of male mice, chronic social defeat stress (CSDS) led to a susceptible or a resilient population, whose proportion was dependent on housing conditions, namely standard housing or enriched environment. Rank-rank-hypergeometric overlap (RRHO), a threshold-free approach that ranks genes by their p value and effect size direction, was used to identify genes from a continuous gradient of significancy that were concordant across groups. In mice treated with CORT and in standard-housed susceptible mice, differentially expressed genes (DEGs) were concordant for gene networks involved in neurotransmission, cytoskeleton function, and vascularization. Weighted gene co-expression analysis generated 54 gene hub modules and revealed two modules in which both CORT and CSDS-induced enrichment in DEGs, whose function was concordant with the RRHO predictions, and correlated with behavioral resilience or susceptibility. These data showed transcriptional concordance across models in which the stress coping depends upon hormonal, environmental, or genetic factors revealing common genomic drivers that embody the multifaceted nature of stress-related disorders.
Collapse
Affiliation(s)
- Salvatore G. Caradonna
- grid.134907.80000 0001 2166 1519Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY USA
| | - Tie-Yuan Zhang
- grid.14709.3b0000 0004 1936 8649Douglas Mental Health University Institute, McGill University, Montreal, QC Canada
| | - Nicholas O’Toole
- grid.14709.3b0000 0004 1936 8649Douglas Mental Health University Institute, McGill University, Montreal, QC Canada
| | - Mo-Jun Shen
- grid.452264.30000 0004 0530 269XSingapore Institute for Clinical Sciences, Singapore, Singapore
| | - Huzefa Khalil
- grid.214458.e0000000086837370Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI USA
| | - Nathan R. Einhorn
- grid.134907.80000 0001 2166 1519Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY USA
| | - Xianglan Wen
- grid.14709.3b0000 0004 1936 8649Douglas Mental Health University Institute, McGill University, Montreal, QC Canada
| | - Carine Parent
- grid.14709.3b0000 0004 1936 8649Douglas Mental Health University Institute, McGill University, Montreal, QC Canada
| | - Francis S. Lee
- grid.5386.8000000041936877XDepartment of Psychiatry, Sackler Institute for Developmental Psychobiology, Weill Cornell Medical College, New York, NY USA
| | - Huda Akil
- grid.214458.e0000000086837370Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI USA
| | - Michael J. Meaney
- grid.14709.3b0000 0004 1936 8649Douglas Mental Health University Institute, McGill University, Montreal, QC Canada ,grid.452264.30000 0004 0530 269XSingapore Institute for Clinical Sciences, Singapore, Singapore ,grid.4280.e0000 0001 2180 6431Yong Loo Lin School of Medicine, Singapore, Singapore ,grid.14709.3b0000 0004 1936 8649Sackler Program for Epigenetics & Psychobiology, McGill University, Montreal, QC Canada
| | - Bruce S. McEwen
- grid.134907.80000 0001 2166 1519Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY USA
| | - Jordan Marrocco
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
26
|
Laham BJ, Gould E. How Stress Influences the Dynamic Plasticity of the Brain’s Extracellular Matrix. Front Cell Neurosci 2022; 15:814287. [PMID: 35145379 PMCID: PMC8821883 DOI: 10.3389/fncel.2021.814287] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 12/27/2021] [Indexed: 12/14/2022] Open
Abstract
Diffuse and structured extracellular matrix (ECM) comprise ∼20% of the brain’s volume and play important roles in development and adult plasticity. Perineuronal nets (PNNs), specialized ECM structures that surround certain types of neurons in the brain, emerge during the postnatal period, making their development and maintenance potentially sensitive to experience. Recent studies have shown that stress affects diffuse ECM as well as PNNs, and that such effects are dependent on life stage and brain region. Given that the ECM participates in synaptic plasticity, the generation of neuronal oscillations, and synchronous firing across brain regions, all of which have been linked to cognition and emotional regulation, ECM components may be candidate therapeutic targets for stress-induced neuropsychiatric disease. This review considers the influence of stress over diffuse and structured ECM during postnatal life with a focus on functional outcomes and the potential for translational relevance.
Collapse
|
27
|
Waters RC, Worth HM, Vasquez B, Gould E. Inhibition of adult neurogenesis reduces avoidance behavior in male, but not female, mice subjected to early life adversity. Neurobiol Stress 2022; 17:100436. [PMID: 35146080 PMCID: PMC8819473 DOI: 10.1016/j.ynstr.2022.100436] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/08/2022] [Accepted: 01/24/2022] [Indexed: 12/20/2022] Open
Abstract
Early life adversity (ELA) increases the risk of developing neuropsychiatric illnesses such as anxiety disorders. However, the mechanisms connecting these negative early life experiences to illness later in life remain unclear. In rodents, plasticity mechanisms, specifically adult neurogenesis in the ventral hippocampus, have been shown to be altered by ELA and important for buffering against detrimental stress-induced outcomes. The current study sought to explore whether adult neurogenesis contributes to ELA-induced changes in avoidance behavior. Using the GFAP-TK transgenic model, which allows for the inhibition of adult neurogenesis, and CD1 littermate controls, we subjected mice to an ELA paradigm of maternal separation and early weaning (MSEW) or control rearing. We found that mice with intact adult neurogenesis showed no behavioral changes in response to MSEW. After reducing adult neurogenesis, however, male mice previously subjected to MSEW had an unexpected decrease in avoidance behavior. This finding was not observed in female mice, suggesting that a sex difference exists in the role of adult-born neurons in buffering against ELA-induced changes in behavior. Taken together with the existing literature on ELA and avoidance behavior, this work suggests that strain differences exist in susceptibility to ELA and that adult-born neurons may play a role in regulating adaptive behavior.
Collapse
|
28
|
Radulovic J, Ivkovic S, Adzic M. From chronic stress and anxiety to neurodegeneration: Focus on neuromodulation of the axon initial segment. HANDBOOK OF CLINICAL NEUROLOGY 2022; 184:481-495. [PMID: 35034756 DOI: 10.1016/b978-0-12-819410-2.00025-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
To adapt to the sustained demands of chronic stress, discrete brain circuits undergo structural and functional changes often resulting in anxiety disorders. In some individuals, anxiety disorders precede the development of motor symptoms of Parkinson's disease (PD) caused by degeneration of neurons in the substantia nigra (SN). Here, we present a circuit framework for probing a causal link between chronic stress, anxiety, and PD, which postulates a central role of abnormal neuromodulation of the SN's axon initial segment by brainstem inputs. It is grounded in findings demonstrating that the earliest PD pathologies occur in the stress-responsive, emotion regulation network of the brainstem, which provides the SN with dense aminergic and cholinergic innervation. SN's axon initial segment (AIS) has unique features that support the sustained and bidirectional propagation of activity in response to synaptic inputs. It is therefore, especially sensitive to circuit-mediated stress-induced imbalance of neuromodulation, and thus a plausible initiating site of neurodegeneration. This could explain why, although secondary to pathophysiologies in other brainstem nuclei, SN degeneration is the most extensive. Consequently, the cardinal symptom of PD, severe motor deficits, arise from degeneration of the nigrostriatal pathway rather than other brainstem nuclei. Understanding when and how circuit dysfunctions underlying anxiety can progress to neurodegeneration, raises the prospect of timed interventions for reversing, or at least impeding, the early pathophysiologies that lead to PD and possibly other neurodegenerative disorders.
Collapse
Affiliation(s)
- Jelena Radulovic
- Department of Neuroscience, Albert Einstein Medical College, Bronx, NY, United States; Department of Psychiatry and Behavioral Sciences, Albert Einstein Medical College, Bronx, NY, United States.
| | - Sanja Ivkovic
- Department of Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Miroslav Adzic
- Department of Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
29
|
Sahani V, Hurd YL, Bachi K. Neural Underpinnings of Social Stress in Substance Use Disorders. Curr Top Behav Neurosci 2022; 54:483-515. [PMID: 34971448 PMCID: PMC9177516 DOI: 10.1007/7854_2021_272] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
BACKGROUND Drug addiction is a complex brain disorder that is characterized by craving, withdrawal, and relapse, which can be perpetuated by social stress. Stemming from an acute life event, chronic stress, or trauma in a social context, social stress has a major role in the initiation and trajectory of substance use. Preclinical literature shows that early life stress exposure and social isolation facilitate and enhance drug self-administration. Epidemiological evidence links childhood adversity to increased risk for drug use and demonstrates that cumulative stress experiences are predictive of substance use severity in a dose-dependent manner. Stress and drug use induce overlapping brain alterations leading to downregulation or deficits in brain reward circuitry, thereby resulting in greater sensitization to the rewarding properties of drugs. Though stress in the context of addiction has been studied at the neural level, a gap in our understanding of the neural underpinnings of social stress in humans remains. METHODS We conducted a systematic review of in vivo structural and functional neuroimaging studies to evaluate the neural processes associated with social stress in individuals with substance use disorder. Results were considered in relation to participants' history of social stress and with regard to the effects of social stress induced during the neuroimaging paradigm. RESULTS An exhaustive search yielded 21 studies that matched inclusion criteria. Social stress induces broad structural and functional neural effects in individuals with substance use disorder throughout their lifespan and across drug classes. A few patterns emerged across studies: (1) many of the brain regions altered in individuals who were exposed to chronic social stress and during acute stress induction have been implicated in addiction networks (including the prefrontal cortex, insula, hippocampus, and amygdala); (2) individuals with childhood maltreatment and substance use history had decreased gray matter or activation in regions of executive functioning (including the medial prefrontal cortex, orbitofrontal cortex, anterior cingulate cortex), the hippocampal complex, and the supplementary motor area; and (3) during stress-induction paradigms, activation in the anterior cingulate cortex, caudate, and amygdala was most commonly observed. CONCLUSIONS/IMPLICATIONS A distinct overlap is shown between social stress-related circuitry and addiction circuitry, particularly in brain regions implicated in drug-seeking, craving, and relapse. Given the few studies that have thoroughly investigated social stress, the evidence accumulated to date needs to be replicated and extended, particularly using research designs and methods that disentangle the effects of substance use from social stress. Future clinical studies can leverage this information to evaluate the impact of exposure to trauma or adverse life events within substance use research. Expanding knowledge in this emerging field could help clarify neural mechanisms underlying addiction risk and progression to guide causal-experimental inquiry and novel prevention and treatment strategies.
Collapse
Affiliation(s)
- Vyoma Sahani
- Department of Psychiatry, Addiction Institute of Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yasmin L Hurd
- Department of Psychiatry, Addiction Institute of Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Keren Bachi
- Department of Psychiatry, Addiction Institute of Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
30
|
Piacentino D, Sani G, Kotzalidis GD, Cappelletti S, Longo L, Rizzato S, Fabi F, Frati P, Fineschi V, Leggio L. Anabolic androgenic steroids used as performance and image enhancing drugs in professional and amateur athletes: Toxicological and psychopathological findings. Hum Psychopharmacol 2022; 37:e2815. [PMID: 34528289 PMCID: PMC8727496 DOI: 10.1002/hup.2815] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/31/2021] [Accepted: 08/24/2021] [Indexed: 01/03/2023]
Abstract
OBJECTIVE The use of anabolic androgenic steroids (AASs) as performance and image enhancing drugs (PIEDs), once restricted to professional athletes, now includes amateurs and regular gym visitors. AAS use is associated with psychopathology, yet this relationship is complex and not fully understood. We aimed to assess the presence of AASs and other misused substances in athletes' biological samples and link toxicological to psychopathological findings. METHODS A multicentre, cross-sectional study in fitness centres in Italy recruited 122 professional and amateur athletes training in several sports (84 men; age range = 18-45 years). Athletes completed questionnaires, interviews, and toxicology testing for AASs, other PIEDs, illicit drugs, and non-prescribed psychotropics. Toxicology was conducted in blood, urine, and hair. RESULTS Self-reported and toxicologically detected use rates of AASs and other misused substances showed slight-to-fair agreement (Fleiss' κ = 0.104-0.375). There was slight-to-moderate agreement among the three biological samples used for AAS testing (κ = 0.112-0.436). Thirty-one athletes (25.4%) tested positive for AASs. More sport hours/week, narcissistic or antisocial personality disorders, and higher nonplanning impulsiveness scores predicted AAS use (pseudo-R2 = 0.665). AAS users did not differ significantly from non-users in major psychopathology, but their Hypomania Checklist-32 score, which also predicted AAS use, was significantly higher (p < 0.001), suggesting increased odds for cyclothymic disorder or subthreshold hypomania. CONCLUSIONS Our results have implications for studying AAS users, as they identify a cluster of variables that may be relevant in future understanding of AAS use risks (e.g., personality disorders). Possible disagreements between AAS assessment methods should be considered when implementing harm reduction interventions, such as needle and syringe distribution, health education, and counselling, as well as surveillance programmes.
Collapse
Affiliation(s)
- Daria Piacentino
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore and Bethesda, MD, USA,Center on Compulsive Behaviors, National Institutes of Health, Bethesda, MD, USA,Neurosciences, Mental Health, and Sensory Organs (NESMOS) Department, School of Medicine and Psychology, Sapienza University of Rome, Rome, Italy,Correspondence: Daria Piacentino, M.D., Ph.D., M.S., Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, NIDA IRP and NIAAA DICBR, National Institutes of Health, 251 Bayview Blvd, Room 02A406, Baltimore, MD 21224, Phone: 443-740-2793,
| | - Gabriele Sani
- Department of Neuroscience, Psychiatry Section, Catholic University of Rome, Rome, Italy
| | - Georgios D. Kotzalidis
- Neurosciences, Mental Health, and Sensory Organs (NESMOS) Department, School of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Simone Cappelletti
- Anatomical, Histological, Forensic Medicine, and Orthopedic Sciences (SAIMLAL) Department, Sapienza University of Rome, Rome, Italy
| | - Livia Longo
- Neurosciences, Mental Health, and Sensory Organs (NESMOS) Department, School of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Salvatore Rizzato
- Neurosciences, Mental Health, and Sensory Organs (NESMOS) Department, School of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Francesco Fabi
- Centre for Statistical and Social Studies (CE3S), Rome, Italy
| | - Paola Frati
- Anatomical, Histological, Forensic Medicine, and Orthopedic Sciences (SAIMLAL) Department, Sapienza University of Rome, Rome, Italy
| | - Vittorio Fineschi
- Anatomical, Histological, Forensic Medicine, and Orthopedic Sciences (SAIMLAL) Department, Sapienza University of Rome, Rome, Italy
| | - Lorenzo Leggio
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, National Institutes of Health, Baltimore and Bethesda, MD, USA,Center on Compulsive Behaviors, National Institutes of Health, Bethesda, MD, USA,Medication Development Program, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA,Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University School of Public Health, Providence, RI, USA,Division of Addiction Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA,Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| |
Collapse
|
31
|
Brandner S, Schroeter S, Çalışkan G, Salar S, Kobow K, Coras R, Blümcke I, Hamer H, Schwarz M, Buchfelder M, Maslarova A. Glucocorticoid modulation of synaptic plasticity in the human temporal cortex of epilepsy patients: Does chronic stress contribute to memory impairment? Epilepsia 2021; 63:209-221. [PMID: 34687218 DOI: 10.1111/epi.17107] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/11/2021] [Accepted: 10/11/2021] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Memory impairment is common in patients with temporal lobe epilepsy and seriously affects life quality. Chronic stress is a recognized cofactor in epilepsy and can also impair memory function. Furthermore, increased cortisol levels have been reported in epilepsy patients. Animal models have suggested that aggravating effects of stress on memory and synaptic plasticity were mediated via glucocorticoids. The aim of this study was, therefore, to investigate the effect of glucocorticoid receptor (GR) modulation on synaptic plasticity in the human cortex of epilepsy patients. METHODS We performed field potential recordings in acute slices from the temporal neocortex of patients who underwent surgery for drug-resistant temporal lobe epilepsy. Synaptic plasticity was investigated by a theta-burst stimulation (TBS) protocol for induction of long-term potentiation (LTP) in the presence of GR modulators. RESULTS LTP was impaired in temporal cortex from epilepsy patients. Pretreatment of the slices with the GR antagonist mifepristone (RU486) improved LTP induction, suggesting that LTP impairment was due to baseline GR activation in the human cortex. The highly potent GR agonist dexamethasone additionally weakened synaptic strength in an activity-dependent manner when applied after TBS. SIGNIFICANCE Our results show a direct negative glucocorticoid effect on synaptic potentiation in the human cortex and imply chronic activation of GRs. Chronic stress may therefore contribute to memory impairment in patients with temporal lobe epilepsy. Furthermore, the activity-dependent acute inhibitory effect of dexamethasone suggests a mechanism of synaptic downscaling by which postictally increased cortisol levels may prevent pathologic plasticity upon seizures.
Collapse
Affiliation(s)
- Sebastian Brandner
- Department of Neurosurgery, Erlangen University Hospital, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Sarah Schroeter
- Department of Neurosurgery, Erlangen University Hospital, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany.,Department of Orthopedic, Trauma, and Hand Surgery, Osnabrück Clinic, Osnabrück, Germany
| | - Gürsel Çalışkan
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto von Guericke University Magdeburg, Magdeburg, Germany.,Center for Behavioral Brain Sciences, Magdeburg, Germany
| | - Seda Salar
- Department of Psychiatry and Psychotherapy, Erlangen University Hospital, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Katja Kobow
- Department of Neuropathology, Erlangen University Hospital, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Roland Coras
- Department of Neuropathology, Erlangen University Hospital, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Ingmar Blümcke
- Department of Neuropathology, Erlangen University Hospital, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Hajo Hamer
- Department of Neurology, Epilepsy Center, Erlangen University Hospital, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Michael Schwarz
- Department of Neurology, Epilepsy Center, Erlangen University Hospital, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Michael Buchfelder
- Department of Neurosurgery, Erlangen University Hospital, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Anna Maslarova
- Department of Neurosurgery, Erlangen University Hospital, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
32
|
Papp M, Gruca P, Lason M, Litwa E, Solecki W, Willner P. Insufficiency of ventral hippocampus to medial prefrontal cortex transmission explains antidepressant non-response. J Psychopharmacol 2021; 35:1253-1264. [PMID: 34617804 PMCID: PMC8521380 DOI: 10.1177/02698811211048281] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND There is extensive evidence that antidepressant drugs restore normal brain function by repairing damage to ventral hippocampus (vHPC) and medial prefrontal cortex (mPFC). While the damage is more extensive in hippocampus, the evidence of treatments, such as deep brain stimulation, suggests that functional changes in prefrontal cortex may be more critical. We hypothesized that antidepressant non-response may result from an insufficiency of transmission from vHPC to mPFC. METHOD Antidepressant non-responsive Wistar Kyoto (WKY) rats were subjected to chronic mild stress (CMS), then treated with chronic daily administration of the antidepressant drug venlafaxine (VEN) and/or repeated weekly optogenetic stimulation (OGS) of afferents to mPFC originating from vHPC or dorsal HPC (dHPC). RESULTS As in many previous studies, CMS decreased sucrose intake, open-arm entries on the elevated plus maze (EPM), and novel object recognition (NOR). Neither VEN nor vHPC-mPFC OGS alone was effective in reversing the effects of CMS, but the combination of chronic VEN and repeated OGS restored normal behaviour on all three measures. dHPC-mPFC OGS restored normal behaviour in the EPM and NOR test irrespective of concomitant VEN treatment, and had no effect on sucrose intake. CONCLUSIONS The synergism between VEN and vHPC-mPFC OGS supports the hypothesis that the antidepressant non-responsiveness of WKY rats results from a failure of antidepressant treatment fully to restore transmission in the vHPC-mPFC pathway.
Collapse
Affiliation(s)
- Mariusz Papp
- Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland,Mariusz Papp, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna Street, Krakow 31-343, Poland.
| | - Piotr Gruca
- Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Magdalena Lason
- Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Ewa Litwa
- Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Wojciech Solecki
- Department of Neurobiology and Neuropsychology, Institute of Applied Psychology, Jagiellonian University, Krakow, Poland
| | - Paul Willner
- Department of Psychology, Swansea University, Swansea, UK
| |
Collapse
|
33
|
Koyama Y, Nawa N, Yamaoka Y, Nishimura H, Sonoda S, Kuramochi J, Miyazaki Y, Fujiwara T. Interplay between social isolation and loneliness and chronic systemic inflammation during the COVID-19 pandemic in Japan: Results from U-CORONA study. Brain Behav Immun 2021; 94:51-59. [PMID: 33705870 PMCID: PMC7939973 DOI: 10.1016/j.bbi.2021.03.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 03/05/2021] [Accepted: 03/05/2021] [Indexed: 01/16/2023] Open
Abstract
In the face of the global coronavirus disease 2019 (COVID-19) pandemic, billions of people were forced to stay at home due to the implementation of social distancing and lockdown policies. As a result, individuals lost their social relationships, leading to social isolation and loneliness. Both social isolation and loneliness are major risk factors for poor physical and mental health status through enhanced chronic inflammation; however, there might be an interplay between social isolation and loneliness on the association with chronic inflammation. We aimed to clarify the link between social relationships and inflammation in the context of the COVID-19 pandemic by distinguishing whether social isolation only, loneliness only, or both were associated with chronic inflammation markers among community-dwelling adults. The data of 624 people (aged 18-92 years, mean 51.4) from the Utsunomiya COVID-19 seROprevalence Neighborhood Association (U-CORONA) study, which targeted randomly sampled households in Utsunomiya city, Japan, were analyzed. Social isolation was assessed as a structural social network by asking the number of social roles they have on a daily basis. Loneliness was measured with the UCLA loneliness scale. As chronic inflammation biomarkers, neutrophil-to-lymphocyte ratio (NLR) and the concentration of high-sensitivity C-reactive protein (CRP) were measured. Generalized estimating equations method was employed to take into account the correlations within households. Isolated-Lonely condition (i.e., being both socially isolated and feeling lonely) was associated with higher NLR among men (B = 0.141, 95%CI = -0.01 to 0.29). Interestingly, Nonisolated-Lonely condition (i.e., not socially isolated but feeling lonely) was associated with lower CRP among women (B = -0.462, 95%CI = -0.82 to -0.10) and among the working-age population (B = -0.495, 95%CI = -0.76 to -0.23). In conclusion, being both socially isolated and feeling lonely was associated with chronic inflammation. Assessing both social isolation and loneliness is critical for proper interventions to mitigate the impact of poor social relationships on health, especially in the context of the COVID-19 pandemic.
Collapse
Affiliation(s)
- Yuna Koyama
- Department of Global Health Promotion, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Nobutoshi Nawa
- Department of Medical Education Research and Development, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Yui Yamaoka
- Department of Global Health Promotion, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Hisaaki Nishimura
- Department of Global Health Promotion, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Shiro Sonoda
- Kuramochi Clinic Interpark, Utsunomiya, Tochigi, Japan
| | - Jin Kuramochi
- Kuramochi Clinic Interpark, Utsunomiya, Tochigi, Japan
| | - Yasunari Miyazaki
- Department of Respiratory Medicine, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Takeo Fujiwara
- Department of Global Health Promotion, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.
| |
Collapse
|
34
|
Aghayeva U, Bhattacharya A, Sural S, Jaeger E, Churgin M, Fang-Yen C, Hobert O. DAF-16/FoxO and DAF-12/VDR control cellular plasticity both cell-autonomously and via interorgan signaling. PLoS Biol 2021; 19:e3001204. [PMID: 33891586 PMCID: PMC8099054 DOI: 10.1371/journal.pbio.3001204] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 05/05/2021] [Accepted: 03/23/2021] [Indexed: 01/08/2023] Open
Abstract
Many cell types display the remarkable ability to alter their cellular phenotype in response to specific external or internal signals. Such phenotypic plasticity is apparent in the nematode Caenorhabditis elegans when adverse environmental conditions trigger entry into the dauer diapause stage. This entry is accompanied by structural, molecular, and functional remodeling of a number of distinct tissue types of the animal, including its nervous system. The transcription factor (TF) effectors of 3 different hormonal signaling systems, the insulin-responsive DAF-16/FoxO TF, the TGFβ-responsive DAF-3/SMAD TF, and the steroid nuclear hormone receptor, DAF-12/VDR, a homolog of the vitamin D receptor (VDR), were previously shown to be required for entering the dauer arrest stage, but their cellular and temporal focus of action for the underlying cellular remodeling processes remained incompletely understood. Through the generation of conditional alleles that allowed us to spatially and temporally control gene activity, we show here that all 3 TFs are not only required to initiate tissue remodeling upon entry into the dauer stage, as shown before, but are also continuously required to maintain the remodeled state. We show that DAF-3/SMAD is required in sensory neurons to promote and then maintain animal-wide tissue remodeling events. In contrast, DAF-16/FoxO or DAF-12/VDR act cell-autonomously to control anatomical, molecular, and behavioral remodeling events in specific cell types. Intriguingly, we also uncover non-cell autonomous function of DAF-16/FoxO and DAF-12/VDR in nervous system remodeling, indicating the presence of several insulin-dependent interorgan signaling axes. Our findings provide novel perspectives into how hormonal systems control tissue remodeling.
Collapse
Affiliation(s)
- Ulkar Aghayeva
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, New York, United States of America
| | - Abhishek Bhattacharya
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, New York, United States of America
| | - Surojit Sural
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, New York, United States of America
| | - Eliza Jaeger
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, New York, United States of America
| | - Matthew Churgin
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Christopher Fang-Yen
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Oliver Hobert
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
35
|
Ambhore NS, Kalidhindi RSR, Sathish V. Sex-Steroid Signaling in Lung Diseases and Inflammation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1303:243-273. [PMID: 33788197 DOI: 10.1007/978-3-030-63046-1_14] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Sex/gender difference exists in the physiology of multiple organs. Recent epidemiological reports suggest the influence of sex-steroids in modulating a wide variety of disease conditions. Sex-based discrepancies have been reported in pulmonary physiology and various chronic inflammatory responses associated with lung diseases like asthma, chronic obstructive pulmonary disease (COPD), pulmonary fibrosis, and rare lung diseases. Notably, emerging clinical evidence suggests that several respiratory diseases affect women to a greater degree, with increased severity and prevalence than men. Although sex-specific differences in various lung diseases are evident, such differences are inherent to sex-steroids, which are major biological variables in men and women who play a central role to control these differences. The focus of this chapter is to comprehend the sex-steroid biology in inflammatory lung diseases and to understand the mechanistic role of sex-steroids signaling in regulating these diseases. Exploring the roles of sex-steroid signaling in the regulation of lung diseases and inflammation is crucial for the development of novel and effective therapy. Overall, we will illustrate the importance of differential sex-steroid signaling in lung diseases and their possible clinical implications for the development of complementary and alternative medicine to treat lung diseases.
Collapse
Affiliation(s)
- Nilesh Sudhakar Ambhore
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, ND, USA
| | | | - Venkatachalem Sathish
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, ND, USA.
| |
Collapse
|
36
|
Šutulović N, Grubač Ž, Šuvakov S, Jerotić D, Puškaš N, Macut D, Rašić-Marković A, Simić T, Stanojlović O, Hrnčić D. Experimental Chronic Prostatitis/Chronic Pelvic Pain Syndrome Increases Anxiety-Like Behavior: The Role of Brain Oxidative Stress, Serum Corticosterone, and Hippocampal Parvalbumin-Positive Interneurons. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6687493. [PMID: 33815658 PMCID: PMC7990537 DOI: 10.1155/2021/6687493] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 02/08/2021] [Accepted: 02/10/2021] [Indexed: 12/21/2022]
Abstract
Mechanisms of the brain-related comorbidities in chronic prostatitis/chronic pelvic pain syndrome (CP/CPPS) are still largely unknown, although CP/CPPS is one of the major urological problems in middle-aged men, while these neuropsychological incapacities considerably diminish life quality. The objectives of this study were to assess behavioral patterns in rats with CP/CPPS and to determine whether these patterns depend on alterations in the brain oxidative stress, corticosterone, and hippocampal parvalbumin-positive (PV+) interneurons. Adult male Wistar albino rats from CP/CPPS (intraprostatic injection of 3% λ-carrageenan, day 0) and sham (0.9% NaCl) groups were subjected to pain and anxiety-like behavior tests (days 2, 3, and 7). Afterwards, rats were sacrificed and biochemical and immunohistochemical analyses were performed. Scrotal allodynia and prostatitis were proven in CP/CPPS, but not in sham rats. Ethological tests (open field, elevated plus maze, and light/dark tests) revealed significantly increased anxiety-like behavior in rats with CP/CPPS comparing to their sham-operated mates starting from day 3, and there were significant intercorrelations among parameters of these tests. Increased oxidative stress in the hippocampus, thalamus, and cerebral cortex, as well as increased serum corticosterone levels and decreased number of hippocampal PV+ neurons, was shown in CP/CPPS rats, compared to sham rats. Increased anxiety-like behavior in CP/CPPS rats was significantly correlated with these brain biochemical and hippocampal immunohistochemical alterations. Therefore, the potential mechanisms of observed behavioral alterations in CP/CPPS rats could be the result of an interplay between increased brain oxidative stress, elevated serum corticosterone level, and loss of hippocampal PV+ interneurons.
Collapse
Affiliation(s)
- Nikola Šutulović
- Institute of Medical Physiology “Richard Burian”, Belgrade University Faculty of Medicine, 11000 Belgrade, Serbia
| | - Željko Grubač
- Institute of Medical Physiology “Richard Burian”, Belgrade University Faculty of Medicine, 11000 Belgrade, Serbia
| | - Sonja Šuvakov
- Institute of Clinical and Medical Biochemistry, Belgrade University Faculty of Medicine, 11000 Belgrade, Serbia
| | - Djurdja Jerotić
- Institute of Clinical and Medical Biochemistry, Belgrade University Faculty of Medicine, 11000 Belgrade, Serbia
| | - Nela Puškaš
- Institute of Histology and Embryology “Aleksandar Đ. Kostić”, Belgrade University Faculty of Medicine, 11000 Belgrade, Serbia
| | - Djuro Macut
- Clinic of Endocrinology, Diabetes and Metabolic Disease, CCS, Belgrade University Faculty of Medicine, 11000 Belgrade, Serbia
| | - Aleksandra Rašić-Marković
- Institute of Medical Physiology “Richard Burian”, Belgrade University Faculty of Medicine, 11000 Belgrade, Serbia
| | - Tatjana Simić
- Institute of Clinical and Medical Biochemistry, Belgrade University Faculty of Medicine, 11000 Belgrade, Serbia
| | - Olivera Stanojlović
- Institute of Medical Physiology “Richard Burian”, Belgrade University Faculty of Medicine, 11000 Belgrade, Serbia
| | - Dragan Hrnčić
- Institute of Medical Physiology “Richard Burian”, Belgrade University Faculty of Medicine, 11000 Belgrade, Serbia
| |
Collapse
|
37
|
Quinn KG, Spector A, Takahashi L, Voisin DR. Conceptualizing the Effects of Continuous Traumatic Violence on HIV Continuum of Care Outcomes for Young Black Men Who Have Sex with Men in the United States. AIDS Behav 2021; 25:758-772. [PMID: 32944841 PMCID: PMC7886964 DOI: 10.1007/s10461-020-03040-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/14/2020] [Indexed: 01/06/2023]
Abstract
The United States (US) is on track to achieve the 90-90-90 targets set forth by UNAIDS and the National HIV/AIDS strategy, yet significant racial disparities in HIV care outcomes remain, particularly for young Black men who have sex with men (YBMSM). Research has demonstrated that various types of violence are key aspects of syndemics that contribute to disparities in HIV risk. However, little research has looked collectively at cumulative violent experiences and how those might affect HIV treatment and care outcomes. Drawing on extant literature and theoretical underpinnings of syndemics, we provide a conceptual model that highlights how continuous traumatic violence experienced by YBMSM may affect HIV outcomes and contribute to racial disparities in HIV outcomes. The findings of this focused review suggest a need for research on how continuous exposure to various types of violence influence HIV prevention and treatment outcomes for young Black MSM.
Collapse
Affiliation(s)
- Katherine G Quinn
- Center for AIDS Intervention Research (CAIR), Medical College of Wisconsin, 2071 N. Summit Ave., Milwaukee, WI, 53202, USA.
| | - Antoinette Spector
- Medical College of Wisconsin, Institute for Health Equity, Milwaukee, USA
| | | | | |
Collapse
|
38
|
Tanner JJ, Johnson AJ, Terry EL, Cardoso J, Garvan C, Staud R, Deutsch G, Deshpande H, Lai S, Addison A, Redden D, Goodin BR, Price CC, Fillingim RB, Sibille KT. Resilience, pain, and the brain: Relationships differ by sociodemographics. J Neurosci Res 2021; 99:1207-1235. [PMID: 33606287 DOI: 10.1002/jnr.24790] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 11/23/2020] [Accepted: 01/03/2021] [Indexed: 12/22/2022]
Abstract
Chronic musculoskeletal (MSK) pain is disabling to individuals and burdensome to society. A relationship between telomere length and resilience was reported in individuals with consideration for chronic pain intensity. While chronic pain associates with brain changes, little is known regarding the neurobiological interface of resilience. In a group of individuals with chronic MSK pain, we examined the relationships between a previously investigated resilience index, clinical pain and functioning measures, and pain-related brain structures, with consideration for sex and ethnicity/race. A cross-sectional analysis of 166 non-Hispanic Black and non-Hispanic White adults, 45-85 years of age with pain ≥ 1 body site (s) over the past 3 months was completed. Measures of clinical pain and functioning, biobehavioral and psychosocial resilience, and structural MRI were completed. Our findings indicate higher levels of resilience associate with lower levels of clinical pain and functional limitations. Significant associations between resilience, ethnicity/race, and/or sex, and pain-related brain gray matter structure were demonstrated in the right amygdaloid complex, bilateral thalamus, and postcentral gyrus. Our findings provide compelling evidence that in order to decipher the neurobiological code of chronic pain and related protective factors, it will be important to improve how chronic pain is phenotyped; to include an equal representation of females in studies including analyses stratifying by sex, and to consider other sociodemographic factors.
Collapse
Affiliation(s)
- Jared J Tanner
- Department of Clinical and Health Psychology, College of Public Health and Health Professions, University of Florida, Gainesville, FL, USA
| | - Alisa J Johnson
- Pain Research & Intervention Center of Excellence, University of Florida, Gainesville, FL, USA.,Department of Community Dentistry and Behavioral Science, University of Florida, Gainesville, FL, USA
| | - Ellen L Terry
- Pain Research & Intervention Center of Excellence, University of Florida, Gainesville, FL, USA.,Department of Biobehavioral Nursing Science, College of Nursing, University of Florida, Gainesville, FL, USA
| | - Josue Cardoso
- Pain Research & Intervention Center of Excellence, University of Florida, Gainesville, FL, USA
| | - Cynthia Garvan
- Department of Anesthesiology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Roland Staud
- Department of Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Georg Deutsch
- Department of Radiology, University of Alabama, Birmingham Medical Center, Birmingham, AL, USA
| | - Hrishikesh Deshpande
- Department of Radiology, University of Alabama, Birmingham Medical Center, Birmingham, AL, USA.,Department of Anesthesiology, University of Alabama, Birmingham Medical Center, Birmingham, AL, USA
| | - Song Lai
- Department of Radiation Oncology & CTSI Human Imaging Core, University of Florida, Gainesville, FL, USA
| | - Adriana Addison
- Department of Anesthesiology, University of Alabama, Birmingham Medical Center, Birmingham, AL, USA.,Department of Psychology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - David Redden
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Burel R Goodin
- Department of Anesthesiology, University of Alabama, Birmingham Medical Center, Birmingham, AL, USA.,Department of Psychology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Catherine C Price
- Department of Clinical and Health Psychology, College of Public Health and Health Professions, University of Florida, Gainesville, FL, USA.,Department of Anesthesiology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Roger B Fillingim
- Pain Research & Intervention Center of Excellence, University of Florida, Gainesville, FL, USA.,Department of Community Dentistry and Behavioral Science, University of Florida, Gainesville, FL, USA
| | - Kimberly T Sibille
- Pain Research & Intervention Center of Excellence, University of Florida, Gainesville, FL, USA.,Department of Anesthesiology, College of Medicine, University of Florida, Gainesville, FL, USA.,Department of Aging and Geriatric Research, College of Medicine, UF Pain Research & Intervention Center of Excellence, University of Florida, Gainesville, FL, USA
| |
Collapse
|
39
|
Boyce WT, Levitt P, Martinez FD, McEwen BS, Shonkoff JP. Genes, Environments, and Time: The Biology of Adversity and Resilience. Pediatrics 2021; 147:peds.2020-1651. [PMID: 33495368 DOI: 10.1542/peds.2020-1651] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/28/2020] [Indexed: 11/24/2022] Open
Abstract
Exposures to adverse environments, both psychosocial and physicochemical, are prevalent and consequential across a broad range of childhood populations. Such adversity, especially early in life, conveys measurable risk to learning and behavior and to the foundations of both mental and physical health. Using an interactive gene-environment-time (GET) framework, we survey the independent and interactive roles of genetic variation, environmental context, and developmental timing in light of advances in the biology of adversity and resilience, as well as new discoveries in biomedical research. Drawing on this rich evidence base, we identify 4 core concepts that provide a powerful catalyst for fresh thinking about primary health care for young children: (1) all biological systems are inextricably integrated, continuously "reading" and adapting to the environment and "talking back" to the brain and each other through highly regulated channels of cross-system communication; (2) adverse environmental exposures induce alterations in developmental trajectories that can lead to persistent disruptions of organ function and structure; (3) children vary in their sensitivity to context, and this variation is influenced by interactions among genetic factors, family and community environments, and developmental timing; and (4) critical or sensitive periods provide unmatched windows of opportunity for both positive and negative influences on multiple biological systems. These rapidly moving frontiers of investigation provide a powerful framework for new, science-informed thinking about health promotion and disease prevention in the early childhood period.
Collapse
Affiliation(s)
- W Thomas Boyce
- Departments of Pediatrics and Psychiatry, University of California, San Francisco, San Francisco, California
| | - Pat Levitt
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California.,Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Fernando D Martinez
- Asthma and Airway Disease Research Center, The University of Arizona, Tucson, Arizona
| | - Bruce S McEwen
- The Rockefeller University, New York, New York.,Deceased
| | - Jack P Shonkoff
- Center on the Developing Child and .,Harvard Graduate School of Education, Harvard University, Cambridge, Massachusetts.,Department of Social & Behavioral Sciences, Harvard T.H. Chan School of Public Health and.,Harvard Medical School and Boston Children's Hospital, Harvard University, Boston, Massachusetts.,Boston Children's Hospital, Boston, Massachusetts; and.,Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
40
|
Risal S, Manti M, Lu H, Fornes R, Larsson H, Benrick A, Deng Q, Cesta CE, Rosenqvist MA, Stener-Victorin E. Prenatal androgen exposure causes a sexually dimorphic transgenerational increase in offspring susceptibility to anxiety disorders. Transl Psychiatry 2021; 11:45. [PMID: 33441551 PMCID: PMC7806675 DOI: 10.1038/s41398-020-01183-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 11/30/2020] [Accepted: 12/15/2020] [Indexed: 12/11/2022] Open
Abstract
If and how obesity and elevated androgens in women with polycystic ovary syndrome (PCOS) affect their offspring's psychiatric health is unclear. Using data from Swedish population health registers, we showed that daughters of mothers with PCOS have a 78% increased risk of being diagnosed with anxiety disorders. We next generated a PCOS-like mouse (F0) model induced by androgen exposure during late gestation, with or without diet-induced maternal obesity, and showed that the first generation (F1) female offspring develop anxiety-like behavior, which is transgenerationally transmitted through the female germline into the third generation of female offspring (F3) in the androgenized lineage. In contrast, following the male germline, F3 male offspring (mF3) displayed anxiety-like behavior in the androgenized and the obese lineages. Using a targeted approach to search for molecular targets within the amygdala, we identified five differentially expressed genes involved in anxiety-like behavior in F3 females in the androgenized lineage and eight genes in the obese lineage. In mF3 male offspring, three genes were dysregulated in the obese lineage but none in the androgenized lineage. Finally, we performed in vitro fertilization (IVF) using a PCOS mouse model of continuous androgen exposure. We showed that the IVF generated F1 and F2 offspring in the female germline did not develop anxiety-like behavior, while the F2 male offspring (mF2) in the male germline did. Our findings provide evidence that elevated maternal androgens in PCOS and maternal obesity may underlie the risk of a transgenerational transmission of anxiety disorders in children of women with PCOS.
Collapse
Affiliation(s)
- Sanjiv Risal
- grid.4714.60000 0004 1937 0626Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Maria Manti
- grid.4714.60000 0004 1937 0626Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Haojiang Lu
- grid.4714.60000 0004 1937 0626Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Romina Fornes
- grid.4714.60000 0004 1937 0626Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Henrik Larsson
- grid.4714.60000 0004 1937 0626Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden ,grid.15895.300000 0001 0738 8966School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Anna Benrick
- grid.8761.80000 0000 9919 9582Department of Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden ,grid.412798.10000 0001 2254 0954School of Health Sciences, University of Skövde, Skövde, Sweden
| | - Qiaolin Deng
- grid.4714.60000 0004 1937 0626Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Carolyn E. Cesta
- grid.4714.60000 0004 1937 0626Department of Medicine, Solna, Centre for Pharmacoepidemiology, Karolinska Institutet, Stockholm, Sweden
| | - Mina A. Rosenqvist
- grid.15895.300000 0001 0738 8966School of Medical Sciences, Örebro University, Örebro, Sweden
| | | |
Collapse
|
41
|
Peters E, Hübner J, Katalinic A. [Stress, coping strategies and health-related quality of life during the corona pandemic in April 2020 in Germany]. Dtsch Med Wochenschr 2020; 146:e11-e20. [PMID: 33260231 PMCID: PMC7815330 DOI: 10.1055/a-1275-3792] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
INTRODUCTION International studies indicate that pandemics and quarantine can lead to significantly increased stress levels and mental illness in those affected. Stress levels and quality of life in selected population groups in the early phase of the lockdown of the corona pandemic were examined. Associations of coping strategies with perceived stress levels and associations of activities to increase well-being with health-related quality of life as an aspect of well-being are presented. METHODS Data from the first survey wave of the CoPa study were evaluated, which were collected via online survey. Group differences regarding stress and quality of life were explorative tested by means of Chi-square tests and T-tests. Associations of coping strategies with stress and of activities to increase well-being with health-related quality of life were calculated using linear regression analysis. RESULTS Among the 5315 participants, persons at risk of mental health and those who did not go out in public showed signs of depression, anxiety disorders and stress significantly more often than other participants. Persons with children under 12 years of age showed significantly higher stress levels than others and their health-related quality of life was comparable. Perceived social support and self-efficacy proved to be resources for stress. Humor, physical activity, healthy eating, maintaining daily routines and pursuing specific goals were positively associated with health-related quality of life. DISCUSSION Persons with mental health risks need therapeutic services in times of reduced contact. Selected measures to increase well-being appear to be effective and should be recommended.
Collapse
Affiliation(s)
- Elke Peters
- Institut für Sozialmedizin und Epidemiologie, Universität zu Lübeck, Lübeck
| | - Joachim Hübner
- Institut für Sozialmedizin und Epidemiologie, Universität zu Lübeck, Lübeck
| | - Alexander Katalinic
- Institut für Sozialmedizin und Epidemiologie, Universität zu Lübeck, Lübeck.,Institut für Krebsepidemiologie e. V., Universität zu Lübeck, Lübeck
| |
Collapse
|
42
|
Zafar T. Potential biomarkers of emotional stress induced neurodegeneration. eNeurologicalSci 2020; 21:100292. [PMID: 33294647 PMCID: PMC7695868 DOI: 10.1016/j.ensci.2020.100292] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 11/10/2020] [Accepted: 11/12/2020] [Indexed: 01/14/2023] Open
Abstract
Mental health is a matter of great significance and interest both socially and scientifically. The present review aims to provide an informative platform about the classical biomarkers available to identify and diagnose the neurodegeneration induced by emotional stress and depression. Present article provides an expert comprehensive overview of the universally accepted markers and their mechanism of action involved in emotional stress assessment and its management. This envisioned piece of work emphasize on the incorporation of clinical markers in classical psychiatry experiments will make the information more significant, reliable and universally accepted. The information summaries in the article will facilitates the researchers of clinical psychiatry, neuropharmacology and neuropsychiatry in management of depressive disorders along with the identification of possible neurodegenerative association. Health is an overall outcome of emotional, physiological, endocrinological and neurological factors. Emotional stress and depression are silent causes of neurodegeneration. Clinical markers play a vital role in the management of neurological health. Endocrine secretions play a vital role in mediation of various neurotransmission, nerves excitability and neurodegeneration.
Collapse
Affiliation(s)
- Tabassum Zafar
- Department of Bioscience, Faculty of Life Sciences, Barkatullah University, Bhopal 462026, Madhya Pradesh, India
| |
Collapse
|
43
|
Associations between Alzheimer's disease polygenic risk scores and hippocampal subfield volumes in 17,161 UK Biobank participants. Neurobiol Aging 2020; 98:108-115. [PMID: 33259984 DOI: 10.1016/j.neurobiolaging.2020.11.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 09/20/2020] [Accepted: 11/01/2020] [Indexed: 11/23/2022]
Abstract
Hippocampal volume is an important biomarker of Alzheimer's disease (AD), and genetic risk of AD is associated with hippocampal atrophy. However, the hippocampus is not a uniform structure and has a number of subfields, the associations of which with age, sex, and polygenic risk score for AD (PRSAD) have been inadequately investigated. We examined these associations in 17,161 cognitively normal UK Biobank participants (44-80 years). Age was negatively associated with all the hippocampal subfield volumes and females had smaller volumes than men. Higher PRSAD was associated with lower volumes in the bilateral whole hippocampus, hippocampal-amygdala-transition-area, and hippocampal tail; right subiculum; left cornu ammonis 1, cornu ammonis 4, molecular layer, and granule cell layer of dentate gyrus. Older individuals (median age 63 years, n = 8984) showed greater subfield vulnerability to high PRSAD compared to the younger group (n = 8177), but the effect did not differ by sex. The pattern of subfield involvement in relation to the PRSAD in community dwelling healthy individuals sheds additional light on the pathogenesis of AD.
Collapse
|
44
|
Lehtola SJ, Tuulari JJ, Scheinin NM, Karlsson L, Parkkola R, Merisaari H, Lewis JD, Fonov VS, Louis Collins D, Evans A, Saunavaara J, Hashempour N, Lähdesmäki T, Acosta H, Karlsson H. Newborn amygdalar volumes are associated with maternal prenatal psychological distress in a sex-dependent way. Neuroimage Clin 2020; 28:102380. [PMID: 32805677 PMCID: PMC7453059 DOI: 10.1016/j.nicl.2020.102380] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 08/07/2020] [Accepted: 08/07/2020] [Indexed: 12/20/2022]
Abstract
Maternal psychological distress during pregnancy (PPD)1 has been associated with changes in offspring amygdalar and hippocampal volumes. Studies on child amygdalae suggest that sex moderates the vulnerability of fetal brains to prenatal stress. However, this has not yet been observed in these structures in newborns. Newborn studies are crucial, as they minimize the confounding influence of postnatal life. We investigated the effects of maternal prenatal psychological symptoms on newborn amygdalar and hippocampal volumes and their interactions with newborn sex in 123 newborns aged 2-5 weeks (69 males, 54 females). Based on earlier studies, we anticipated small, but statistically significant effects of PPD on the volumes of these structures. Maternal psychological distress was measured at gestational weeks (GW)2 14, 24 and 34 using Symptom Checklist-90 (SCL-90, anxiety scale)3 and Edinburgh Postnatal Depression Scale (EPDS)4 questionnaires. Newborn sex was found to moderate the relationship between maternal distress symptoms at GW 24 and the volumes of left and right amygdala. This relationship was negative and significant only in males. No significant main effect or sex-based moderation was found for hippocampal volumes. This newborn study provides evidence for a sex-dependent influence of maternal psychiatric symptoms on amygdalar structural development. This association may be relevant to later psychopathology.
Collapse
Affiliation(s)
- Satu J Lehtola
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Institute of Clinical Medicine University of Turku, Turku, Finland.
| | - Jetro J Tuulari
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Institute of Clinical Medicine University of Turku, Turku, Finland; Department of Psychiatry, University of Turku and Turku University Hospital, Turku, Finland; Turku Collegium for Science and Medicine, University of Turku, Turku, Finland; Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Noora M Scheinin
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Institute of Clinical Medicine University of Turku, Turku, Finland; Department of Psychiatry, University of Turku and Turku University Hospital, Turku, Finland
| | - Linnea Karlsson
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Institute of Clinical Medicine University of Turku, Turku, Finland; Department of Child Psychiatry, University of Turku and Turku University Hospital, Turku, Finland; Center for Population Health Research, University of Turku and Turku University Hospital, Finland
| | - Riitta Parkkola
- Department of Radiology, University of Turku and Turku University Hospital, Turku, Finland
| | - Harri Merisaari
- Department of Future Technologies, University of Turku, Turku, Finland
| | - John D Lewis
- Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Vladimir S Fonov
- Montreal Neurological Institute, McGill University, Montreal, Canada
| | - D Louis Collins
- Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Alan Evans
- Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Jani Saunavaara
- Department of Medical Physics, Turku University Hospital, Turku, Finland
| | - Niloofar Hashempour
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Institute of Clinical Medicine University of Turku, Turku, Finland
| | - Tuire Lähdesmäki
- Department of Pediatric Neurology, University of Turku and Turku University Hospital, Turku, Finland
| | - Henriette Acosta
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Institute of Clinical Medicine University of Turku, Turku, Finland
| | - Hasse Karlsson
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Institute of Clinical Medicine University of Turku, Turku, Finland; Department of Psychiatry, University of Turku and Turku University Hospital, Turku, Finland; Center for Population Health Research, University of Turku and Turku University Hospital, Finland
| |
Collapse
|
45
|
Nasrin Faraji, Shiravi A, Bahari Z, Shirvani H, Meftahi GH. Basolateral Amygdala α1-Adrenergic Receptor Suppression Attenuates Stress-Induced Anxiety-Like Behavior and Spine Morphology Impairment on Hippocampal CA1 Pyramidal Neurons. NEUROCHEM J+ 2020. [DOI: 10.1134/s1819712420010079] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
46
|
Reding KM, Grayson DS, Miranda-Dominguez O, Ray S, Wilson ME, Toufexis D, Fair DA, Sanchez MM. Effects of social subordination and oestradiol on resting-state amygdala functional connectivity in adult female rhesus monkeys. J Neuroendocrinol 2020; 32:e12822. [PMID: 31846515 PMCID: PMC7066536 DOI: 10.1111/jne.12822] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 10/17/2019] [Accepted: 12/12/2019] [Indexed: 12/18/2022]
Abstract
Preclinical studies demonstrate that chronic stress modulates the effects of oestradiol (E2) on behaviour through the modification of the amygdala and the medial prefrontal cortex (mPFC) neuronal structure. Clinical studies suggest that alterations in amygdala functional connectivity (FC) with the mPFC may be associated with stress-related phenotypes, including mood and anxiety disorders. Thus, identifying the effects of stress and E2 on amygdala-mPFC circuits is critical for understanding the neurobiology underpinning the vulnerability to stress-related disorders in women. In the present study, we used a well-validated rhesus monkey model of chronic psychosocial stress (subordinate social rank) to examine effects of E2 on subordinate (SUB) (i.e. high stress) and dominant (DOM) (i.e. low stress) female resting-state amygdala FC with the mPFC and with the whole-brain. In the non-E2 treatment control condition, SUB was associated with stronger left amygdala FC to subgenual cingulate (Brodmann area [BA] 25: BA25), a region implicated in several psychopathologies in people. In SUB females, E2 treatment strengthened right amygdala-BA25 FC, induced a net positive amygdala-visual cortex FC that was positively associated with frequency of submissive behaviours, and weakened positive amygdala-para/hippocampus FC. Our findings show that subordinate social rank alters amygdala FC and the impact of E2 on amygdala FC with BA25 and with regions involved in visual processing and memory encoding.
Collapse
Affiliation(s)
- Katherine M. Reding
- Division of Developmental and Cognitive Neuroscience,
Yerkes National Primate Research Center, Emory University
| | - David S. Grayson
- Center for Neuroscience, University of California –
Davis
- Departments of Behavioral Neuroscience, Psychiatry, and
Advanced Imaging Research Center, Oregon Health and Science University
| | - Oscar Miranda-Dominguez
- Departments of Behavioral Neuroscience, Psychiatry, and
Advanced Imaging Research Center, Oregon Health and Science University
| | - Siddarth Ray
- Departments of Behavioral Neuroscience, Psychiatry, and
Advanced Imaging Research Center, Oregon Health and Science University
| | - Mark E. Wilson
- Division of Developmental and Cognitive Neuroscience,
Yerkes National Primate Research Center, Emory University
- Department of Psychiatry & Behavioral Sciences, Emory
University
| | - Donna Toufexis
- Department of Psychological Science, University of
Vermont
| | - Damien A. Fair
- Departments of Behavioral Neuroscience, Psychiatry, and
Advanced Imaging Research Center, Oregon Health and Science University
| | - Mar M. Sanchez
- Division of Developmental and Cognitive Neuroscience,
Yerkes National Primate Research Center, Emory University
- Department of Psychiatry & Behavioral Sciences, Emory
University
| |
Collapse
|
47
|
Sobolewski M, Abston K, Conrad K, Marvin E, Harvey K, Susiarjo M, Cory-Slechta DA. Lineage- and Sex-Dependent Behavioral and Biochemical Transgenerational Consequences of Developmental Exposure to Lead, Prenatal Stress, and Combined Lead and Prenatal Stress in Mice. ENVIRONMENTAL HEALTH PERSPECTIVES 2020; 128:27001. [PMID: 32073883 PMCID: PMC7064322 DOI: 10.1289/ehp4977] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 01/02/2020] [Accepted: 01/06/2020] [Indexed: 05/04/2023]
Abstract
BACKGROUND Lead (Pb) exposure and prenatal stress (PS) during development are co-occurring risk factors with shared biological substrates. PS has been associated with transgenerational passage of altered behavioral phenotypes, whereas the transgenerational behavioral or biochemical consequences of Pb exposure, and modification of any such effects by PS, is unknown. OBJECTIVES The present study sought to determine whether Pb, PS, or combined Pb and PS exposures produced adverse transgenerational consequences on brain and behavior. METHODS Maternal Pb and PS exposures were carried out in F0 mice. Outside breeders were used at each subsequent breeding, producing four F1-F2 lineages: [F1 female-F2 female (FF), FM (male), MF, and MM]. F3 offspring were generated from each of these lineages and examined for outcomes previously found to be altered by Pb, PS, or combined Pb and PS in F1 offspring: behavioral performance [fixed-interval (FI) schedule of food reward, locomotor activity, and anxiety-like behavior], dopamine function [striatal expression of tyrosine hydroxylase (Th)], glucocorticoid receptor (GR) and plasma corticosterone, as well as brain-derived neurotrophic factor (BDNF) and total percent DNA methylation of Th and Bdnf genes in the frontal cortex and hippocampus. RESULTS Maternal F0 Pb exposure produced runting in F3 offspring. Considered across lineages, F3 females exhibited Pb-related alterations in behavior, striatal BDNF levels, frontal cortical Th total percentage DNA methylation levels and serum corticosterone levels, whereas F3 males showed Pb- and PS-related alterations in behavior and total percent DNA methylation of hippocampal Bdnf. However, numerous lineage-specific effects were observed, most of greater magnitude than those observed across lineages, with outcomes differing by F3 sex. DISCUSSION These findings support the possibility that exposures of previous generations to Pb or PS may influence the brain and behavior of future generations. Observed changes were sex-dependent, with F3 females showing multiple changes through Pb-exposed lineages. Lineage effects may occur through maternal responses to pregnancy, altered maternal behavior, epigenetic modifications, or a combination of mechanisms, but they have significant public health ramifications regardless of mechanism. https://doi.org/10.1289/EHP4977.
Collapse
Affiliation(s)
- Marissa Sobolewski
- Department of Environmental Medicine, University of Rochester School of Medicine, Rochester, New York, USA
| | - Kadijah Abston
- Department of Environmental Medicine, University of Rochester School of Medicine, Rochester, New York, USA
| | - Katherine Conrad
- Department of Environmental Medicine, University of Rochester School of Medicine, Rochester, New York, USA
| | - Elena Marvin
- Department of Environmental Medicine, University of Rochester School of Medicine, Rochester, New York, USA
| | - Katherine Harvey
- Department of Environmental Medicine, University of Rochester School of Medicine, Rochester, New York, USA
| | - Martha Susiarjo
- Department of Environmental Medicine, University of Rochester School of Medicine, Rochester, New York, USA
| | - Deborah A Cory-Slechta
- Department of Environmental Medicine, University of Rochester School of Medicine, Rochester, New York, USA
| |
Collapse
|
48
|
Cui L, Wang F, Yin Z, Chang M, Song Y, Wei Y, Lv J, Zhang Y, Tang Y, Gong X, Xu K. Effects of the LHPP gene polymorphism on the functional and structural changes of gray matter in major depressive disorder. Quant Imaging Med Surg 2020; 10:257-268. [PMID: 31956547 DOI: 10.21037/qims.2019.12.01] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Background A single-nucleotide polymorphism (SNP) of the LHPP gene (rs35936514) has been reported to be associated with major depressive disorder (MDD) in genome-wide association studies. However, the systems-level neural effects of rs35936514 that mediate the association are unknown. We hypothesized that variations in rs35936514 would be associated with structural and functional changes in gray matter (GM) at rest in MDD patients. Methods A total of 50 MDD patients and 113 healthy controls (HCs) were studied. Functional connectivity (FC) was analyzed by defining the bilateral hippocampus as the seed region. Voxel-based morphometry (VBM) was performed to assess the patterns of GM volume. The subjects were further divided into two groups: a CC homozygous group (CC; 24 MDD and 56 HC) and a risk T-allele carrier group (CT/TT genotypes; 26 MDD and 57 HC). A 2×2 analysis of variance (ANOVA: diagnosis × genotype) was used to determine the interaction effects and main effect (P<0.05). Results Significant diagnosis × genotype interaction effects on brain morphology and FC were noted. Compared to other subgroups, the MDD patients with the T allele showed an increased hippocampal FC in the bilateral calcarine cortex and cuneus and a decreased hippocampal FC in the right dorsolateral prefrontal cortex (DLPFC), bilateral anterior cingulate cortex (ACC), and medial prefrontal cortex (MPFC), in addition to reduced GM volume in the right DLPFC, bilateral temporal cortex, and posterior cingulate cortex (PCC). Conclusions LHPP gene polymorphisms may affect functional and structural changes in the GM at rest and may play an important role in the pathophysiological mechanisms of MDD.
Collapse
Affiliation(s)
- Lingling Cui
- Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang 110001, China.,State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Fei Wang
- Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang 110001, China.,Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang 110001, China.,Brain Function Research Sections, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Zhiyang Yin
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang 110001, China.,Brain Function Research Sections, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Miao Chang
- Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang 110001, China.,Brain Function Research Sections, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Yanzhuo Song
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang 110001, China.,Brain Function Research Sections, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Yange Wei
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang 110001, China.,Brain Function Research Sections, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Jing Lv
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang 110001, China.,Brain Function Research Sections, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Yifan Zhang
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang 110001, China.,Brain Function Research Sections, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Yanqing Tang
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang 110001, China.,Brain Function Research Sections, The First Affiliated Hospital of China Medical University, Shenyang 110001, China.,Department of Geriatrics, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Xiaohong Gong
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Ke Xu
- Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| |
Collapse
|
49
|
Sigurdardottir HL, Lanzenberger R, Kranz GS. Genetics of sex differences in neuroanatomy and function. HANDBOOK OF CLINICAL NEUROLOGY 2020; 175:179-193. [PMID: 33008524 DOI: 10.1016/b978-0-444-64123-6.00013-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2023]
Abstract
Sex differences are observed at many distinct biologic levels, such as in the anatomy and functioning of the brain, behavior, and susceptibility to neuropsychiatric disorders. Previously, these differences were believed to entirely result from the secretion of gonadal hormones; however, recent research has demonstrated that differences are also the consequence of direct or nonhormonal effects of genes located on the sex chromosomes. This chapter reviews the four core genotype model that separates the effects of hormones and sex chromosomes and highlights a few genes that are believed to be partly responsible for sex dimorphism of the brain, in particular, the Sry gene. Genetics of the brain's neurochemistry is discussed and the susceptibility to certain neurologic and psychiatric disorders is reviewed. Lastly, we discuss the sex-specific genetic contribution in disorders of sexual development. The precise molecular mechanisms underlying these differences are currently not entirely known. An increased knowledge and understanding of the role of candidate genes will undeniably be of great aid in elucidating the molecular basis of sex-biased disorders and potentially allow for more sex-specific therapies.
Collapse
Affiliation(s)
- Helen L Sigurdardottir
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria.
| | - Rupert Lanzenberger
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Georg S Kranz
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria; Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong, People's Republic of China; The State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, People's Republic of China
| |
Collapse
|
50
|
Gildawie KR, Honeycutt JA, Brenhouse HC. Region-specific Effects of Maternal Separation on Perineuronal Net and Parvalbumin-expressing Interneuron Formation in Male and Female Rats. Neuroscience 2019; 428:23-37. [PMID: 31887358 DOI: 10.1016/j.neuroscience.2019.12.010] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 12/02/2019] [Accepted: 12/08/2019] [Indexed: 12/12/2022]
Abstract
Early life experiences play a vital role in contributing to healthy brain development. Adverse experiences have a lasting impact on the prefrontal cortex (PFC) and basolateral amygdala (BLA), brain regions associated with emotion regulation. Early life adversity via maternal separation (MS) has sex-specific effects on expression of parvalbumin (PV), which is expressed in fast-spiking GABAergic interneurons that are preferentially enwrapped by perineuronal nets (PNNs). Importantly, PNN formation coincides with the closure of developmental critical periods and regulates PV-expressing interneuron activity. Since aberrant PNN organization has been reported following adverse experiences in adolescent and adult rats, we investigated the impact of adversity early in life in the form of MS on the developing brain. Rat pups were separated from their dams for 4 h per day from postnatal day (P) 2-20. Tissue sections from juvenile (P20), adolescent (P40), and early adult (P70) animals containing the PFC and BLA were fluorescently stained to visualize Wisteria floribunda agglutinin+ PNNs and PV-expressing interneurons, and density and intensity was quantified. Our results confirm past reports that PFC PNNs form gradually throughout development; however, PNN density plateaus in adolescence, while intensity continues to increase into adulthood. Importantly, MS delays PNN formation in the prelimbic PFC and results in sex-specific aberrations in PNN structural integrity that do not appear until adulthood. The present findings reveal sex-, age-, and region-specific effects of early life adversity on PNN and PV maturation, implicating neuroplastic alterations following early life adversity that may be associated with sex differences in psychopathology and resilience.
Collapse
Affiliation(s)
- Kelsea R Gildawie
- Department of Psychology, Developmental Neuropsychobiology Laboratory, Northeastern University, Boston, MA 02115, USA
| | - Jennifer A Honeycutt
- Department of Psychology, Developmental Neuropsychobiology Laboratory, Northeastern University, Boston, MA 02115, USA
| | - Heather C Brenhouse
- Department of Psychology, Developmental Neuropsychobiology Laboratory, Northeastern University, Boston, MA 02115, USA.
| |
Collapse
|