1
|
Akhtar K, Rashid G, Rather TB, Maqbool I, Parveiz I, Bhat GA, Parray FQ, Yasin SB, Mudassar S. Downregulation of Hedgehog Interacting Protein (HHIP) in Gastric Cancer: Implications for Tumorigenesis. Indian J Clin Biochem 2025; 40:46-58. [PMID: 39835242 PMCID: PMC11741958 DOI: 10.1007/s12291-024-01293-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 12/17/2024] [Indexed: 01/22/2025]
Abstract
The increasing incidence of gastric cancer (GC) in the Kashmir Valley is concerning, but its root causes are largely unknown. Dysregulated activation of the Hedgehog signaling pathway has been linked to various cancers, and the Human Hedgehog Interacting Protein (HHIP), a tumor suppressor, is frequently dysregulated in malignancies. However, the expression of the HHIP gene in GC is inconsistent and poorly understood. This study aimed to examine HHIP gene expression in gastric cancer. We used methylation-specific PCR, Western Blot analysis, and quantitative reverse transcription PCR (qRT-PCR) to assess the hypermethylation and expression levels of HHIP gene promoters. The correlation between these results and clinical parameters (e.g. age, gender, histological type, class, stage, and lymph node metastasis) was studied with samples from 53 GC patients confirmed by histology. In 69.81% (37 out of 53) of the tumor tissue, HHIP hypermethylation was found. Of the 45 cases examined for mRNA expression, 53.33% (24 out of 45) showed a decrease in the HHIP mRNA level compared to the normal sample. In addition, 49.05% (26 out of 53) showed a decline in the expression of HHIP proteins. Almost all GC samples with reduced protein expression also showed a reduction in mRNA levels. These results suggest that the hypermethylation of the HHIP promoter leads to a decrease in the regulation of HHIP, which contributes to the activation of the hedgehog signal path and may play a critical role in the progress of GC. Our study highlights the significant link between HHIP hypermethylation and reduced gene expression at both mRNA and protein levels, suggesting that target HHIP gene methylation could be a promising treatment strategy for gastric cancer.
Collapse
Affiliation(s)
- Kulsum Akhtar
- Department of Clinical Biochemistry, SKIMS, Srinagar, India
| | - Gowhar Rashid
- Department of Clinical Biochemistry, SKIMS, Srinagar, India
| | | | - Irfan Maqbool
- Department of Clinical Biochemistry, Government Degree College, Baramulla, India
| | - Ishrat Parveiz
- Department of Clinical Biochemistry, SKIMS, Srinagar, India
| | | | - Fazl Q. Parray
- Department of General and Invasive Surgery, SKIMS, Srinagar, India
| | | | - Syed Mudassar
- Department of Clinical Biochemistry, SKIMS, Srinagar, India
| |
Collapse
|
2
|
Sykes DJ, Solanki S, Chukkapalli S, Williams K, Newman EA, Resnicow K, Shah YM. Structural enrichment attenuates colitis-associated colon cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.13.580099. [PMID: 38405737 PMCID: PMC10888747 DOI: 10.1101/2024.02.13.580099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Colorectal cancer (CRC) is a major public health concern and disproportionately impacts racial/ethnic minority populations in the US. Animal models are helpful in examining human health disparities because many stress-induced human health conditions can be recapitulated using mouse models. Azoxymethane (AOM)/ dextran sodium sulfate (DSS) treatment can be used to model colitis-associated cancers. While colitis-associated cancers account for only 2% of colon cancers, the AOM/DSS model is useful for examining links between inflammation, immunity, and colon cancer. Mice were housed in enriched and impoverished environments for 1-month prior to behavioral testing. Following behavioral testing the mice were subjected to the AOM/DSS model. While our analysis revealed no significant behavioral variances between the impoverished and enriched housing conditions, we found significant effects in tumorigenesis. Enriched mice had fewer tumors and smaller tumor volumes compared to impoverished mice. African Americans are at higher risk for early onset colorectal cancers in part due to social economic status. Furthermore, housing conditions and environment may reflect social economic status. Research aimed at understanding links between social economic status and colorectal cancer progression is important for eliminating disparities in health outcomes.
Collapse
|
3
|
McCallum RT, Thériault RK, Manduca JD, Russell ISB, Culmer AM, Doost JS, Martino TA, Perreault ML. Nrf2 activation rescues stress-induced depression-like behaviour and inflammatory responses in male but not female rats. Biol Sex Differ 2024; 15:16. [PMID: 38350966 PMCID: PMC10863247 DOI: 10.1186/s13293-024-00589-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 01/31/2024] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND Major depressive disorder (MDD) is a recurring affective disorder that is two times more prevalent in females than males. Evidence supports immune system dysfunction as a major contributing factor to MDD, notably in a sexually dimorphic manner. Nuclear factor erythroid 2-related factor 2 (Nrf2), a regulator of antioxidant signalling during inflammation, is dysregulated in many chronic inflammatory disorders; however, its role in depression and the associated sex differences have yet to be explored. Here, we investigated the sex-specific antidepressant and immunomodulatory effects of the potent Nrf2 activator dimethyl fumarate (DMF), as well as the associated gene expression profiles. METHODS Male and female rats were treated with vehicle or DMF (25 mg/kg) whilst subjected to 8 weeks of chronic unpredictable stress. The effect of DMF treatment on stress-induced depression- and anxiety-like behaviours, as well as deficits in recognition and spatial learning and memory were then assessed. Sex differences in hippocampal (HIP) microglial activation and gene expression response were also evaluated. RESULTS DMF treatment during stress exposure had antidepressant effects in male but not female rats, with no anxiolytic effects in either sex. Recognition learning and memory and spatial learning and memory were impaired in chronically stressed males and females, respectively, and DMF treatment rescued these deficits. DMF treatment also prevented stress-induced HIP microglial activation in males. Conversely, females displayed no HIP microglial activation associated with stress exposure. Last, chronic stress elicited sex-specific alterations in HIP gene expression, many of which were normalized in animals treated with DMF. Of note, most of the differentially expressed genes in males normalized by DMF were related to antioxidant, inflammatory or immune responses. CONCLUSIONS Collectively, these findings support a greater role of immune processes in males than females in a rodent model of depression. This suggests that pharmacotherapies that target Nrf2 have the potential to be an effective sex-specific treatment for depression.
Collapse
Affiliation(s)
- Ryan T McCallum
- Department of Biomedical Sciences, University of Guelph, 50 Stone Rd. E., Guelph, ON, N1G 2W1, Canada
| | - Rachel-Karson Thériault
- Department of Biomedical Sciences, University of Guelph, 50 Stone Rd. E., Guelph, ON, N1G 2W1, Canada
| | - Joshua D Manduca
- Department of Biomedical Sciences, University of Guelph, 50 Stone Rd. E., Guelph, ON, N1G 2W1, Canada
| | - Isaac S B Russell
- Department of Biomedical Sciences, University of Guelph, 50 Stone Rd. E., Guelph, ON, N1G 2W1, Canada
| | - Angel M Culmer
- Department of Biomedical Sciences, University of Guelph, 50 Stone Rd. E., Guelph, ON, N1G 2W1, Canada
| | - Janan Shoja Doost
- Department of Biomedical Sciences, University of Guelph, 50 Stone Rd. E., Guelph, ON, N1G 2W1, Canada
| | - Tami A Martino
- Department of Biomedical Sciences, University of Guelph, 50 Stone Rd. E., Guelph, ON, N1G 2W1, Canada
| | - Melissa L Perreault
- Department of Biomedical Sciences, University of Guelph, 50 Stone Rd. E., Guelph, ON, N1G 2W1, Canada.
| |
Collapse
|
4
|
Guez-Barber D, Eisch AJ, Cristancho AG. Developmental Brain Injury and Social Determinants of Health: Opportunities to Combine Preclinical Models for Mechanistic Insights into Recovery. Dev Neurosci 2023; 45:255-267. [PMID: 37080174 PMCID: PMC10614252 DOI: 10.1159/000530745] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 04/14/2023] [Indexed: 04/22/2023] Open
Abstract
Epidemiological studies show that social determinants of health are among the strongest factors associated with developmental outcomes after prenatal and perinatal brain injuries, even when controlling for the severity of the initial injury. Elevated socioeconomic status and a higher level of parental education correlate with improved neurologic function after premature birth. Conversely, children experiencing early life adversity have worse outcomes after developmental brain injuries. Animal models have provided vital insight into mechanisms perturbed by developmental brain injuries, which have indicated directions for novel therapeutics or interventions. Animal models have also been used to learn how social environments affect brain maturation through enriched environments and early adverse conditions. We recognize animal models cannot fully recapitulate human social circumstances. However, we posit that mechanistic studies combining models of developmental brain injuries and early life social environments will provide insight into pathways important for recovery. Some studies combining enriched environments with neonatal hypoxic injury models have shown improvements in developmental outcomes, but further studies are needed to understand the mechanisms underlying these improvements. By contrast, there have been more limited studies of the effects of adverse conditions on developmental brain injury extent and recovery. Uncovering the biological underpinnings for early life social experiences has translational relevance, enabling the development of novel strategies to improve outcomes through lifelong treatment. With the emergence of new technologies to analyze subtle molecular and behavioral phenotypes, here we discuss the opportunities for combining animal models of developmental brain injury with social construct models to deconvolute the complex interactions between injury, recovery, and social inequity.
Collapse
Affiliation(s)
- Danielle Guez-Barber
- Division of Child Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amelia J. Eisch
- Department of Anesthesiology and Critical Care, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ana G. Cristancho
- Division of Child Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
5
|
Clinton SM, Unroe KA, Shupe EA, McCoy CR, Glover ME. Resilience to Stress: Lessons from Rodents about Nature versus Nurture. Neuroscientist 2022; 28:283-298. [PMID: 33567987 PMCID: PMC11092422 DOI: 10.1177/1073858421989357] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Individual differences in human temperament influence how we respond to stress and can confer vulnerability (or resilience) to emotional disorders. For example, high levels of behavioral inhibition in children predict increased risk of mood and anxiety disorders in later life. The biological underpinnings of temperament are unknown, although improved understanding can offer insight into the pathogenesis of emotional disorders. Our laboratory has used a rat model of temperamental differences to study neurodevelopmental factors that lead to a highly inhibited, stress vulnerable phenotype. Selective breeding for high versus low behavioral response to novelty created two rat strains that exhibit dramatic behavior differences over multiple domains relevant to emotional disorders. Low novelty responder (bLR) rats exhibit high levels of behavioral inhibition, passive stress coping, anhedonia, decreased sociability and vulnerability to chronic stress compared to high novelty responders (bHRs). On the other hand, bHRs exhibit high levels of behavioral dis-inhibition, active coping, and aggression. This review article summarizes our work with the bHR/bLR model showing the developmental emergence of the bHR/bLR phenotypes, the role the environment plays in shaping it, and the involvement of epigenetic processes such as DNA methylation that mediate differences in emotionality and stress reactivity.
Collapse
Affiliation(s)
- Sarah M. Clinton
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| | - Keaton A. Unroe
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| | - Elizabeth A. Shupe
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| | - Chelsea R. McCoy
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| | - Matthew E. Glover
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, 24061, USA
| |
Collapse
|
6
|
Central and Peripheral Fatigue in Physical Exercise Explained: A Narrative Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19073909. [PMID: 35409591 PMCID: PMC8997532 DOI: 10.3390/ijerph19073909] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/16/2022] [Accepted: 03/21/2022] [Indexed: 02/07/2023]
Abstract
The study of the origin and implications of fatigue in exercise has been widely investigated, but not completely understood given the complex multifactorial mechanisms involved. Then, it is essential to understand the fatigue mechanism to help trainers and physicians to prescribe an adequate training load. The present narrative review aims to analyze the multifactorial factors of fatigue in physical exercise. To reach this aim, a consensus and critical review were performed using both primary sources, such as scientific articles, and secondary ones, such as bibliographic indexes, web pages, and databases. The main search engines were PubMed, SciELO, and Google Scholar. Central and peripheral fatigue are two unison constructs part of the Integrative Governor theory, in which both psychological and physiological drives and requirements are underpinned by homeostatic principles. The relative activity of each one is regulated by dynamic negative feedback activity, as the fundamental general operational controller. Fatigue is conditioned by factors such as gender, affecting men and women differently. Sleep deprivation or psychological disturbances caused, for example, by stress, can affect neural activation patterns, realigning them and slowing down simple mental operations in the context of fatigue. Then, fatigue can have different origins not only related with physiological factors. Therefore, all these prisms must be considered for future approaches from sport and clinical perspectives.
Collapse
|
7
|
Interspecific differences in sociability, social novelty preference, anxiety- and depression-like behaviors between Brandt's voles and C57BL/6J mice. Behav Processes 2022; 197:104624. [DOI: 10.1016/j.beproc.2022.104624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/08/2022] [Accepted: 03/10/2022] [Indexed: 11/24/2022]
|
8
|
Rainville JR, Lipuma T, Hodes GE. Translating the Transcriptome: Sex Differences in the Mechanisms of Depression and Stress, Revisited. Biol Psychiatry 2022; 91:25-35. [PMID: 33865609 PMCID: PMC10197090 DOI: 10.1016/j.biopsych.2021.02.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 02/01/2021] [Accepted: 02/01/2021] [Indexed: 12/28/2022]
Abstract
The past decade has produced a plethora of studies examining sex differences in the transcriptional profiles of stress and mood disorders. As we move forward from accepting the existence of extensive molecular sex differences in the brain to exploring the purpose of these sex differences, our approach must become more systemic and less reductionist. Earlier studies have examined specific brain regions and/or cell types. To use this knowledge to develop the next generation of personalized medicine, we need to comprehend how transcriptional changes across the brain and/or the body relate to each other. We provide an overview of the relationships between baseline and depression/stress-related transcriptional sex differences and explore contributions of preclinically identified mechanisms and their impacts on behavior.
Collapse
Affiliation(s)
- Jennifer R Rainville
- Department of Neuroscience, Virginia Polytechnic and State University, Blacksburg, Virginia
| | - Timothy Lipuma
- Department of Neuroscience, Virginia Polytechnic and State University, Blacksburg, Virginia
| | - Georgia E Hodes
- Department of Neuroscience, Virginia Polytechnic and State University, Blacksburg, Virginia.
| |
Collapse
|
9
|
Yu Q, Wang C, Xu H, Wu Y, Ding H, Liu N, Zhang N, Wang C. The mediating role of transmembrane protein 132D methylation in predicting the occurrence of panic disorder in physical abuse. Front Psychiatry 2022; 13:972522. [PMID: 36032246 PMCID: PMC9403743 DOI: 10.3389/fpsyt.2022.972522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 07/22/2022] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE Genome Wide Association study (GWAS) has revealed that the transmembrane protein 132D (TMEM132D) is a gene of sensitive for panic disorder (PD). As the main type of childhood trauma experience, childhood abuse has become a public health issue attracting much attention at home and abroad, and has been proved to be a risk factor for the onset of PD. However, how it affects the occurrence and development of panic disorder has not yet been revealed. We examined the relationship between TMEM132D methylation, childhood abuse and symptoms based on this finding. MATERIALS AND METHODS Thirty-two patients with PD and 22 healthy controls (HCs) were recruited after age, gender, and the education level were matched. The DNA methylation levels of CpG sites across the genome were examined with genomic DNA samples (PD, N = 32, controls, N = 22) extracted from subjects' elbow venous blood. A mediation model was used to explore the relationship between the methylation degree of different CpG sites and childhood maltreatment and clinical symptoms. RESULTS We found that the PD group had significantly lower methylation at CpG1, CpG2, CpG3, CpG4, CpG5, CpG6, CpG7, CpG8, CpG11, CpG14, and CpG18 than did the HCs (p < 0.05). The CpG2 (r = 0.5953, p = 0.0117) site in the priming region of TEME132D gene were positively associated with PDSS score. The CpG2 (r = 0.4889, p = 0.046) site in the priming region of TEME132D gene were positively associated with physical abuse. Furthermore, path analyses showed that the methylation of CpG2 of TMEM132D played a fully mediating role in the relationship between physical abuse and PD symptom severity (95. CONCLUSION Childhood abuse experiences, especially physical abuse, are significantly related to PD. The methylation of CpG2 of TMEM132D was shown to have a fully mediating effect between panic disorder and physical abuse. The interaction between TMEM132D methylation and physical abuse can predict panic disorder.
Collapse
Affiliation(s)
- Qianmei Yu
- School of Psychology, Nanjing Normal University, Nanjing, China.,Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Chiyue Wang
- Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Huazheng Xu
- Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Yun Wu
- School of Psychology, Nanjing Normal University, Nanjing, China.,Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Huachen Ding
- Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Na Liu
- School of Psychology, Nanjing Normal University, Nanjing, China.,Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Ning Zhang
- School of Psychology, Nanjing Normal University, Nanjing, China.,Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Chun Wang
- School of Psychology, Nanjing Normal University, Nanjing, China.,Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| |
Collapse
|
10
|
Borba LA, Broseghini LDR, Manosso LM, de Moura AB, Botelho MEM, Arent CO, Behenck JP, Hilsendeger A, Kammer LH, Valvassori SS, Quevedo J, Réus GZ. Environmental enrichment improves lifelong persistent behavioral and epigenetic changes induced by early-life stress. J Psychiatr Res 2021; 138:107-116. [PMID: 33848966 PMCID: PMC10494235 DOI: 10.1016/j.jpsychires.2021.04.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 03/26/2021] [Accepted: 04/02/2021] [Indexed: 02/08/2023]
Abstract
This study aimed to evaluate the effects of environmental enrichment (EE) in Wistar rats subjected to maternal deprivation (MD). MD was performed in the first post-natal days (PND) ten for 3 h/day. The groups were: control; deprived without EE; and deprived with EE. The EE was applied for 3 h/day. Forced swimming test (FST) and open field test were performed, and histone deacetylase (HDAC) and DNA methyltransferase (DNMT) activities in the prefrontal cortex (PFC) and hippocampus were evaluated on 31, 41, and 61 PND. MD altered spontaneous locomotor activity and immobility time in FST, but the effects were sex- and developmental period dependent. In deprived females at PND 31, 41, and 61, HDAC and DNMT increased in the PFC and hippocampus. In females exposed to EE for 20 days, there was a decrease of HDAC in the hippocampus and DNMT in the PFC and hippocampus. Exposure of females to EE for 40 days can reverse HDAC and DNMT increase in all brain areas. In deprived males at PND 31, 41, and 61, HDAC and DNMT increased in the hippocampus, and in the group exposed to EE for 40 days, there was a decrease in hippocampal activity. In PFC of male deprived rats at PND 61 and EE for 40 days, there was a reduction of HDAC and DNMT. MD induced lifelong persistent behavioral and epigenetic changes, and such effects were more evident in female than male rats. EE can be considered an essential non-pharmacological strategy to treat long-term trauma-induced early life changes.
Collapse
Affiliation(s)
- Laura A Borba
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciuma, SC, Brazil
| | - Lia D R Broseghini
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciuma, SC, Brazil
| | - Luana M Manosso
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciuma, SC, Brazil
| | - Airam B de Moura
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciuma, SC, Brazil
| | - Maria Eduarda M Botelho
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciuma, SC, Brazil
| | - Camila O Arent
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciuma, SC, Brazil
| | - João Paulo Behenck
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciuma, SC, Brazil
| | - Amanda Hilsendeger
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciuma, SC, Brazil
| | - Letícia H Kammer
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciuma, SC, Brazil
| | - Samira S Valvassori
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciuma, SC, Brazil
| | - João Quevedo
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciuma, SC, Brazil; Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA; Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA; Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Gislaine Z Réus
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciuma, SC, Brazil.
| |
Collapse
|
11
|
Alshammari TK. Sexual dimorphism in pre-clinical studies of depression. Prog Neuropsychopharmacol Biol Psychiatry 2021; 105:110120. [PMID: 33002519 DOI: 10.1016/j.pnpbp.2020.110120] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 09/17/2020] [Accepted: 09/25/2020] [Indexed: 12/21/2022]
Abstract
Although there is a sex bias in the pathological mechanisms exhibited by brain disorders, investigation of the female brain in biomedical science has long been neglected. Use of the male model has generally been the preferred option as the female animal model exhibits both biological variability and hormonal fluctuations. Existing studies that compare behavioral and/or molecular alterations in animal models of brain diseases are generally underrepresented, and most utilize the male model. Nevertheless, in recent years there has been a trend toward the increased inclusion of females in brain studies. However, current knowledge regarding sex-based differences in depression and stress-related disorders is limited. This can be improved by reviewing preclinical studies that highlight sex differences in depression. This paper therefore presents a review of sex-based preclinical studies of depression. These shed light on the discrepancies between males and females regarding the biological mechanisms that underpin mechanistic alterations in the diseased brain. This review also highlights the conclusions drawn by preclinical studies to advance our understanding of mood disorders, encouraging researchers to promote ways of investigating and managing sexually dimorphic disorders.
Collapse
Affiliation(s)
- Tahani K Alshammari
- Department of Pharmacology and Toxicology, Pharmacy College, King Saud University, Saudi Arabia; Prince Naïf Bin Abdul-Aziz Health Research Center, King Saud University, Saudi Arabia.
| |
Collapse
|
12
|
Thériault RK, Manduca JD, Perreault ML. Sex differences in innate and adaptive neural oscillatory patterns link resilience and susceptibility to chronic stress in rats. J Psychiatry Neurosci 2021; 46:E258-E270. [PMID: 33769022 PMCID: PMC8061734 DOI: 10.1503/jpn.200117] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Major depressive disorder is a chronic illness with a higher incidence in women. Dysregulated neural oscillatory activity is an emerging mechanism thought to underlie major depressive disorder, but whether sex differences in these rhythms contribute to the development of symptoms is unknown. METHODS We exposed male and female rats to chronic unpredictable stress and characterized them as stress-resilient or stress-susceptible based on behavioural output in the forced swim test and the sucrose preference test. To identify sex-specific neural oscillatory patterns associated with stress response, we recorded local field potentials from the prefrontal cortex, cingulate cortex, nucleus accumbens and dorsal hippocampus throughout stress exposure. RESULTS At baseline, female stress-resilient rats innately exhibited higher theta coherence in hippocampal connections compared with stress-susceptible female rats. Following stress exposure, additional oscillatory changes manifested: stress-resilient females were characterized by increased dorsal hippocampal theta power and cortical gamma power, and stress-resilient males were characterized by a widespread increase in high gamma coherence. In stress-susceptible animals, we observed a pattern of increased delta and reduced theta power; the changes were restricted to the cingulate cortex and dorsal hippocampus in males but occurred globally in females. Finally, stress exposure was accompanied by the time-dependent recruitment of specific neural pathways, which culminated in system-wide changes that temporally coincided with the onset of depression-like behaviour. LIMITATIONS We could not establish causality between the electrophysiological changes and behaviours with the methodology we employed. CONCLUSION Sex-specific neurophysiological patterns can function as early markers for stress vulnerability and the onset of depression-like behaviours in rats.
Collapse
Affiliation(s)
- Rachel-Karson Thériault
- From the department of Molecular and Cellular Biology, University of Guelph, Guelph, Ont., Canada (Thériault, Manduca, Perreault) and the Collaborative Neuroscience Program, University of Guelph, Guelph, Ont., Canada (Thériault, Perreault)
| | - Joshua D Manduca
- From the department of Molecular and Cellular Biology, University of Guelph, Guelph, Ont., Canada (Thériault, Manduca, Perreault) and the Collaborative Neuroscience Program, University of Guelph, Guelph, Ont., Canada (Thériault, Perreault)
| | - Melissa L Perreault
- From the department of Molecular and Cellular Biology, University of Guelph, Guelph, Ont., Canada (Thériault, Manduca, Perreault) and the Collaborative Neuroscience Program, University of Guelph, Guelph, Ont., Canada (Thériault, Perreault)
| |
Collapse
|
13
|
Powers B, Joyce C, Kleinman JE, Hyde TM, Ajilore O, Leow A, Sodhi MS. Sex differences in the transcription of glutamate transporters in major depression and suicide. J Affect Disord 2020; 277:244-252. [PMID: 32836031 DOI: 10.1016/j.jad.2020.07.055] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 05/29/2020] [Accepted: 07/05/2020] [Indexed: 11/28/2022]
Abstract
BACKGROUND Accumulating evidence indicates that the glutamate system contributes to the pathophysiology of major depressive disorder (MDD) and suicide. We previously reported higher mRNA expression of glutamate receptors in the dorsolateral prefrontal cortex (DLPFC) of females with MDD. METHODS In the current study, we measured the expression of mRNAs encoding glutamate transporters in the DLPFC of MDD subjects who died by suicide (MDD-S, n = 51), MDD non-suicide subjects (MDD-NS, n = 28), and individuals who did not have a history of neurological illness (CTRL, n = 32). RESULTS Females but not males with MDD showed higher expression of EAATs and VGLUTs relative to CTRLs. VGLUT expression was significantly higher in the female MDD-S group, relative to the other groups. EAAT expression was lower in the male violent suicides. LIMITATIONS This study has limitations common to most human studies, including medication history and demographic differences between the diagnostic groups. We mitigated the effects of confounders by including them as covariates in our analyses. CONCLUSIONS We report sex differences in the expression of glutamate transporter genes in the DLPFC in MDD. Increased neuronal glutamate transporter expression may increase synaptic glutamate, leading to neuronal and glial loss in the DLPFC in MDD. These deficits may lower DLPFC activity, impair problem solving and impair executive function in depression, perhaps increasing vulnerability to suicidal behavior. These data add to accumulating support for the hypothesis that glutamatergic transmission is dysregulated in MDD and suicide. Glutamate transporters may be novel targets for the development of rapidly acting antidepressant therapies.
Collapse
Affiliation(s)
- Brian Powers
- Department of Molecular Pharmacology & Neuroscience, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, United States
| | - Cara Joyce
- Biostatistics Collaborative Core, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States
| | - Joel E Kleinman
- Lieber Institute for Brain Development and Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Thomas M Hyde
- Lieber Institute for Brain Development and Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Johns Hopkins University School of Medicine, Department of Neurology, Baltimore, MD, United States
| | - Olusola Ajilore
- Dept. Psychiatry, University of Illinois at Chicago, Chicago, IL, United States
| | - Alex Leow
- Dept. Psychiatry, University of Illinois at Chicago, Chicago, IL, United States
| | - Monsheel S Sodhi
- Department of Molecular Pharmacology & Neuroscience, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, United States; Dept. Psychiatry, University of Illinois at Chicago, Chicago, IL, United States.
| |
Collapse
|
14
|
Muir J, Tse YC, Iyer ES, Biris J, Cvetkovska V, Lopez J, Bagot RC. Ventral Hippocampal Afferents to Nucleus Accumbens Encode Both Latent Vulnerability and Stress-Induced Susceptibility. Biol Psychiatry 2020; 88:843-854. [PMID: 32682566 DOI: 10.1016/j.biopsych.2020.05.021] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 04/25/2020] [Accepted: 05/13/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Stress is a major risk factor for depression, but not everyone responds to stress in the same way. Identifying why certain individuals are more susceptible is essential for targeted treatment and prevention. In rodents, nucleus accumbens (NAc) afferents from the ventral hippocampus (vHIP) are implicated in stress-induced susceptibility, but little is known about how this pathway might encode future vulnerability or specific behavioral phenotypes. METHODS We used fiber photometry to record in vivo activity in vHIP-NAc afferents during tests of depressive- and anxiety-like behavior in male and female mice, both before and after a sex-specific chronic variable stress protocol, to probe relationships between prestress neural activity and behavior and potential predictors of poststress behavioral adaptation. Furthermore, we examined chronic variable stress-induced alterations in vHIP-NAc activity in vivo and used ex vivo slice electrophysiology to identify the mechanism of this change. RESULTS We identified behavioral specificity of the vHIP-NAc pathway to anxiety-like and social interaction behavior. We also showed that this activity is broadly predictive of stress-induced susceptibility in both sexes, while prestress behavior is predictive only of anxiety-like behavior. We observed a stress-induced increase in in vivo vHIP-NAc activity coincident with an increase in spontaneous excitatory postsynaptic current frequency. CONCLUSIONS We implicate vHIP-NAc in social interaction and anxiety-like behavior and identify markers of vulnerability in this neural signal, with elevated prestress vHIP-NAc activity predicting increased susceptibility across behavioral domains. Our findings indicate that individual differences in neural activity and behavior play a role in predetermining susceptibility to later stress, providing insight into mechanisms of vulnerability.
Collapse
Affiliation(s)
- Jessie Muir
- Integrated Program in Neuroscience, McGill University, Montréal, Quebec, Canada
| | - Yiu Chung Tse
- Department of Psychology, McGill University, Montréal, Quebec, Canada
| | - Eshaan S Iyer
- Integrated Program in Neuroscience, McGill University, Montréal, Quebec, Canada
| | - Julia Biris
- Department of Psychology, McGill University, Montréal, Quebec, Canada
| | | | - Joëlle Lopez
- Department of Psychology, McGill University, Montréal, Quebec, Canada
| | - Rosemary C Bagot
- Department of Psychology, McGill University, Montréal, Quebec, Canada; Ludmer Centre for Neuroinformatics and Mental Health, Montréal, Quebec, Canada.
| |
Collapse
|
15
|
Abstract
Stress is associated with the onset of several stress-related mental disorders that occur more frequently in women than in men, such as major depression or posttraumatic stress disorder (PTSD). The hypothalamic-pituitary-adrenal (HPA) axis is the major component of the neuroendocrine network responding to internal and external challenges. The proper functioning of the HPA axis is critical for the maintenance of mental and physical health, as dysregulations of the HPA axis have been linked to several mental and physical disorders. Numerous studies have observed distinct sex differences in the regulation of the HPA axis in response to stress, and it is supposed that these differences may partially explain the female predominance in stress-related mental disorders. Preclinical models have clearly shown that the HPA axis in females is activated more rapidly and produces a larger output of stress hormones than in males. However, studies with humans often produced inconsistent findings, which might be traced back to the variation of investigated stressors, the use of contraceptives in some of the studies, and different menstrual cycle stages of the female subjects. This article discusses rodent and human literature of sex differences in the function of the HPA axis.
Collapse
Affiliation(s)
- Carolin Leistner
- Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - Andreas Menke
- Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital of Wuerzburg, Wuerzburg, Germany; Comprehensive Heart Failure Center, University Hospital of Wuerzburg, Wuerzburg, Germany; Department of Psychosomatic Medicine and Psychotherapy, Medical Park Chiemseeblick, Bernau-Felden, Germany.
| |
Collapse
|
16
|
Verma M, Agarwal N, Verma M, Kumar V. Epigenetics and animal models: applications in cancer control and treatment. Anim Biotechnol 2020. [DOI: 10.1016/b978-0-12-811710-1.00004-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
17
|
|
18
|
Kim GS, Smith AK, Xue F, Michopoulos V, Lori A, Armstrong DL, Aiello AE, Koenen KC, Galea S, Wildman DE, Uddin M. Methylomic profiles reveal sex-specific differences in leukocyte composition associated with post-traumatic stress disorder. Brain Behav Immun 2019; 81:280-291. [PMID: 31228611 PMCID: PMC6754791 DOI: 10.1016/j.bbi.2019.06.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 06/18/2019] [Accepted: 06/18/2019] [Indexed: 02/07/2023] Open
Abstract
Post-traumatic stress disorder (PTSD) is a debilitating mental disorder precipitated by trauma exposure. However, only some persons exposed to trauma develop PTSD. There are sex differences in risk; twice as many women as men develop a lifetime diagnosis of PTSD. Methylomic profiles derived from peripheral blood are well-suited for investigating PTSD because DNA methylation (DNAm) encodes individual response to trauma and may play a key role in the immune dysregulation characteristic of PTSD pathophysiology. In the current study, we leveraged recent methodological advances to investigate sex-specific differences in DNAm-based leukocyte composition that are associated with lifetime PTSD. We estimated leukocyte composition on a combined methylation array dataset (483 participants, ∼450 k CpG sites) consisting of two civilian cohorts, the Detroit Neighborhood Health Study and Grady Trauma Project. Sex-stratified Mann-Whitney U test and two-way ANCOVA revealed that lifetime PTSD was associated with significantly higher monocyte proportions in males, but not in females (Holm-adjusted p-val < 0.05). No difference in monocyte proportions was observed between current and remitted PTSD cases in males, suggesting that this sex-specific difference may reflect a long-standing trait of lifetime history of PTSD, rather than current state of PTSD. Associations with lifetime PTSD or PTSD status were not observed in any other leukocyte subtype and our finding in monocytes was confirmed using cell estimates based on a different deconvolution algorithm, suggesting that our sex-specific findings are robust across cell estimation approaches. Overall, our main finding of elevated monocyte proportions in males, but not in females with lifetime history of PTSD provides evidence for a sex-specific difference in peripheral blood leukocyte composition that is detectable in methylomic profiles and that may reflect long-standing changes associated with PTSD diagnosis.
Collapse
Affiliation(s)
- Grace S Kim
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Medical Scholars Program, University of Illinois College of Medicine, Urbana, IL, USA
| | - Alicia K Smith
- Department of Psychiatry & Behavioral Sciences, Emory University, Atlanta, GA, USA; Department of Gynecology and Obstetrics, Emory University, Atlanta, GA, USA
| | - Fei Xue
- Department of Statistics, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Vasiliki Michopoulos
- Department of Psychiatry & Behavioral Sciences, Emory University, Atlanta, GA, USA
| | - Adriana Lori
- Department of Psychiatry & Behavioral Sciences, Emory University, Atlanta, GA, USA
| | - Don L Armstrong
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Allison E Aiello
- Gillings School of Global Public Health, University of North Carolina - Chapel Hill, Chapel Hill, NC, USA
| | - Karestan C Koenen
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Sandro Galea
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| | - Derek E Wildman
- Genomics Program, College of Public Health, University of South Florida, Tampa, FL, USA
| | - Monica Uddin
- Genomics Program, College of Public Health, University of South Florida, Tampa, FL, USA.
| |
Collapse
|
19
|
Dioli C, Patrício P, Sousa N, Kokras N, Dalla C, Guerreiro S, Santos-Silva MA, Rego AC, Pinto L, Ferreiro E, Sotiropoulos I. Chronic stress triggers divergent dendritic alterations in immature neurons of the adult hippocampus, depending on their ultimate terminal fields. Transl Psychiatry 2019; 9:143. [PMID: 31028242 PMCID: PMC6486609 DOI: 10.1038/s41398-019-0477-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 02/02/2019] [Accepted: 03/08/2019] [Indexed: 12/18/2022] Open
Abstract
Chronic stress, a suggested precipitant of brain pathologies, such as depression and Alzheimer's disease, is known to impact on brain plasticity by causing neuronal remodeling as well as neurogenesis suppression in the adult hippocampus. Although many studies show that stressful conditions reduce the number of newborn neurons in the adult dentate gyrus (DG), little is known about whether and how stress impacts on dendritic development and structural maturation of these newborn neurons. We, herein, demonstrate that chronic stress impacts differentially on doublecortin (DCX)-positive immature neurons in distinct phases of maturation. Specifically, the density of the DCX-positive immature neurons whose dendritic tree reaches the inner molecular layer (IML) of DG is reduced in stressed animals, whereas their dendritic complexity is increased. On the contrary, no change on the density of DCX-positive neurons whose dendritic tree extends to the medial/outer molecular layer (M/OML) of the DG is found under stress conditions, whereas the dendritic complexity of these cells is diminished. In addition, DCX+ cells displayed a more complex and longer arbor in the dendritic compartments located in the granular cell layer of the DG under stress conditions; on the contrary, their dendritic segments localized into the M/OML were shorter and less complex. These findings suggest that the neuroplastic effects of chronic stress on dendritic maturation and complexity of DCX+ immature neurons vary based on the different maturation stage of DCX-positive cells and the different DG sublayer, highlighting the complex and dynamic stress-driven neuroplasticity of immature neurons in the adult hippocampus.
Collapse
Affiliation(s)
- Chrysoula Dioli
- 0000 0001 2159 175Xgrid.10328.38Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal ,0000 0001 2159 175Xgrid.10328.38ICVS/3B’s - PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| | - Patrícia Patrício
- 0000 0001 2159 175Xgrid.10328.38Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal ,0000 0001 2159 175Xgrid.10328.38ICVS/3B’s - PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| | - Nuno Sousa
- 0000 0001 2159 175Xgrid.10328.38Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal ,0000 0001 2159 175Xgrid.10328.38ICVS/3B’s - PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| | - Nikolaos Kokras
- 0000 0001 2155 0800grid.5216.0First Department of Psychiatry, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece ,0000 0001 2155 0800grid.5216.0Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Christina Dalla
- 0000 0001 2155 0800grid.5216.0Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Sara Guerreiro
- 0000 0001 2159 175Xgrid.10328.38Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal ,0000 0001 2159 175Xgrid.10328.38ICVS/3B’s - PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| | - Miguel A. Santos-Silva
- 0000 0001 2159 175Xgrid.10328.38Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal ,0000 0001 2159 175Xgrid.10328.38ICVS/3B’s - PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| | - Ana Cristina Rego
- 0000 0000 9511 4342grid.8051.cCenter for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal ,0000 0000 9511 4342grid.8051.cInstitute of Biochemistry, Faculty of Medicine, University of Coimbra (FMUC), Coimbra, Portugal
| | - Luísa Pinto
- 0000 0001 2159 175Xgrid.10328.38Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal ,0000 0001 2159 175Xgrid.10328.38ICVS/3B’s - PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| | - Elisabete Ferreiro
- 0000 0000 9511 4342grid.8051.cCenter for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal ,0000 0000 9511 4342grid.8051.cInstitute for Interdisciplinary Research of the University of Coimbra (IIIUC), Coimbra, Portugal
| | - Ioannis Sotiropoulos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal. .,ICVS/3B's - PT Government Associate Laboratory, 4710-057, Braga/Guimarães, Portugal.
| |
Collapse
|
20
|
H3K9 Acetylation of Tph2 Involved in Depression-like Behavior in Male, but not Female, Juvenile Offspring Rat Induced by Prenatal Stress. Neuroscience 2018; 381:138-148. [PMID: 29625215 DOI: 10.1016/j.neuroscience.2018.03.043] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 03/23/2018] [Accepted: 03/26/2018] [Indexed: 12/31/2022]
Abstract
Increasing evidence has shown that prenatal stress (PS) could cause depression-like behavior in the offspring, which is sex-specific. However, the underlying mechanisms remain to be elucidated. This study is to investigate the involvement of tryptophan hydroxylase 2 (Tph2) H3K9 acetylation (H3K9ac) modification on PS-induced depression-like behavior in juvenile offspring rats (JOR). PS models were established, with or without trichostatin A (TSA) treatment. Animal behavior was assessed by the sucrose preference test (SPT) and forced swimming test (FST). The mRNA and protein expression levels of TPH2 in the dorsal raphenucleus (DRN), hippocampus, and prefrontal cortex were detected with quantitative real-time PCR and Western blot analysis, respectively. The Tph2 H3K9ac levels in the hippocampus were also analyzed. SPT and FST showed significantly reduced sucrose preference and significantly prolonged immobility in PS-induced male juvenile offspring rats (MJOR). Moreover, the mRNA and protein expression levels of TPH2 in the DRN and hippocampus were significantly declined, while the hippocampal Tph2 H3K9ac levels were significantly declined in the PS-induced MJOR. Furthermore, the PS-induced effects in MJOR could be reversed by the microinjection of TSA. However, no significant effects were observed for the female juvenile offspring rats (FJORs). In conclusion, our results showed that the Tph2 H3K9ac modification is only involved in PS-induced depression-like behavior in MJOR, in a sex-specific manner. These findings might contribute to the understanding of the disease pathogenesis and clinical treatment in future.
Collapse
|
21
|
Kokras N, Dalla C. Preclinical sex differences in depression and antidepressant response: Implications for clinical research. J Neurosci Res 2017; 95:731-736. [PMID: 27870451 DOI: 10.1002/jnr.23861] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 06/29/2016] [Accepted: 07/11/2016] [Indexed: 01/11/2023]
Abstract
Women suffer from depression and anxiety disorders more often than men, and as a result they receive antidepressants to a greater extent. Sex differences in antidepressant response in humans have been modestly studied, and results have been controversial. At the same time, preclinical studies on animal models of depression and antidepressant response have provided insights with regard to sex differences that could be useful for the design and interpretation of future clinical trials. This Mini-Review discusses such sex-differentiated findings with regard to the presentation of depression, endophenotypes, and antidepressant response. In particular, men and women differ in symptomatology of depression, and animal models of depression have revealed sex differences in behavioral indices. However, although in experimental studies behavioral indices and models are adjusted to identify sex differences properly, this is not the case in the use of depression rating scales in clinical studies. Accordingly, preclinical studies highlight the importance of sex differences at the baseline behavioral response and the underlying mechanisms that often converge after antidepressant treatment. This is also a neglected issue in human studies. Finally, preclinical research suggests that, in the quest for potential biomarkers for depression, sex should be an important factor to consider. Careful consideration of sex differences in preclinical research could facilitate and ameliorate the design and quality of clinical studies for disease biomarkers and novel fast-acting antidepressants that are so essential for both men and women suffering from depression. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Nikolaos Kokras
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece.,First Department of Psychiatry, Eginition Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Christina Dalla
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
22
|
Epigenome-wide association study of DNA methylation in panic disorder. Clin Epigenetics 2017; 9:6. [PMID: 28149334 PMCID: PMC5270210 DOI: 10.1186/s13148-016-0307-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Accepted: 12/26/2016] [Indexed: 12/22/2022] Open
Abstract
Background Panic disorder (PD) is considered to be a multifactorial disorder emerging from interactions among multiple genetic and environmental factors. To date, although genetic studies reported several susceptibility genes with PD, few of them were replicated and the pathogenesis of PD remains to be clarified. Epigenetics is considered to play an important role in etiology of complex traits and diseases, and DNA methylation is one of the major forms of epigenetic modifications. In this study, we performed an epigenome-wide association study of PD using DNA methylation arrays so as to investigate the possibility that different levels of DNA methylation might be associated with PD. Methods The DNA methylation levels of CpG sites across the genome were examined with genomic DNA samples (PD, N = 48, control, N = 48) extracted from peripheral blood. Methylation arrays were used for the analysis. β values, which represent the levels of DNA methylation, were normalized via an appropriate pipeline. Then, β values were converted to M values via the logit transformation for epigenome-wide association study. The relationship between each DNA methylation site and PD was assessed by linear regression analysis with adjustments for the effects of leukocyte subsets. Results Forty CpG sites showed significant association with PD at 5% FDR correction, though the differences of the DNA methylation levels were relatively small. Most of the significant CpG sites (37/40 CpG sites) were located in or around CpG islands. Many of the significant CpG sites (27/40 CpG sites) were located upstream of genes, and all such CpG sites with the exception of two were hypomethylated in PD subjects. A pathway analysis on the genes annotated to the significant CpG sites identified several pathways, including “positive regulation of lymphocyte activation.” Conclusions Although future studies with larger number of samples are necessary to confirm the small DNA methylation abnormalities associated with PD, there is a possibility that several CpG sites might be associated, together as a group, with PD. Electronic supplementary material The online version of this article (doi:10.1186/s13148-016-0307-1) contains supplementary material, which is available to authorized users.
Collapse
|
23
|
Beeghly M, Partridge T, Tronick E, Muzik M, Mashhadi MR, Boeve JL, Irwin JL. ASSOCIATIONS BETWEEN EARLY MATERNAL DEPRESSIVE SYMPTOM TRAJECTORIES AND TODDLERS' FELT SECURITY AT 18 MONTHS: ARE BOYS AND GIRLS AT DIFFERENTIAL RISK? Infant Ment Health J 2017; 38:53-67. [PMID: 28042661 PMCID: PMC5225085 DOI: 10.1002/imhj.21617] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The goal of this study was to evaluate whether there are sex differences in children's vulnerability to caregiving risk, as indexed by trajectories of maternal depressive symptoms assessed from 2 to 18 months' postpartum, and children's rated attachment security in toddlerhood, adjusting for maternal social support and demographic risk. Analyses utilized longitudinal data collected for 182 African American mother-child dyads from economically diverse backgrounds. Participants were recruited at the time of the child's birth and followed to 18 months' postpartum. Results of conditional latent growth models indicated that an increasing rate of change in level of maternal depressive symptoms over time negatively predicted toddlers' felt attachment security. Higher social support was associated with decreasing levels of maternal depressive symptoms over time whereas higher demographic risk was associated with increasing levels of maternal depressive symptoms. A subsequent multigroup conditional latent growth model revealed that child sex moderated these associations. For male (but not female) children, a rapid increase in maternal depressive symptoms was associated with lower felt attachment security at 18 months. These findings suggest that boys, as compared to girls, may be more vulnerable to early caregiving risks such as maternal depression, with negative consequences for mother-child attachment security in toddlerhood.
Collapse
Affiliation(s)
| | | | | | - Maria Muzik
- University of Michigan Health Sciences Center, Ann Arbor MI
| | | | | | | |
Collapse
|
24
|
Immune and Neuroendocrine Mechanisms of Stress Vulnerability and Resilience. Neuropsychopharmacology 2017; 42:62-80. [PMID: 27291462 PMCID: PMC5143517 DOI: 10.1038/npp.2016.90] [Citation(s) in RCA: 245] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Revised: 05/17/2016] [Accepted: 05/18/2016] [Indexed: 12/15/2022]
Abstract
Diagnostic criteria for mood disorders including major depressive disorder (MDD) largely ignore biological factors in favor of behavioral symptoms. Compounding this paucity of psychiatric biomarkers is a need for therapeutics to adequately treat the 30-50% of MDD patients who are unresponsive to traditional antidepressant medications. Interestingly, MDD is highly prevalent in patients suffering from chronic inflammatory conditions, and MDD patients exhibit higher levels of circulating pro-inflammatory cytokines. Together, these clinical findings suggest a role for the immune system in vulnerability to stress-related psychiatric illness. A growing body of literature also implicates the immune system in stress resilience and coping. In this review, we discuss the mechanisms by which peripheral and central immune cells act on the brain to affect stress-related neurobiological and neuroendocrine responses. We specifically focus on the roles of pro-inflammatory cytokine signaling, peripheral monocyte infiltration, microglial activation, and hypothalamic-pituitary-adrenal axis hyperactivity in stress vulnerability. We also highlight recent evidence suggesting that adaptive immune responses and treatment with immune modulators (exogenous glucocorticoids, humanized antibodies against cytokines) may decrease depressive symptoms and thus represent an attractive alternative to the current antidepressant treatments.
Collapse
|
25
|
Sarkar A, Kabbaj M. Sex Differences in Effects of Ketamine on Behavior, Spine Density, and Synaptic Proteins in Socially Isolated Rats. Biol Psychiatry 2016; 80:448-456. [PMID: 26957131 PMCID: PMC4940294 DOI: 10.1016/j.biopsych.2015.12.025] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 12/18/2015] [Accepted: 12/30/2015] [Indexed: 01/12/2023]
Abstract
BACKGROUND The mechanistic underpinnings of sex differences in occurrence of depression and efficacy of antidepressant treatments are poorly understood. We examined the effects of isolation stress (IS) and the fast-acting antidepressant ketamine on anhedonia and depression-like behavior, spine density, and synaptic proteins in male and female rats. METHODS We used a chronic social IS paradigm to test the effects of ketamine (0, 2.5 mg/kg, and 5 mg/kg) on behavior and levels of synaptic proteins synapsin-1, postsynaptic density protein 95, and glutamate receptor 1 in male rats and female rats in diestrus. Medial prefrontal cortex spine density was also examined in male rats and female rats that received ketamine during either the diestrus or the proestrus phase of their estrous cycle. RESULTS Male rats showed anhedonia and depression-like behavior after 8 weeks of IS, concomitant with decreases in spine density and levels of synapsin-1, postsynaptic density protein 95, and glutamate receptor 1 in the medial prefrontal cortex; these changes were reversed by a single injection of ketamine (5 mg/kg). After 11 weeks of IS, female rats showed depression-like behavior but no signs of anhedonia. Although both doses of ketamine rescued depression-like behavior in female rats, the decline observed in synaptic proteins and spine density in IS and in diestrus female rats could not be reversed by ketamine. Spine density was higher in female rats during proestrus than in diestrus. CONCLUSIONS Our findings implicate a role for synaptic proteins synapsin-1, postsynaptic density protein 95, and glutamate receptor 1 and medial prefrontal cortex spine density in the antidepressant effects of ketamine in male rats subjected to IS but not in female rats subjected to IS, suggesting dissimilar underlying mechanisms for efficacy of ketamine in the two sexes.
Collapse
Affiliation(s)
- Ambalika Sarkar
- Department of Biomedical Sciences, College of Medicine, Program in Neuroscience, Florida State University, Tallahassee, Florida, USA
| | - Mohamed Kabbaj
- Department of Biomedical Sciences, Program in Neuroscience, College of Medicine, Florida State University, Tallahassee, Florida.
| |
Collapse
|
26
|
Carroll ME, Lynch WJ. How to study sex differences in addiction using animal models. Addict Biol 2016; 21:1007-29. [PMID: 27345022 PMCID: PMC4970981 DOI: 10.1111/adb.12400] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 02/17/2016] [Accepted: 02/27/2016] [Indexed: 11/30/2022]
Abstract
The importance of studying sex as a biological variable in biomedical research is becoming increasingly apparent. There is a particular need in preclinical studies of addiction to include both sexes, as female animals are often excluded from studies, leaving large gaps in our knowledge of not only sex differences and potential prevention and treatment strategies but also with regard to the basic neurobiology of addiction. This review focuses on methodology that has been developed in preclinical studies to examine sex differences in the behavioral aspects and neurobiological mechanisms related to addiction across the full range of the addiction process, including initiation (acquisition), maintenance, escalation, withdrawal, relapse to drug seeking and treatment. This review also discusses strategic and technical issues that need to be considered when comparing females and males, including the role of ovarian hormones and how sex differences interact with other major vulnerability factors in addiction, such as impulsivity, compulsivity and age (adolescent versus adult). Novel treatments for addiction are also discussed, such as competing non-drug rewards, repurposed medications such as progesterone and treatment combinations. Practical aspects of conducting research comparing female and male animals are also considered. Making sex differences a point of examination requires additional effort and consideration; however, such studies are necessary given mounting evidence demonstrating that the addiction process occurs differently in males and females. These studies should lead to a better understanding of individual differences in the development of addiction and effective treatments for males and females.
Collapse
Affiliation(s)
- Marilyn E. Carroll
- Marilyn E. Carroll, Department of Psychiatry, University of Minnesota, MMC 392, Minneapolis, MN, USA
| | - Wendy J. Lynch
- Wendy J. Lynch, Department of Psychiatry and Neurobehavioral Sciences, University of Virginia, PO Box 801402, Charlottesville, VA
| |
Collapse
|
27
|
Barra de la Tremblaye P, Plamondon H. Alterations in the corticotropin-releasing hormone (CRH) neurocircuitry: Insights into post stroke functional impairments. Front Neuroendocrinol 2016; 42:53-75. [PMID: 27455847 DOI: 10.1016/j.yfrne.2016.07.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 07/04/2016] [Accepted: 07/06/2016] [Indexed: 10/21/2022]
Abstract
Although it is well accepted that changes in the regulation of the hypothalamic-pituitary adrenal (HPA) axis may increase susceptibility to affective disorders in the general population, this link has been less examined in stroke patients. Yet, the bidirectional association between depression and cardiovascular disease is strong, and stress increases vulnerability to stroke. Corticotropin-releasing hormone (CRH) is the central stress hormone of the HPA axis pathway and acts by binding to CRH receptors (CRHR) 1 and 2, which are located in several stress-related brain regions. Evidence from clinical and animal studies suggests a role for CRH in the neurobiological basis of depression and ischemic brain injury. Given its importance in the regulation of the neuroendocrine, autonomic, and behavioral correlates of adaptation and maladaptation to stress, CRH is likely associated in the pathophysiology of post stroke emotional impairments. The goals of this review article are to examine the clinical and experimental data describing (1) that CRH regulates the molecular signaling brain circuit underlying anxiety- and depression-like behaviors, (2) the influence of CRH and other stress markers in the pathophysiology of post stroke emotional and cognitive impairments, and (3) context and site specific interactions of CRH and BDNF as a basis for the development of novel therapeutic targets. This review addresses how the production and release of the neuropeptide CRH within the various regions of the mesocorticolimbic system influences emotional and cognitive behaviors with a look into its role in psychiatric disorders post stroke.
Collapse
Affiliation(s)
- P Barra de la Tremblaye
- School of Psychology, Behavioral Neuroscience Program, University of Ottawa, 136 Jean-Jacques Lussier, Vanier Building, Ottawa, Ontario K1N 6N5, Canada
| | - H Plamondon
- School of Psychology, Behavioral Neuroscience Program, University of Ottawa, 136 Jean-Jacques Lussier, Vanier Building, Ottawa, Ontario K1N 6N5, Canada.
| |
Collapse
|
28
|
Reis-Silva TM, Cohn DW, Sandini TM, Udo MS, Teodorov E, Bernardi MM. Prenatal lipopolysaccharide exposure affects sexual dimorphism in different germlines of mice with a depressive phenotype. Life Sci 2016; 149:129-37. [DOI: 10.1016/j.lfs.2016.02.068] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 02/14/2016] [Accepted: 02/16/2016] [Indexed: 12/21/2022]
|
29
|
Boyce WT, Kobor MS. Development and the epigenome: the 'synapse' of gene-environment interplay. Dev Sci 2015; 18:1-23. [PMID: 25546559 DOI: 10.1111/desc.12282] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 10/14/2014] [Indexed: 12/21/2022]
Abstract
This paper argues that there is a revolution afoot in the developmental science of gene-environment interplay. We summarize, for an audience of developmental researchers and clinicians, how epigenetic processes - chromatin structural modifications that regulate gene expression without changing DNA sequences - may offer a strong, parsimonious account for the convergence of genetic and contextual variation in the genesis of adaptive and maladaptive development. Epigenetic processes may play a plausible explanatory role in understanding: divergent trajectories and sexual dimorphisms in brain development; statistical interactions between genes and environments; the biological embedding of early psychosocial adversities; the linkages of such adversities to disorders of mental health; the striking individual variation in the strength of those linkages; the molecular origins of critical and sensitive periods; and the transgenerational inheritance of risk and protection. Taken together, these arguments converge in a claim that epigenetic processes constitute a promising and illuminating point of connection - a 'synapse' - between genes and environments.
Collapse
Affiliation(s)
- W Thomas Boyce
- Departments of Pediatrics and Psychiatry, University of California, San Francisco, USA; Child and Brain Development Program, Canadian Institute for Advanced Research, Canada
| | | |
Collapse
|
30
|
Shimada-Sugimoto M, Otowa T, Hettema JM. Genetics of anxiety disorders: Genetic epidemiological and molecular studies in humans. Psychiatry Clin Neurosci 2015; 69:388-401. [PMID: 25762210 DOI: 10.1111/pcn.12291] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/09/2015] [Indexed: 12/30/2022]
Abstract
This review provides a broad overview of the state of research in the genetics of anxiety disorders (AD). Genetic epidemiological studies report a moderate level of familial aggregation (odds ratio: 4-6) and heritability estimates are about 30-50%. Twin studies suggest that the genetic architecture of AD is not isomorphic with their classifications, sharing risk factors with each other. So far, linkage and association studies of AD have produced inconclusive results. Genome-wide association studies of AD can provide an unbiased survey of common genetic variations across the entire genome. Given the shared causes of AD that transcend our current diagnostic classifications, clustering anxiety phenotypes into broader groups may be a powerful approach to identifying susceptibility locus for AD. Using such a shared genetic risk factor, meta-analyses of genome-wide association studies of AD conducted by large consortia are needed. Environmental factors also make a substantial contribution to the cause of AD. Although candidate gene studies of gene by environmental (G × E) interaction have appeared recently, no genome-wide search for G × E interactions have been performed. Epigenetic modification of DNA appears to have important effects on gene expression mediating environmental influences on disease risk. Given that G × E can be linked to an epigenetic modification, a combination analysis of genome-wide G × E interaction and methylation could be an alternative method to find risk variants for AD. This genetic research will enable us to utilize more effective strategies for the prevention and treatment of AD in the near future.
Collapse
Affiliation(s)
- Mihoko Shimada-Sugimoto
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takeshi Otowa
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - John M Hettema
- Department of Psychiatry, Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, USA
| |
Collapse
|
31
|
Ménard C, Hodes GE, Russo SJ. Pathogenesis of depression: Insights from human and rodent studies. Neuroscience 2015; 321:138-162. [PMID: 26037806 DOI: 10.1016/j.neuroscience.2015.05.053] [Citation(s) in RCA: 391] [Impact Index Per Article: 39.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 05/14/2015] [Accepted: 05/21/2015] [Indexed: 12/30/2022]
Abstract
Major depressive disorder (MDD) will affect one out of every five people in their lifetime and is the leading cause of disability worldwide. Nevertheless, mechanisms associated with the pathogenesis of MDD have yet to be completely understood and current treatments remain ineffective in a large subset of patients. In this review, we summarize the most recent discoveries and insights for which parallel findings have been obtained in human depressed subjects and rodent models of mood disorders in order to examine the potential etiology of depression. These mechanisms range from synaptic plasticity mechanisms to epigenetics and the immune system where there is strong evidence to support a functional role in the development of specific depression symptomology. Ultimately we conclude by discussing how novel therapeutic strategies targeting central and peripheral processes might ultimately aid in the development of effective new treatments for MDD and related stress disorders.
Collapse
Affiliation(s)
- C Ménard
- Fishberg Department of Neuroscience and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - G E Hodes
- Fishberg Department of Neuroscience and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - S J Russo
- Fishberg Department of Neuroscience and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
32
|
Effects of prenatal and postnatal depression, and maternal stroking, at the glucocorticoid receptor gene. Transl Psychiatry 2015; 5:e560. [PMID: 25942041 PMCID: PMC4471282 DOI: 10.1038/tp.2014.140] [Citation(s) in RCA: 129] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 10/20/2014] [Accepted: 11/17/2014] [Indexed: 12/21/2022] Open
Abstract
In animal models, prenatal and postnatal stress is associated with elevated hypothalamic-pituitary axis (HPA) reactivity mediated via altered glucocorticoid receptor (GR) gene expression. Postnatal tactile stimulation is associated with reduced HPA reactivity mediated via increased GR gene expression. In this first study in humans to examine the joint effects of prenatal and postnatal environmental exposures, we report that GR gene (NR3C1) 1-F promoter methylation in infants is elevated in the presence of increased maternal postnatal depression following low prenatal depression, and that this effect is reversed by self-reported stroking of the infants by their mothers over the first weeks of life.
Collapse
|
33
|
Epigenetic and transgenerational reprogramming of brain development. Nat Rev Neurosci 2015; 16:332-44. [PMID: 25921815 DOI: 10.1038/nrn3818] [Citation(s) in RCA: 338] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Neurodevelopmental programming - the implementation of the genetic and epigenetic blueprints that guide and coordinate normal brain development - requires tight regulation of transcriptional processes. During prenatal and postnatal time periods, epigenetic processes fine-tune neurodevelopment towards an end product that determines how an organism interacts with and responds to exposures and experiences throughout life. Epigenetic processes also have the ability to reprogramme the epigenome in response to environmental challenges, such as maternal stress, making the organism more or less adaptive depending on the future challenges presented. Epigenetic marks generated within germ cells as a result of environmental influences throughout life can also shape future generations long before conception occurs.
Collapse
|
34
|
Zerres K, Eggermann T. [Genetics and epigenetics. Explanatory approaches for (gender-specific) mechanisms of disease development]. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2015; 57:1047-53. [PMID: 25070408 DOI: 10.1007/s00103-014-2013-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Whereas the central role of DNA as the carrier of genetic information has long been well known, the impact of epigenetic mechanisms as mediators between genes and environment is now becoming increasingly clear. Epigenetics helps explain the partially reversible interplay between gene function and environment and even permits observation of the transgenerational transmission of epigenetic modifications. Of special interest are gender-specific mechanisms of gene regulation which, among others, offer an explanation for gender differences in human diseases. Since the study of epigenetic mechanisms and their impact on the etiology of common diseases is in its infancy, it is too early to draw general conclusions from the current state of knowledge. Moreover, completely new strategies are needed to research these effects. In addition to molecular findings, definitions of specific phenotypes are required, including biographic data of affected individuals and their ancestors. Epigenetics needs to be viewed in the context of the theory of evolution, classical genetics, and environmental research. Its aim is not to substitute the knowledge in these disciplines, but rather to provide a key to link their findings, thereby opening up new possibilities in terms of interpretation and understanding of gender differences in medicine. If these epigenetic mechanisms are better understood, particularly in terms of specific diseases, it is conceivable that these disorders could be influenced and treated in a more targeted manner in the future.
Collapse
Affiliation(s)
- K Zerres
- Institut für Humangenetik, Universitätsklinikum der RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Deutschland,
| | | |
Collapse
|
35
|
Gelman PL, Flores-Ramos M, López-Martínez M, Fuentes CC, Grajeda JPR. Hypothalamic-pituitary-adrenal axis function during perinatal depression. Neurosci Bull 2015; 31:338-50. [PMID: 25732527 DOI: 10.1007/s12264-014-1508-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2014] [Accepted: 09/24/2014] [Indexed: 12/01/2022] Open
Abstract
Abnormal function of the hypothalamic-pituitary-adrenal (HPA) axis is an important pathological finding in pregnant women exhibiting major depressive disorder. They show high levels of cortisol pro-inflammatory cytokines, hypothalamic-pituitary peptide hormones and catecholamines, along with low dehydroepiandrosterone levels in plasma. During pregnancy, the TH2 balance together with the immune system and placental factors play crucial roles in the development of the fetal allograft to full term. These factors, when altered, may generate a persistent dysfunction of the HPA axis that may lead to an overt transfer of cortisol and toxicity to the fetus at the expense of reduced activity of placental 11β-hydroxysteroid dehydrogenase type 2. Epigenetic modifications also may contribute to the dysregulation of the HPA axis. Affective disorders in pregnant women should be taken seriously, and therapies focused on preventing the deleterious effects of stressors should be implemented to promote the welfare of both mother and baby.
Collapse
|
36
|
Baldwin CH, Srivastava LK. Can the neurodevelopmental theory account for sex differences in schizophrenia across the life span? J Psychiatry Neurosci 2015; 40:75-7. [PMID: 25703644 PMCID: PMC4354820 DOI: 10.1503/jpn.150014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Affiliation(s)
| | - Lalit K. Srivastava
- Correspondence to: L.K. Srivastava, Douglas Mental Health University Institute and Department of Psychiatry, McGill University, 6875 LaSalle Boulevard, Montréal, Qc, Canada, H4H 1R3;
| |
Collapse
|
37
|
Abstract
Viable new treatments for depression and anxiety have been slow to emerge, likely owing to the complex and incompletely understood etiology of these disorders. A budding area of research with great therapeutic promise involves the study of resilience, the adaptive maintenance of normal physiology and behavior despite exposure to marked psychological stress. This phenomenon, documented in both humans and animal models, involves coordinated biological mechanisms in numerous bodily systems, both peripheral and central. In this review, we provide an overview of resilience mechanisms throughout the body, discussing current research in animal models investigating the roles of the neuroendocrine, immune, and central nervous systems in behavioral resilience to stress.
Collapse
Affiliation(s)
- Madeline L Pfau
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY, USA, 10029 ; Graduate Program in Neuroscience, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY, USA, 10029
| | - Scott J Russo
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY, USA, 10029 ; Graduate Program in Neuroscience, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY, USA, 10029
| |
Collapse
|
38
|
Does Child Sex Moderate Vulnerability to Postpartum Risk among Infants of Mothers at Risk for Psychopathology? INFANCY 2014. [DOI: 10.1111/infa.12065] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
39
|
Jones NC, O'Brien TJ, Carmant L. Interaction between sex and early-life stress: influence on epileptogenesis and epilepsy comorbidities. Neurobiol Dis 2014; 72 Pt B:233-41. [PMID: 25266701 DOI: 10.1016/j.nbd.2014.09.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 09/08/2014] [Accepted: 09/12/2014] [Indexed: 12/22/2022] Open
Abstract
Epilepsy is a common brain disorder which is characterised by recurring seizures. In addition to suffering from the constant stress of living with this neurological condition, patients also frequently experience comorbid psychiatric and cognitive disorders which significantly impact their quality of life. There is growing appreciation that stress, in particular occurring in early life, can negatively impact brain development, creating an enduring vulnerability to develop epilepsy. This aligns with the solid connections between early life environments and the development of psychiatric conditions, promoting the possibility that adverse early life events could represent a common risk factor for the later development of both epilepsy and comorbid psychiatric disorders. The influence of sex has been little studied, but recent research points to potential important interactions, particularly with regard to effects mediated by HPA axis programming. Understanding these interactions, and the underlying molecular mechanisms, will provide important new insights into the causation of both epilepsy and of psychiatric disorders, and potentially open up novel avenues for treatment.
Collapse
Affiliation(s)
- Nigel C Jones
- Department of Medicine (Royal Melbourne Hospital), Melbourne Brain Centre, University of Melbourne, Parkville, VIC, Australia.
| | - Terence J O'Brien
- Department of Medicine (Royal Melbourne Hospital), Melbourne Brain Centre, University of Melbourne, Parkville, VIC, Australia
| | - Lionel Carmant
- Centre de Recherche du Centre Hospitalier Universitaire Sainte-Justine, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
40
|
Song Y, Zuo Y. Occurrence of HHIP gene CpG island methylation in gastric cancer. Oncol Lett 2014; 8:2340-2344. [PMID: 25295121 PMCID: PMC4186611 DOI: 10.3892/ol.2014.2518] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2014] [Accepted: 08/07/2014] [Indexed: 11/06/2022] Open
Abstract
The present study aimed to observe the methylation status of the CpG islands at the human hedgehog interacting protein (HHIP) gene in gastric cancer tissues, peritumoral tissues and the AGS cell line, to analyze the association between the methylation status of the CpG islands and the tumorigenesis of gastric cancer. The HHIP mRNA expression in 60 human gastric carcinnoma tissues, peritumoral tissues and the gastric carcinoma AGS cell line were detected by reverse transcription polymerase chain reaction (RT-PCR). The HHIP methylation status of the promoter region in the gastric carcinnoma tissues and peritumoral tissues was detected by methylation-specific PCR (MSP). Prior to and following treatment with methyl transferase inhibitor 5-aza-2'-deoxycitydine (5-aza-dc), the HHIP mRNA expression level, the methylation status of the promoter region and the methylation site loci on the CpG islands in the AGS cells were detected by RT-PCR, MSP and bisulfite sequencing PCR (BSP), respectively. The correlation between the methylation status of the CpG islands at the HHIP promoter region and the HHIP mRNA expression level were analyzed. It was found that the expression level of the HHIP mRNA in the gastric carcinoma tissues was significantly lower than that in the adjacent tissues (0.82±0.38 vs. 1.60±0.26, respectively; P<0.001). No significant correlations were observed between the expression of HHIP mRNA and age, gender, tumor-node-metastasis stage, differentiation degree and presence of lymph node metastasis (P>0.05). The degree of methylation of the HHIP gene promotor in the peritumoral tissues (17.7±3.59%) was significantly lower than that in the gastric cancer tissues (62.9±6.14%) and in the AGS cells (99.7±0.67%) (P<0.05). Compared with prior to 5-aza-dc intervention, the HHIP mRNA expression level in the AGS cells was significantly increased subsequent to intervention (0.21±0.12 vs. 4.68±0.22; P<0.01), while the degree of methylation in the AGS cells was significantly decreased (90.2±0.67 vs. 10.1±0.21%; P<0.01), and the methylation sites in CpG islands were significantly reduced. The degree of HHIP methylation showed a negative correlation with the level of mRNA expression (r=-0.693; P<0.01). It can be hypothesized that a high degree of methylation of the HHIP gene promoter CpG islands in gastric cancer tissues and cells causes a decrease in HHIP mRNA expression, which may be involved in the carcinogenesis of gastric cancer.
Collapse
Affiliation(s)
- Yu Song
- Department of Oncology, Zhangjiagang First People's Hospital, Suzhou, Jiangsu 215600, P.R. China
| | - Yun Zuo
- Department of Oncology, Zhangjiagang First People's Hospital, Suzhou, Jiangsu 215600, P.R. China
| |
Collapse
|
41
|
Shen EY, Ahern TH, Cheung I, Straubhaar J, Dincer A, Houston I, de Vries GJ, Akbarian S, Forger NG. Epigenetics and sex differences in the brain: A genome-wide comparison of histone-3 lysine-4 trimethylation (H3K4me3) in male and female mice. Exp Neurol 2014; 268:21-9. [PMID: 25131640 DOI: 10.1016/j.expneurol.2014.08.006] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 07/30/2014] [Accepted: 08/05/2014] [Indexed: 12/11/2022]
Abstract
Many neurological and psychiatric disorders exhibit gender disparities, and sex differences in the brain likely explain some of these effects. Recent work in rodents points to a role for epigenetics in the development or maintenance of neural sex differences, although genome-wide studies have so far been lacking. Here we review the existing literature on epigenetics and brain sexual differentiation and present preliminary analyses on the genome-wide distribution of histone-3 lysine-4 trimethylation in a sexually dimorphic brain region in male and female mice. H3K4me3 is a histone mark primarily organized as 'peaks' surrounding the transcription start site of active genes. We microdissected the bed nucleus of the stria terminalis and preoptic area (BNST/POA) in adult male and female mice and used ChIP-Seq to compare the distribution of H3K4me3 throughout the genome. We found 248 genes and loci with a significant sex difference in H3K4me3. Of these, the majority (71%) had larger H3K4me3 peaks in females. Comparisons with existing databases indicate that genes and loci with increased H3K4me3 in females are associated with synaptic function and with expression atlases from related brain areas. Based on RT-PCR, only a minority of genes with a sex difference in H3K4me3 has detectable sex differences in expression at baseline conditions. Together with previous findings, our data suggest that there may be sex biases in the use of epigenetic marks. Such biases could underlie sex differences in vulnerabilities to drugs or diseases that disrupt specific epigenetic processes.
Collapse
Affiliation(s)
- Erica Y Shen
- Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Todd H Ahern
- Center for Behavioral Neuroscience, Department of Psychology, Quinnipiac University, Hamden, CT 06518, USA
| | - Iris Cheung
- Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, MA 01604, USA
| | - Juerg Straubhaar
- Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, MA 01604, USA
| | - Aslihan Dincer
- Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Genetics and Genomics Science, Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Isaac Houston
- Brudnick Neuropsychiatric Research Institute, University of Massachusetts Medical School, Worcester, MA 01604, USA
| | - Geert J de Vries
- Neuroscience Institute, Georgia State University, Atlanta, GA 30302, USA
| | - Schahram Akbarian
- Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Nancy G Forger
- Neuroscience Institute, Georgia State University, Atlanta, GA 30302, USA.
| |
Collapse
|
42
|
Al Ameri M, Al Mansouri S, Al Maamari A, Bahi A. The histone deacetylase (HDAC) inhibitor valproic acid reduces ethanol consumption and ethanol-conditioned place preference in rats. Brain Res 2014; 1583:122-31. [PMID: 25108044 DOI: 10.1016/j.brainres.2014.07.051] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 07/17/2014] [Accepted: 07/30/2014] [Indexed: 12/25/2022]
Abstract
Recent evidence suggests that epigenetic mechanisms such as chromatin modification (specifically histone acetylation) may play a crucial role in the development of addictive behavior. However, little is known about the role of epigenetic modifications in the rewarding properties of ethanol. In the current study, we studied the effects of systemic injection of the histone deacetylase (HDAC) inhibitor, valproic acid (VPA) on ethanol consumption and ethanol-elicited conditioned place preference (CPP). The effect of VPA (300 mg/kg) on voluntary ethanol intake and preference was assessed using continuous two-bottle choice procedure with escalating concentrations of alcohol (2.5-20% v/v escalating over 4 weeks). Taste sensitivity was studies using saccharin (sweet; 0.03% and 0.06%) and quinine (bitter; 20 µM and 40 µM) tastants solutions. Ethanol conditioned reward was investigated using an unbiased CPP model. Blood ethanol concentration (BEC) was also measured. Compared to vehicle, VPA-injected rats displayed significantly lower preference and consumption of ethanol in a two-bottle choice paradigm, with no significant difference observed with saccharin and quinine. More importantly, 0.5 g/kg ethanol-induced-CPP acquisition was blocked following VPA administration. Finally, vehicle- and VPA-treated mice had similar BECs. Taken together, our results implicated HDAC inhibition in the behavioral and reinforcement-related effects of alcohol and raise the question of whether specific drugs that target HDAC could potentially help to tackle alcoholism in humans.
Collapse
Affiliation(s)
- Mouza Al Ameri
- Department of Anatomy, College of Medicine & Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, UAE
| | - Shamma Al Mansouri
- Department of Anatomy, College of Medicine & Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, UAE
| | - Alyazia Al Maamari
- Department of Anatomy, College of Medicine & Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, UAE
| | - Amine Bahi
- Department of Anatomy, College of Medicine & Health Sciences, United Arab Emirates University, P.O. Box 17666, Al Ain, UAE.
| |
Collapse
|
43
|
Kokras N, Dalla C. Sex differences in animal models of psychiatric disorders. Br J Pharmacol 2014; 171:4595-619. [PMID: 24697577 DOI: 10.1111/bph.12710] [Citation(s) in RCA: 263] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Revised: 03/20/2014] [Accepted: 03/26/2014] [Indexed: 12/14/2022] Open
Abstract
Psychiatric disorders are characterized by sex differences in their prevalence, symptomatology and treatment response. Animal models have been widely employed for the investigation of the neurobiology of such disorders and the discovery of new treatments. However, mostly male animals have been used in preclinical pharmacological studies. In this review, we highlight the need for the inclusion of both male and female animals in experimental studies aiming at gender-oriented prevention, diagnosis and treatment of psychiatric disorders. We present behavioural findings on sex differences from animal models of depression, anxiety, post-traumatic stress disorder, substance-related disorders, obsessive-compulsive disorder, schizophrenia, bipolar disorder and autism. Moreover, when available, we include studies conducted across different stages of the oestrous cycle. By inspection of the relevant literature, it is obvious that robust sex differences exist in models of all psychiatric disorders. However, many times results are conflicting, and no clear conclusion regarding the direction of sex differences and the effect of the oestrous cycle is drawn. Moreover, there is a lack of considerable amount of studies using psychiatric drugs in both male and female animals, in order to evaluate the differential response between the two sexes. Notably, while in most cases animal models successfully mimic drug response in both sexes, test parameters and treatment-sensitive behavioural indices are not always the same for male and female rodents. Thus, there is an increasing need to validate animal models for both sexes and use standard procedures across different laboratories.
Collapse
Affiliation(s)
- N Kokras
- Department of Pharmacology, Medical School, University of Athens, Greece; First Department of Psychiatry, Eginition Hospital, Medical School, University of Athens, Greece
| | | |
Collapse
|
44
|
Davies MN, Krause L, Bell JT, Gao F, Ward KJ, Wu H, Lu H, Liu Y, Tsai PC, Collier DA, Murphy T, Dempster E, Mill J, Battle A, Mostafavi S, Zhu X, Henders A, Byrne E, Wray NR, Martin NG, Spector TD, Wang J. Hypermethylation in the ZBTB20 gene is associated with major depressive disorder. Genome Biol 2014; 15:R56. [PMID: 24694013 PMCID: PMC4072999 DOI: 10.1186/gb-2014-15-4-r56] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 04/02/2014] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Although genetic variation is believed to contribute to an individual's susceptibility to major depressive disorder, genome-wide association studies have not yet identified associations that could explain the full etiology of the disease. Epigenetics is increasingly believed to play a major role in the development of common clinical phenotypes, including major depressive disorder. RESULTS Genome-wide MeDIP-Sequencing was carried out on a total of 50 monozygotic twin pairs from the UK and Australia that are discordant for depression. We show that major depressive disorder is associated with significant hypermethylation within the coding region of ZBTB20, and is replicated in an independent cohort of 356 unrelated case-control individuals. The twins with major depressive disorder also show increased global variation in methylation in comparison with their unaffected co-twins. ZBTB20 plays an essential role in the specification of the Cornu Ammonis-1 field identity in the developing hippocampus, a region previously implicated in the development of major depressive disorder. CONCLUSIONS Our results suggest that aberrant methylation profiles affecting the hippocampus are associated with major depressive disorder and show the potential of the epigenetic twin model in neuro-psychiatric disease.
Collapse
|
45
|
Hunter SK. Sex differences in human fatigability: mechanisms and insight to physiological responses. Acta Physiol (Oxf) 2014; 210:768-89. [PMID: 24433272 DOI: 10.1111/apha.12234] [Citation(s) in RCA: 351] [Impact Index Per Article: 31.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 07/29/2013] [Accepted: 01/08/2014] [Indexed: 12/17/2022]
Abstract
Sex-related differences in physiology and anatomy are responsible for profound differences in neuromuscular performance and fatigability between men and women. Women are usually less fatigable than men for similar intensity isometric fatiguing contractions. This sex difference in fatigability, however, is task specific because different neuromuscular sites will be stressed when the requirements of the task are altered, and the stress on these sites can differ for men and women. Task variables that can alter the sex difference in fatigability include the type, intensity and speed of contraction, the muscle group assessed and the environmental conditions. Physiological mechanisms that are responsible for sex-based differences in fatigability may include activation of the motor neurone pool from cortical and subcortical regions, synaptic inputs to the motor neurone pool via activation of metabolically sensitive small afferent fibres in the muscle, muscle perfusion and skeletal muscle metabolism and fibre type properties. Non-physiological factors such as the sex bias of studying more males than females in human and animal experiments can also mask a true understanding of the magnitude and mechanisms of sex-based differences in physiology and fatigability. Despite recent developments, there is a tremendous lack of understanding of sex differences in neuromuscular function and fatigability, the prevailing mechanisms and the functional consequences. This review emphasizes the need to understand sex-based differences in fatigability to shed light on the benefits and limitations that fatigability can exert for men and women during daily tasks, exercise performance, training and rehabilitation in both health and disease.
Collapse
Affiliation(s)
- S. K. Hunter
- Exercise Science Program; Department of Physical Therapy; Marquette University; Milwaukee WI USA
| |
Collapse
|
46
|
McCarthy MM, Nugent BM. Epigenetic contributions to hormonally-mediated sexual differentiation of the brain. J Neuroendocrinol 2013; 25:1133-40. [PMID: 23919286 PMCID: PMC5330673 DOI: 10.1111/jne.12072] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Revised: 06/18/2013] [Accepted: 06/29/2013] [Indexed: 12/28/2022]
Abstract
It has been long established that hormones exert enduring influences on the developing brain that direct the reproductive response in adulthood, although the cellular mechanisms by which organisational effects are maintained have not been determined satisfactorily. Recent interest in epigenetic modifications to the nervous system has highlighted the potential for hormone-induced changes to the genome that could endure for the lifespan but not be transmitted to the next generation. Preliminary evidence suggests that this is indeed possible because sex differences in the histone code and in the methylation of CpGs in the promoters of specific genes have been identified and, at times, functionally correlated with behaviour. The present review provides an overview of epigenetic processes and discusses the current state-of-the-art, and also identifies future directions.
Collapse
Affiliation(s)
- M M McCarthy
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | | |
Collapse
|
47
|
Changes in the brain expression of alpha-2 subunits of the GABA-A receptor after chronic restraint stress in low- and high-anxiety rats. Behav Brain Res 2013; 253:337-45. [PMID: 23916758 DOI: 10.1016/j.bbr.2013.07.042] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 07/19/2013] [Accepted: 07/23/2013] [Indexed: 01/02/2023]
Abstract
This study assessed the mechanisms underlying the behavioral differences between high- (HR) and low-anxiety (LR) rats selected for their behavior in the contextual fear test (i.e., the duration of the freezing response was used as a discriminating variable). Rats were subjected to chronic restraint stress (21 days, 3h daily). We found that in the HR group, chronic restraint stress decreased rat activity in the Porsolt test and reduced the concentration of corticosterone in the prefrontal cortex. The behavioral changes were accompanied by a lower expression of alpha-2 GABA-A receptor subunits in the secondary motor cortex (M2 area) and in the dentate gyrus of the hippocampus (DG) compared to LR restraint animals. Moreover, restraint stress increased the density of alpha-2 GABA-A subunits in the basolateral amygdala (BLA) in HR rats and decreased the expression of these subunits in the DG and M2 areas compared to the HR control group. The present results suggest that, in HR rats exposed to chronic restraint stress, the function of hippocampal and cortical GABAergic neurotransmission is attenuated and that this effect could have important influences on the functioning of the hypothalamic-pituitary-adrenal axis and on depressive symptoms.
Collapse
|
48
|
Personalized medicine in Alzheimer's disease and depression. Contemp Clin Trials 2013; 36:616-23. [PMID: 23816492 DOI: 10.1016/j.cct.2013.06.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2012] [Revised: 06/14/2013] [Accepted: 06/20/2013] [Indexed: 12/17/2022]
Abstract
Latest research in the mental health field brings new hope to patients and promises to revolutionize the field of psychiatry. Personalized pharmacogenetic tests that aid in diagnosis and treatment choice are now becoming available for clinical practice. Amyloid beta peptide biomarkers in the cerebrospinal fluid of patients with Alzheimer's disease are now available. For the first time, radiologists are able to visualize amyloid plaques specific to Alzheimer's disease in live patients using Positron Emission Tomography-based tests approved by the FDA. A novel blood-based assay has been developed to aid in the diagnosis of depression based on activation of the HPA axis, metabolic, inflammatory and neurochemical pathways. Serotonin reuptake inhibitors have shown increased remission rates in specific ethnic subgroups and Cytochrome P450 gene polymorphisms can predict antidepressant tolerability. The latest research will help to eradicate "trial and error" prescription, ushering in the most personalized medicine to date. Like all major medical breakthroughs, integration of new algorithms and technologies requires sound science and time. But for many mentally ill patients, diagnosis and effective therapy cannot happen fast enough. This review will describe the newest diagnostic tests, treatments and clinical studies for the diagnosis and treatment of Alzheimer's disease and unipolar, major depressive disorder.
Collapse
Key Words
- 5-HTT
- 5-HTTLPR
- 5-Hydroxytryptamine Transporter gene
- AD
- ADNI
- ADRDA
- Alzheimer's Disease Neuroimaging Initiative
- Alzheimer's Disease and Related Disorders Association
- Alzheimer's disease
- Aβ40
- Aβ42
- CREB
- CSF
- CT
- CV
- CYP2C19
- CYP2D6
- CYP450
- Coefficient of Variation
- Computed Tomography
- Cytochrome P450
- Cytochrome P450 2C19
- Cytochrome P450 2D6
- DNA
- DSM
- DSM-IV-TR
- DSM-V
- Deoxyribonucleic Acid
- Depression
- Diagnostic and Statistical Manual of Mental Disorders
- Diagnostic and Statistical Manual of Mental Disorders—Fifth Edition
- Diagnostic and Statistical Manual of Mental Disorders—Fourth Edition-Text Revision
- ELISA
- Enzyme-Linked Immunosorbent Assay
- Epigenetics
- FDA
- FK506-binding protein
- FKBP5
- Food and Drug Administration
- GRIA
- GRIK
- HPA
- IL28RA
- KCNK2
- MDDScore
- MRI
- MTC
- Magnetic Resonance Imaging
- Major Depressive Disorder Score
- Methylthioninium Chloride
- NINCDS
- National Institute of Neurological and Communicative Disorders and Stroke
- P-tau181P
- PAPLN
- PET
- Personalized medicine
- Positron Emission Tomography
- QC
- Quality Control
- RDoC
- RNA
- Research Domain Criteria
- Ribonucleic Acid
- SSRI
- STAR*D
- Selective Serotonin Reuptake Inhibitor
- Sequenced Treatment Alternatives to Relieve Depression
- Serotonin-Transporter-Gene-Linked Polymorphic Region
- T-tau
- Tau phosphorylated at threonine 181
- VNTR
- WHO
- World Health Organization
- beta-amyloid, amino acids 1–40
- beta-amyloid, amino acids 1–42
- cAMP response element-binding protein
- cerebrospinal fluid
- glutamate receptor, ionotropic, AMPA
- glutamate receptor, ionotropic, kainate
- hypothalamic–pituitary–adrenal
- interleukin 28 receptor, alpha (interferon, lambda receptor)
- papilin, proteoglycan-like sulfated glycoprotein
- potassium channel, subfamily K, member 2
- total Tau
- variable nucleotide terminal repeat
Collapse
|