1
|
Shehzadi A, Bibi Z, Qadeer Sarwar M, Ullah A, Rehman A, Abbas Bukhari D. Effect of probiotics on hematological parameters of male and female Wistar rats. Saudi J Biol Sci 2023; 30:103794. [PMID: 37811481 PMCID: PMC10550764 DOI: 10.1016/j.sjbs.2023.103794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 08/15/2023] [Accepted: 08/27/2023] [Indexed: 10/10/2023] Open
Abstract
In the present study, the effect of probiotics on the hematology of Wistar rats was examined. Locally isolated Lactobacillus plantarum MZ707748 (Pro 1), L. plantarum MZ710117 (Pro 2), Weisella confusa MZ727611 (Pro 3), and L. plantarum MZ735961 (Pro 4) were used. One strain of probiotic, L. acidophilus-14 (Pro 5), was purchased commercially. Different groups were designed as G1, G2, G3, G4, and 5, G5/PC consisting only pro 5 and NC & 0 day were untreated. Different groups have different probiotics like G1 containing Pro 1 and Pro 2, G2 comprising Pro 3 and Pro 4, G3 containing Pro 2, Pro 3 and Pro 5, G4 having Pro 1-5, and G5 containing Pro 5. A complete count of blood, serum chemistry, fecal analysis, and histopathological examination of the thymus and liver were done. Statistical differences were seen in the complete blood count parameters (p < 0.05). No difference was observed in AST, ALT, bilirubin, albumin, IL-6, and IgA (p > 0.05) except for TP, creatinine, and globulin (p < 0.05). Fecal strains of probiotic groups were antibiotic-resistant. In males, Lactobacillus helveticus OQ152020, Enterococcus lactis OQ1519891, E. faecium OQ152017, L. gasseri OQ152017, and E. lactis OQ152019 were isolated from positive control, G1, G2, G3, and G4 respectively. In females, Enterococcus sp. OP800231, Limosilactobacillus fermentum OQ151985, E. lactis OP800267, L. plantarum OP800244, and E. faecium OQ151988 were isolated from positive control, G1, G2, G3 and G4, respectively. It was concluded that all probiotic strains were safe to use and had beneficial effects on the hematology of Wistar rats.
Collapse
Affiliation(s)
- Areeba Shehzadi
- Department of Zoology, Government College University, Lahore, Pakistan
| | - Zuhra Bibi
- Department of Zoology, Government College University, Lahore, Pakistan
| | | | - Arif Ullah
- Department of Zoology, Government College University, Lahore, Pakistan
| | - Abdul Rehman
- Institute of Microbiology and Molecular Genetics, University of the Punjab, Lahore, Pakistan
| | | |
Collapse
|
2
|
Kretzschmar A, Schülke JP, Masana M, Dürre K, Müller MB, Bausch AR, Rein T. The Stress-Inducible Protein DRR1 Exerts Distinct Effects on Actin Dynamics. Int J Mol Sci 2018; 19:ijms19123993. [PMID: 30545002 PMCID: PMC6321462 DOI: 10.3390/ijms19123993] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 12/07/2018] [Accepted: 12/10/2018] [Indexed: 12/13/2022] Open
Abstract
Cytoskeletal dynamics are pivotal to memory, learning, and stress physiology, and thus psychiatric diseases. Downregulated in renal cell carcinoma 1 (DRR1) protein was characterized as the link between stress, actin dynamics, neuronal function, and cognition. To elucidate the underlying molecular mechanisms, we undertook a domain analysis of DRR1 and probed the effects on actin binding, polymerization, and bundling, as well as on actin-dependent cellular processes. Methods: DRR1 domains were cloned and expressed as recombinant proteins to perform in vitro analysis of actin dynamics (binding, bundling, polymerization, and nucleation). Cellular actin-dependent processes were analyzed in transfected HeLa cells with fluorescence recovery after photobleaching (FRAP) and confocal microscopy. Results: DRR1 features an actin binding site at each terminus, separated by a coiled coil domain. DRR1 enhances actin bundling, the cellular F-actin content, and serum response factor (SRF)-dependent transcription, while it diminishes actin filament elongation, cell spreading, and actin treadmilling. We also provide evidence for a nucleation effect of DRR1. Blocking of pointed end elongation by addition of profilin indicates DRR1 as a novel barbed end capping factor. Conclusions: DRR1 impacts actin dynamics in several ways with implications for cytoskeletal dynamics in stress physiology and pathophysiology.
Collapse
Affiliation(s)
- Anja Kretzschmar
- Max Planck Institute of Psychiatry, Kraepelinstraße 2-10, 80805 München, Germany.
| | - Jan-Philip Schülke
- Max Planck Institute of Psychiatry, Kraepelinstraße 2-10, 80805 München, Germany.
| | - Mercè Masana
- Max Planck Institute of Psychiatry, Kraepelinstraße 2-10, 80805 München, Germany.
- Department of Psychiatry and Psychotherapy & Focus Program Translational Neuroscience, Johannes Gutenberg Universität Medical Center, 55131 Mainz, Germany.
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, IDIBAPS, CIBERNED, Casanova, 143, 08036 Barcelona, Spain.
| | - Katharina Dürre
- Lehrstuhl für Biophysik E27, Technische Universität München, Garching, Germany.
| | - Marianne B Müller
- Max Planck Institute of Psychiatry, Kraepelinstraße 2-10, 80805 München, Germany.
- Department of Psychiatry and Psychotherapy & Focus Program Translational Neuroscience, Johannes Gutenberg Universität Medical Center, 55131 Mainz, Germany.
| | - Andreas R Bausch
- Lehrstuhl für Biophysik E27, Technische Universität München, Garching, Germany.
| | - Theo Rein
- Max Planck Institute of Psychiatry, Kraepelinstraße 2-10, 80805 München, Germany.
| |
Collapse
|
3
|
Masana M, Westerholz S, Kretzschmar A, Treccani G, Liebl C, Santarelli S, Dournes C, Popoli M, Schmidt MV, Rein T, Müller MB. Expression and glucocorticoid-dependent regulation of the stress-inducible protein DRR1 in the mouse adult brain. Brain Struct Funct 2018; 223:4039-4052. [PMID: 30121783 PMCID: PMC6267262 DOI: 10.1007/s00429-018-1737-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Accepted: 08/13/2018] [Indexed: 12/28/2022]
Abstract
Identifying molecular targets that are able to buffer the consequences of stress and therefore restore brain homeostasis is essential to develop treatments for stress-related disorders. Down-regulated in renal cell carcinoma 1 (DRR1) is a unique stress-induced protein in the brain and has been recently proposed to modulate stress resilience. Interestingly, DRR1 shows a prominent expression in the limbic system of the adult mouse. Here, we analyzed the neuroanatomical and cellular expression patterns of DRR1 in the adult mouse brain using in situ hybridization, immunofluorescence and Western blot. Abundant expression of DRR1 mRNA and protein was confirmed in the adult mouse brain with pronounced differences between distinct brain regions. The strongest DRR1 signal was detected in the neocortex, the CA3 region of the hippocampus, the lateral septum and the cerebellum. DRR1 was also present in circumventricular organs and its connecting regions. Additionally, DRR1 was present in non-neuronal tissues like the choroid plexus and ependyma. Within cells, DRR1 protein was distributed in a punctate pattern in several subcellular compartments including cytosol, nucleus as well as some pre- and postsynaptic specializations. Glucocorticoid receptor activation (dexamethasone 10 mg/kg s.c.) induced DRR1 expression throughout the brain, with particularly strong induction in white matter and fiber tracts and in membrane-rich structures. This specific expression pattern and stress modulation of DRR1 point to a role of DRR1 in regulating how cells sense and integrate signals from the environment and thus in restoring brain homeostasis after stressful challenges.
Collapse
Affiliation(s)
- Mercè Masana
- Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804, Munich, Germany. .,Translational Psychiatry, Department of Psychiatry and Psychotherapy and Focus Program Translational Neuroscience (FTN), Johannes Gutenberg University Medical Center, Hanns-Dieter-Hüsch-Weg 19, 55128, Mainz, Germany. .,Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, IDIBAPS, CIBERNED, Barcelona, Spain.
| | - Sören Westerholz
- Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804, Munich, Germany
| | - Anja Kretzschmar
- Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804, Munich, Germany
| | - Giulia Treccani
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmacologiche e Biomolecolari and CEND, Università di Milano, Milan, Italy.,Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Risskov, Denmark
| | - Claudia Liebl
- Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804, Munich, Germany
| | - Sara Santarelli
- Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804, Munich, Germany
| | - Carine Dournes
- Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804, Munich, Germany
| | - Maurizio Popoli
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmacologiche e Biomolecolari and CEND, Università di Milano, Milan, Italy
| | - Mathias V Schmidt
- Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804, Munich, Germany
| | - Theo Rein
- Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804, Munich, Germany
| | - Marianne B Müller
- Max Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804, Munich, Germany.,Translational Psychiatry, Department of Psychiatry and Psychotherapy and Focus Program Translational Neuroscience (FTN), Johannes Gutenberg University Medical Center, Hanns-Dieter-Hüsch-Weg 19, 55128, Mainz, Germany.,Deutsches Resilienz-Zentrum, Mainz, Germany
| |
Collapse
|
4
|
Murphy MO, Herald JB, Leachman J, Villasante Tezanos A, Cohn DM, Loria AS. A model of neglect during postnatal life heightens obesity-induced hypertension and is linked to a greater metabolic compromise in female mice. Int J Obes (Lond) 2018; 42:1354-1365. [PMID: 29535450 PMCID: PMC6054818 DOI: 10.1038/s41366-018-0035-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 01/05/2018] [Accepted: 01/14/2018] [Indexed: 12/12/2022]
Abstract
.: Exposure to early life stress (ELS) is associated with behavioral-related alterations, increases in body mass index and higher systolic blood pressure in humans. Postnatal maternal separation and early weaning (MSEW) is a mouse model of neglect characterized by a long-term dysregulation of the neuroendocrine system. OBJECTIVES Given the contribution of adrenal-derived hormones to the development of obesity, we hypothesized that exposure to MSEW could contribute to the worsening of cardiometabolic function in response to chronic high-fat diet (HF) feeding by promoting adipose tissue expansion and insulin resistance. SUBJECTS MSEW was performed in C57BL/6 mice from postnatal days 2-16 and weaned at postnatal day 17. Undisturbed litters weaned at postnatal day 21 served as the control (C) group. At the weaning day, mice were placed on a low-fat diet (LF) or HF for 16 weeks. RESULTS When fed a LF, male and female mice exposed to MSEW display similar body weight but increased fat mass compared to controls. However, when fed a HF, only female MSEW mice display increased body weight, fat mass, and adipocyte hypertrophy compared with controls. Also, female MSEW mice display evidence of an early onset of cardiometabolic risk factors, including hyperinsulinemia, glucose intolerance, and hypercholesterolemia. Yet, both male and female MSEW mice fed a HF show increased blood pressure compared with controls. CONCLUSIONS This study shows that MSEW promotes a sex-specific dysregulation of the adipose tissue expansion and glucose homeostasis that precedes the development of obesity-induced hypertension.
Collapse
Affiliation(s)
- Margaret O Murphy
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Joseph B Herald
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Jacqueline Leachman
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | | | - Dianne M Cohn
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Analia S Loria
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
5
|
van der Kooij MA, Masana M, Rust MB, Müller MB. The stressed cytoskeleton: How actin dynamics can shape stress-related consequences on synaptic plasticity and complex behavior. Neurosci Biobehav Rev 2016; 62:69-75. [DOI: 10.1016/j.neubiorev.2015.12.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 12/01/2015] [Accepted: 12/07/2015] [Indexed: 01/06/2023]
|
6
|
Bolten M. Transgenerational Transmission of Stress Pathology. ZEITSCHRIFT FUR PSYCHOLOGIE-JOURNAL OF PSYCHOLOGY 2015. [DOI: 10.1027/2151-2604/a000219] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Abstract. The impact of the environment early in life on long-term outcomes is well known. Stressful experiences during pre- and postnatal development can modulate the genetic programming of specific brain circuits underlying emotional and cognitive aspects of behavioral adaptation to stressful experiences later in life. Furthermore, there is documented evidence for gene-environment interactions in the context of early-life stress. Identical gene variants can be associated with different phenotypes depending on environmental factors. DNA methylation, an enzymatically-catalyzed modification of the DNA, is the mechanism through which phenotypes are regulated. The dynamics and plasticity of epigenetic mechanisms can have short-term, long-term, or transgenerational consequences. In epigenetic research, rodent models have targeted several behavioral and emotional phenotypes. These models have contributed significantly to our understanding of the environmental regulation of the developmental brain in early life. This review will highlight studies with rats and mice on epigenetic processes in fetal programming of stress-related mental disorders.
Collapse
Affiliation(s)
- Margarete Bolten
- Child and Adolescent Psychiatric Clinic, University of Basel, Switzerland
| |
Collapse
|
7
|
Masana M, Jukic M, Kretzschmar A, Wagner K, Westerholz S, Schmidt M, Rein T, Brodski C, Müller M. Deciphering the spatio-temporal expression and stress regulation of Fam107B, the paralog of the resilience-promoting protein DRR1 in the mouse brain. Neuroscience 2015; 290:147-58. [DOI: 10.1016/j.neuroscience.2015.01.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 01/12/2015] [Accepted: 01/16/2015] [Indexed: 11/26/2022]
|
8
|
Araujo FC, Milsted A, Watanabe IKM, Del Puerto HL, Santos RAS, Lazar J, Reis FM, Prokop JW. Similarities and differences of X and Y chromosome homologous genes, SRY and SOX3, in regulating the renin-angiotensin system promoters. Physiol Genomics 2015; 47:177-86. [PMID: 25759379 DOI: 10.1152/physiolgenomics.00138.2014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 03/09/2015] [Indexed: 12/17/2022] Open
Abstract
The renin-angiotensin system (RAS) is subject to sex-specific modulation by hormones and gene products. However, sex differences in the balance between the vasoconstrictor/proliferative ACE/ANG II/AT1 axis, and the vasodilator/antiproliferative ACE2/ANG-(1-7)/MAS axis are poorly known. Data in the rat have suggested the male-specific Y-chromosome gene Sry to contribute to balance between these two axes, but why the testis-determining gene has these functions remains unknown. A combination of in silico genetic/protein comparisons, functional luciferase assays for promoters of the human RAS, and RNA-Seq profiling in rat were used to address if regulation of Sry on the RAS is conserved in the homologous X-chromosome gene, Sox3. Both SRY and SOX3 upregulated the promoter of Angiotensinogen (AGT) and downregulated the promoters of ACE2, AT2, and MAS, likely through overlapping mechanisms. The regulation by both SRY and SOX3 on the MAS promoter indicates a cis regulation through multiple SOX binding sites. The Renin (REN) promoter is upregulated by SRY and downregulated by SOX3, likely through trans and cis mechanisms, respectively. Sry transcripts are found in all analyzed male rat tissues including the kidney, while Sox3 transcripts are found only in the brain and testis, suggesting that the primary tissue for renin production (kidney) can only be regulated by SRY and not SOX3. These results suggest that SRY regulation of the RAS is partially shared with its X-chromosome homolog SOX3, but SRY gained a sex-specific control in the kidney for the rate-limiting step of the RAS, potentially resulting in male-specific blood pressure regulation.
Collapse
Affiliation(s)
- Fabiano C Araujo
- National Institute of Science and Technology in Molecular Medicine and Department of Obstetrics and Gynecology, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Amy Milsted
- Department of Biology, The University of Akron, Akron, Ohio
| | - Ingrid K M Watanabe
- Nephrology Division, Department of Medicine, Federal University of Sao Paulo, Sao Paulo, Brazil
| | - Helen L Del Puerto
- National Institute of Science and Technology in Molecular Medicine and Department of Obstetrics and Gynecology, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Robson A S Santos
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Jozef Lazar
- Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, Wisconsin; and Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Fernando M Reis
- National Institute of Science and Technology in Molecular Medicine and Department of Obstetrics and Gynecology, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Jeremy W Prokop
- Human and Molecular Genetics Center, Medical College of Wisconsin, Milwaukee, Wisconsin; and Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
9
|
Stankiewicz AM, Goscik J, Swiergiel AH, Majewska A, Wieczorek M, Juszczak GR, Lisowski P. Social stress increases expression of hemoglobin genes in mouse prefrontal cortex. BMC Neurosci 2014; 15:130. [PMID: 25472829 PMCID: PMC4269175 DOI: 10.1186/s12868-014-0130-6] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Accepted: 11/18/2014] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND In order to better understand the effects of social stress on the prefrontal cortex, we investigated gene expression in mice subjected to acute and repeated social encounters of different duration using microarrays. RESULTS The most important finding was identification of hemoglobin genes (Hbb-b1, Hbb-b2, Hba-a1, Hba-a2, Beta-S) as potential markers of chronic social stress in mice. Expression of these genes was progressively increased in animals subjected to 8 and 13 days of repeated stress and was correlated with altered expression of Mgp (Mglap), Fbln1, 1500015O10Rik (Ecrg4), SLC16A10, and Mndal. Chronic stress increased also expression of Timp1 and Ppbp that are involved in reaction to vascular injury. Acute stress did not affect expression of hemoglobin genes but it altered expression of Fam107a (Drr1) and Agxt2l1 (Etnppl) that have been implicated in psychiatric diseases. CONCLUSIONS The observed up-regulation of genes associated with vascular system and brain injury suggests that stressful social encounters may affect brain function through the stress-induced dysfunction of the vascular system.
Collapse
Affiliation(s)
- Adrian M Stankiewicz
- Department of Animal Behavior, Institute of Genetics and Animal Breeding, Jastrzebiec, ul. Postepu 36A, 05-552, Magdalenka, Poland.
| | - Joanna Goscik
- Faculty of Computer Science, Bialystok University of Technology, Wiejska 45A, 15-351, Bialystok, Poland.
| | - Artur H Swiergiel
- Department of Human and Animal Physiology, Institute of Biology, University of Gdansk, 80-308, Gdansk, Poland.
| | - Alicja Majewska
- Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences, Warsaw, Poland.
| | - Marek Wieczorek
- Department of Neurobiology, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Pomorska, 141/143, Poland.
| | - Grzegorz R Juszczak
- Department of Animal Behavior, Institute of Genetics and Animal Breeding, Jastrzebiec, ul. Postepu 36A, 05-552, Magdalenka, Poland.
| | - Paweł Lisowski
- Department of Molecular Biology, Institute of Genetics and Animal Breeding, Jastrzebiec, ul. Postepu 36A, 05-552, Magdalenka, Poland. .,iPS Cell-Based Disease Modeling Group, Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, 13092, Berlin, Germany.
| |
Collapse
|
10
|
The stress-inducible actin-interacting protein DRR1 shapes social behavior. Psychoneuroendocrinology 2014; 48:98-110. [PMID: 24998413 DOI: 10.1016/j.psyneuen.2014.06.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 06/10/2014] [Accepted: 06/11/2014] [Indexed: 01/18/2023]
Abstract
Understanding the molecular mechanisms by which stress is translated into changes in complex behavior may help to identify novel treatment strategies for stress-associated psychiatric disorders. The tumor suppressor gene down-regulated in renal cell carcinoma 1 (DRR1) was recently characterized as a new molecular link between stress, synaptic efficacy and behavioral performance, most likely through its ability to modulate actin dynamics. The lateral septum is one of the brain regions prominently involved in the stress response. This brain region features high DRR1 expression in adult mice, even under basal conditions. We therefore aimed to characterize and dissect the functional role of septal DRR1 in modulating complex behavior. DRR1 protein expression was shown to be expressed in both neurons and astrocytes of the lateral septum of adult mice. Septal DRR1 mRNA expression increased after acute defeat stress and glucocorticoid receptor activation. To mimic the stress-induced DRR1 increase in the lateral septum of mice, we performed adenovirus-mediated region-specific overexpression of DRR1 and characterized the behavior of these mice. Overexpression of DRR1 in the septal region increased sociability, but did not change cognitive, anxiety-like or anhedonic behavior. The observed changes in social behavior did not involve alterations of the expression of vasopressin or oxytocin receptors, the canonical social neuropeptidergic circuits of the lateral septum. In summary, our data suggest that the stress-induced increase of DRR1 expression in the lateral septum could be a protective mechanism to buffer or counterbalance negative consequences of stress exposure on social behavior.
Collapse
|
11
|
Daskalakis NP, Bagot RC, Parker KJ, Vinkers CH, de Kloet ER. The three-hit concept of vulnerability and resilience: toward understanding adaptation to early-life adversity outcome. Psychoneuroendocrinology 2013; 38:1858-73. [PMID: 23838101 PMCID: PMC3773020 DOI: 10.1016/j.psyneuen.2013.06.008] [Citation(s) in RCA: 373] [Impact Index Per Article: 33.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 06/06/2013] [Accepted: 06/07/2013] [Indexed: 01/08/2023]
Abstract
Stressful experiences during early-life can modulate the genetic programming of specific brain circuits underlying emotional and cognitive aspects of behavioral adaptation to stressful experiences later in life. Although this programming effect exerted by experience-related factors is an important determinant of mental health, its outcome depends on cognitive inputs and hence the valence an individual assigns to a given environmental context. From this perspective we will highlight, with studies in rodents, non-human primates and humans, the three-hit concept of vulnerability and resilience to stress-related mental disorders, which is based on gene-environment interactions during critical phases of perinatal and juvenile brain development. The three-hit (i.e., hit-1: genetic predisposition, hit-2: early-life environment, and hit-3: later-life environment) concept accommodates the cumulative stress hypothesis stating that in a given context vulnerability is enhanced when failure to cope with adversity accumulates. Alternatively, the concept also points to the individual's predictive adaptive capacity, which underlies the stress inoculation and match/mismatch hypotheses. The latter hypotheses propose that the experience of relatively mild early-life adversity prepares for the future and promotes resilience to similar challenges in later-life; when a mismatch occurs between early and later-life experience, coping is compromised and vulnerability is enhanced. The three-hit concept is fundamental for understanding how individuals can either be prepared for coping with life to come and remain resilient or are unable to do so and succumb to a stress-related mental disorder, under seemingly identical circumstances.
Collapse
Affiliation(s)
- Nikolaos P. Daskalakis
- Traumatic Stress Studies Division & Laboratory of Molecular Neuropsychiatry, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA,PTSD Clinical Research Program & Laboratory of Clinical Neuroendocrinology and Neurochemistry, James J. Peters Veterans Affairs Medical Center, Bronx, USA,Division of Medical Pharmacology, Leiden/ Amsterdam Center for Drug Research & Leiden University Medical Center, Leiden University, Leiden, The Netherlands,Correspondence: Dr. Nikolaos Daskalakis, Laboratory of Molecular Neuropsychiatry, Mount Sinai School of Medicine, One Gustave L. Levy Place, Box 1668, Annenberg building - Room 22-38, New York, NY 10029-6574, USA,
| | - Rosemary C. Bagot
- Neuroscience Division, Douglas Mental Health University Institute, Montreal, Quebec, Canada,Laboratory of Molecular Psychiatry, Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Karen J. Parker
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, USA
| | - Christiaan H. Vinkers
- Department of Psychiatry, Rudolf Magnus Institute of Neuroscience, University Medical Center Utrecht, Utrecht, the Netherlands,Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences and Rudolf Magnus Institute of Neuroscience, Utrecht University, Utrecht, the Netherlands
| | - E. R. de Kloet
- Division of Medical Pharmacology, Leiden/ Amsterdam Center for Drug Research & Leiden University Medical Center, Leiden University, Leiden, The Netherlands
| |
Collapse
|
12
|
Barouei J, Moussavi M, Hodgson DM. Effect of maternal probiotic intervention on HPA axis, immunity and gut microbiota in a rat model of irritable bowel syndrome. PLoS One 2012; 7:e46051. [PMID: 23071537 PMCID: PMC3469551 DOI: 10.1371/journal.pone.0046051] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Accepted: 08/28/2012] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE To examine whether maternal probiotic intervention influences the alterations in the brain-immune-gut axis induced by neonatal maternal separation (MS) and/or restraint stress in adulthood (AS) in Wistar rats. DESIGN Dams had free access to drinking water supplemented with Bifidobacterium animalis subsp lactis BB-12® (3 × 10(9) CFU/mL) and Propionibacterium jensenii 702 (8.0 × 10(8) CFU/mL) from 10 days before conception until postnatal day (PND) 22 (weaning day), or to control ad lib water. Offspring were subjected to MS from PND 2 to 14 or left undisturbed. From PND 83 to 85, animals underwent 30 min/day AS, or were left undisturbed as controls. On PND 24 and 86, blood samples were collected for corticosterone, ACTH and IgA measurement. Colonic contents were analysed for the composition of microflora and luminal IgA levels. RESULTS Exposure to MS significantly increased ACTH levels and neonatal fecal counts of aerobic and anaerobic bacteria, E. coli, enterococci and clostridia, but reduced plasma IgA levels compared with non-MS animals. Animals exposed to AS exhibited significantly increased ACTH and corticosterone levels, decreased aerobic bacteria and bifidobacteria, and increased Bacteroides and E. coli counts compared to non-AS animals. MS coupled with AS induced significantly decreased anaerobes and clostridia compared with the non-stress adult controls. Maternal probiotic intervention significantly increased neonatal corticosterone levels which persisted until at least week 12 in females only, and also resulted in elevated adult ACTH levels and altered neonatal microflora comparable to that of MS. However, it improved plasma IgA responses, increased enterococci and clostridia in MS adults, increased luminal IgA levels, and restored anaerobes, bifidobacteria and E. coli to normal in adults. CONCLUSION Maternal probiotic intervention induced activation of neonatal stress pathways and an imbalance in gut microflora. Importantly however, it improved the immune environment of stressed animals and protected, in part, against stress-induced disturbances in adult gut microflora.
Collapse
Affiliation(s)
- Javad Barouei
- Laboratory of Microbiology, School of Environmental and Life Sciences, the University of Newcastle, Callaghan, New South Wales, Australia.
| | | | | |
Collapse
|
13
|
van Eekelen JAM, Ellis JA, Pennell CE, Craig J, Saffery R, Mattes E, Olsson CA. Stress-sensitive neurosignalling in depression: an integrated network biology approach to candidate gene selection for genetic association analysis. Ment Illn 2012; 4:e21. [PMID: 25478122 PMCID: PMC4253374 DOI: 10.4081/mi.2012.e21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Revised: 07/06/2012] [Accepted: 08/01/2012] [Indexed: 01/18/2023] Open
Abstract
Genetic risk for depressive disorders is poorly understood despite consistent suggestions of a high heritable component. Most genetic studies have focused on risk associated with single variants, a strategy which has so far only yielded small (often non-replicable) risks for depressive disorders. In this paper we argue that more substantial risks are likely to emerge from genetic variants acting in synergy within and across larger neurobiological systems (polygenic risk factors). We show how knowledge of major integrated neurobiological systems provides a robust basis for defining and testing theoretically defensible polygenic risk factors. We do this by describing the architecture of the overall stress response. Maladaptation via impaired stress responsiveness is central to the aetiology of depression and anxiety and provides a framework for a systems biology approach to candidate gene selection. We propose principles for identifying genes and gene networks within the neurosystems involved in the stress response and for defining polygenic risk factors based on the neurobiology of stress-related behaviour. We conclude that knowledge of the neurobiology of the stress response system is likely to play a central role in future efforts to improve genetic prediction of depression and related disorders.
Collapse
Affiliation(s)
- J. Anke M. van Eekelen
- Developmental Neuroscience, Telethon Institute for Child Health Research and Centre for Child Health Research, University of Western Australia, Perth
| | - Justine A. Ellis
- Environmental and Genetic Epidemiology, Murdoch Childrens Research Institute, The Royal Children's Hospital and Department of Physiology, University of Melbourne
| | - Craig E. Pennell
- The School of Women's and Infants' Health, University of Western Australia at King Edward Memorial Hospital
| | - Jeff Craig
- Developmental Epigenetics, Early Development and Disease, Department of Paediatrics, Murdoch Childrens Research Institute, Royal Children's Hospital
| | - Richard Saffery
- Developmental Epigenetics, Early Development and Disease, Department of Paediatrics, Murdoch Childrens Research Institute, Royal Children's Hospital
| | - Eugen Mattes
- Developmental Neuroscience, Telethon Institute for Child Health Research and Centre for Child Health Research, University of Western Australia, Perth
| | - Craig A. Olsson
- School of Psychology, Deakin University Australia; Murdoch Childrens Research Institute; University of Melbourne, Australia
| |
Collapse
|
14
|
Angiotensin II AT1 receptor blocker candesartan prevents the fast up-regulation of cerebrocortical benzodiazepine-1 receptors induced by acute inflammatory and restraint stress. Behav Brain Res 2012; 232:84-92. [PMID: 22503782 DOI: 10.1016/j.bbr.2012.03.041] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Revised: 03/23/2012] [Accepted: 03/28/2012] [Indexed: 01/12/2023]
Abstract
Centrally acting Angiotensin II AT(1) receptor blockers (ARBs) protect from stress-induced disorders and decrease anxiety in a model of inflammatory stress, the systemic injection of bacterial endotoxin lipopolysaccharide (LPS). In order to better understand the anxiolytic effect of ARBs, we treated rats with LPS (50 μg/kg) with or without 3 days of pretreatment with the ARB candesartan (1mg/kg/day), and studied cortical benzodiazepine (BZ) and corticotrophin-releasing factor (CRF) receptors. We compared the cortical BZ and CRF receptors expression pattern induced by LPS with that produced in restraint stress. Inflammation stress produced a generalized increase in cortical BZ(1) receptors and reduced mRNA expression of the GABA(A) receptor γ(2) subunit in cingulate cortex; changes were prevented by candesartan pretreatment. Moreover, restraint stress produced similar increases in cortical BZ(1) receptor binding, and candesartan prevented these changes. Treatment with candesartan alone increased cortical BZ(1) binding, and decreased γ(2) subunit mRNA expression in the cingulate cortex. Conversely, we did not find changes in CRF(1) receptor expression in any of the cortical areas studied, either after inflammation or restraint stress. Cortical CRF(2) receptor binding was undetectable, but CRF(2) mRNA expression was decreased by inflammation stress, a change prevented by candesartan. We conclude that stress promotes rapid and widespread changes in cortical BZ(1) receptor expression; and that the stress-induced BZ(1) receptor expression is under the control of AT(1) receptor activity. The results suggest that the anti-anxiety effect of ARBs may be associated with their capacity to regulate stress-induced alterations in cortical BZ(1) receptors.
Collapse
|
15
|
Lajud N, Roque A, Cajero M, Gutiérrez-Ospina G, Torner L. Periodic maternal separation decreases hippocampal neurogenesis without affecting basal corticosterone during the stress hyporesponsive period, but alters HPA axis and coping behavior in adulthood. Psychoneuroendocrinology 2012; 37:410-20. [PMID: 21862224 DOI: 10.1016/j.psyneuen.2011.07.011] [Citation(s) in RCA: 121] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2010] [Revised: 07/08/2011] [Accepted: 07/18/2011] [Indexed: 02/03/2023]
Abstract
Although not directly evaluated, the early rise of glucocorticoid (GC) levels, as occur after exposure to adverse early life experience, are assumed to affect hippocampal ontogeny by altering the hippocampus negative feedback on adult HPA axis. To test whether hippocampal ontogeny is affected by early exposure to stress we estimated the survival of recently formed hippocampal granule cells in rat pups subjected to periodic maternal separation (180 min/day; MS180) from postnatal days (PND) 1 to 14. Accordingly, MS180 pups injected with bromodeoxyuridine (BrdU, 50 mg/kg, ip) at PND 5 showed decreased density of doublecortin (DCX) positive BrdU-labeled cells at PND 15. MS180 and AFR pups showed similar corticosterone (CORT) basal levels between PND 3 and 12, whereas adult MS180 rats presented with higher CORT levels than AFR adults. Nonetheless, both AFR and MS180 pups and adults showed similar transient increments of CORT levels in response to stress. In addition, MS180 had no effect on the adult anxiety-like behavior evaluated in the elevated plus maze, but evoked a passive coping strategy in the forced swimming test. The data show that the decrease in hippocampal neurogenesis is an early onset phenomenon, and suggests that adverse experiences alter hippocampal ontogeny without chronic elevation of GC levels.
Collapse
Affiliation(s)
- Naima Lajud
- División de Neurociencias, Centro de Investigación Biomédica de Michoacán - Instituto Mexicano del Seguro Social, Morelia 58341, Michoacán, Mexico
| | | | | | | | | |
Collapse
|
16
|
Meier JC, Harvey RJ, Seeburg P. Frontiers in molecular neuroscience - résumé and perspective. Front Mol Neurosci 2012; 4:58. [PMID: 22232574 PMCID: PMC3248788 DOI: 10.3389/fnmol.2011.00058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Accepted: 12/15/2011] [Indexed: 11/13/2022] Open
Affiliation(s)
- Jochen C Meier
- Max Delbrück Center for Molecular Medicine Berlin, Germany
| | | | | |
Collapse
|
17
|
Coffee RL, Williamson AJ, Adkins CM, Gray MC, Page TL, Broadie K. In vivo neuronal function of the fragile X mental retardation protein is regulated by phosphorylation. Hum Mol Genet 2011; 21:900-15. [PMID: 22080836 DOI: 10.1093/hmg/ddr527] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Fragile X syndrome (FXS), caused by loss of the Fragile X Mental Retardation 1 (FMR1) gene product (FMRP), is the most common heritable cause of intellectual disability and autism spectrum disorders. It has been long hypothesized that the phosphorylation of serine 500 (S500) in human FMRP controls its function as an RNA-binding translational repressor. To test this hypothesis in vivo, we employed neuronally targeted expression of three human FMR1 transgenes, including wild-type (hFMR1), dephosphomimetic (S500A-hFMR1) and phosphomimetic (S500D-hFMR1), in the Drosophila FXS disease model to investigate phosphorylation requirements. At the molecular level, dfmr1 null mutants exhibit elevated brain protein levels due to loss of translational repressor activity. This defect is rescued for an individual target protein and across the population of brain proteins by the phosphomimetic, whereas the dephosphomimetic phenocopies the null condition. At the cellular level, dfmr1 null synapse architecture exhibits increased area, branching and bouton number. The phosphomimetic fully rescues these synaptogenesis defects, whereas the dephosphomimetic provides no rescue. The presence of Futsch-positive (microtubule-associated protein 1B) supernumerary microtubule loops is elevated in dfmr1 null synapses. The human phosphomimetic restores normal Futsch loops, whereas the dephosphomimetic provides no activity. At the behavioral level, dfmr1 null mutants exhibit strongly impaired olfactory associative learning. The human phosphomimetic targeted only to the brain-learning center restores normal learning ability, whereas the dephosphomimetic provides absolutely no rescue. We conclude that human FMRP S500 phosphorylation is necessary for its in vivo function as a neuronal translational repressor and regulator of synaptic architecture, and for the manifestation of FMRP-dependent learning behavior.
Collapse
Affiliation(s)
- R Lane Coffee
- Department of Biological Sciences, Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN 37232, USA
| | | | | | | | | | | |
Collapse
|
18
|
Ackermann M, Kubitza M, Maier K, Brawanski A, Hauska G, Piña AL. The vertebrate homolog of sulfide-quinone reductase is expressed in mitochondria of neuronal tissues. Neuroscience 2011; 199:1-12. [PMID: 22067608 DOI: 10.1016/j.neuroscience.2011.10.044] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2011] [Revised: 10/15/2011] [Accepted: 10/17/2011] [Indexed: 12/24/2022]
Abstract
Hydrogen sulfide (H₂S) can be consumed by both invertebrates and vertebrates as an inorganic substrate. The pathway metabolizing H₂S probably involves three mitochondrial enzymes, one of which is sulfide-quinone oxidoreductase (SQR), known as sulfide-quinone reductase-like protein (SQRDL) in vertebrates. Evidence from fission yeast suggests that SQR might have a role in regulating sulfide levels in the cell. Regulation might be essential for H₂S to act as a gaseous transmitter (gasotransmitter). The brain is an organ with high activity of gasotransmitters, like nitric oxide (NO) and H₂S, which are known to affect synaptic transmission. In this study, we provide evidence that SQRDL is expressed in the mammalian brain. Real-time polymerase chain reaction (PCR) showed an increase in the number of Sqrdl transcripts in the brain with increasing age. Cellular fractionation and subsequent analysis by Western blotting indicated that the protein is located in mitochondria, which is the site of sulfide consumption in the cell. With an immunohistochemical approach, we demonstrated that the SQRDL protein is expressed in neurons, oligodendrocytes, and endothelial cells. Taken together, our data suggest that brain tissue harbors the machinery required for local regulation of sulfide levels.
Collapse
Affiliation(s)
- M Ackermann
- Department of Neurosurgery at the University Clinic, University of Regensburg, Regensburg, Germany
| | | | | | | | | | | |
Collapse
|
19
|
Tumor suppressor down-regulated in renal cell carcinoma 1 (DRR1) is a stress-induced actin bundling factor that modulates synaptic efficacy and cognition. Proc Natl Acad Sci U S A 2011; 108:17213-8. [PMID: 21969592 DOI: 10.1073/pnas.1103318108] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Stress has been identified as a major causal factor for many mental disorders. However, our knowledge about the chain of molecular and cellular events translating stress experience into altered behavior is still rather scant. Here, we have characterized a murine ortholog of the putative tumor suppressor gene DRR1 as a unique stress-induced protein in brain. It binds to actin, promotes bundling and stabilization of actin filaments, and impacts on actin-dependent neurite outgrowth. Endogenous DRR1 localizes to some, but not all, synapses, with preference for the presynaptic region. Hippocampal virus-mediated enhancement of DRR1 expression reduced spine density, diminished the probability of synaptic glutamate release, and altered cognitive performance. DRR1 emerges as a protein to link stress with actin dynamics, which in addition is able to act on synaptic function and cognition.
Collapse
|
20
|
Profiling trait anxiety: transcriptome analysis reveals cathepsin B (Ctsb) as a novel candidate gene for emotionality in mice. PLoS One 2011; 6:e23604. [PMID: 21897848 PMCID: PMC3163650 DOI: 10.1371/journal.pone.0023604] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2011] [Accepted: 07/20/2011] [Indexed: 11/19/2022] Open
Abstract
Behavioral endophenotypes are determined by a multitude of counteracting but precisely balanced molecular and physiological mechanisms. In this study, we aim to identify potential novel molecular targets that contribute to the multigenic trait “anxiety”. We used microarrays to investigate the gene expression profiles of different brain regions within the limbic system of mice which were selectively bred for either high (HAB) or low (LAB) anxiety-related behavior, and also show signs of comorbid depression-like behavior. We identified and confirmed sex-independent differences in the basal expression of 13 candidate genes, using tissue from the entire brain, including coronin 7 (Coro7), cathepsin B (Ctsb), muscleblind-like 1 (Mbnl1), metallothionein 1 (Mt1), solute carrier family 25 member 17 (Slc25a17), tribbles homolog 2 (Trib2), zinc finger protein 672 (Zfp672), syntaxin 3 (Stx3), ATP-binding cassette, sub-family A member 2 (Abca2), ectonucleotide pyrophosphatase/phosphodiesterase 5 (Enpp5), high mobility group nucleosomal binding domain 3 (Hmgn3) and pyruvate dehydrogenase beta (Pdhb). Additionally, we confirmed brain region-specific differences in the expression of synaptotagmin 4 (Syt4). Our identification of about 90 polymorphisms in Ctsb suggested that this gene might play a critical role in shaping our mouse model's behavioral endophenotypes. Indeed, the assessment of anxiety-related and depression-like behaviors of Ctsb knock-out mice revealed an increase in depression-like behavior in females. Altogether, our results suggest that Ctsb has significant effects on emotionality, irrespective of the tested mouse strain, making it a promising target for future pharmacotherapy.
Collapse
|
21
|
Le-Niculescu H, Case NJ, Hulvershorn L, Patel SD, Bowker D, Gupta J, Bell R, Edenberg HJ, Tsuang MT, Kuczenski R, Geyer MA, Rodd ZA, Niculescu AB. Convergent functional genomic studies of ω-3 fatty acids in stress reactivity, bipolar disorder and alcoholism. Transl Psychiatry 2011; 1:e4. [PMID: 22832392 PMCID: PMC3309466 DOI: 10.1038/tp.2011.1] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2010] [Accepted: 02/24/2011] [Indexed: 12/28/2022] Open
Abstract
Omega-3 fatty acids have been proposed as an adjuvant treatment option in psychiatric disorders. Given their other health benefits and their relative lack of toxicity, teratogenicity and side effects, they may be particularly useful in children and in females of child-bearing age, especially during pregnancy and postpartum. A comprehensive mechanistic understanding of their effects is needed. Here we report translational studies demonstrating the phenotypic normalization and gene expression effects of dietary omega-3 fatty acids, specifically docosahexaenoic acid (DHA), in a stress-reactive knockout mouse model of bipolar disorder and co-morbid alcoholism, using a bioinformatic convergent functional genomics approach integrating animal model and human data to prioritize disease-relevant genes. Additionally, to validate at a behavioral level the novel observed effects on decreasing alcohol consumption, we also tested the effects of DHA in an independent animal model, alcohol-preferring (P) rats, a well-established animal model of alcoholism. Our studies uncover sex differences, brain region-specific effects and blood biomarkers that may underpin the effects of DHA. Of note, DHA modulates some of the same genes targeted by current psychotropic medications, as well as increases myelin-related gene expression. Myelin-related gene expression decrease is a common, if nonspecific, denominator of neuropsychiatric disorders. In conclusion, our work supports the potential utility of omega-3 fatty acids, specifically DHA, for a spectrum of psychiatric disorders such as stress disorders, bipolar disorder, alcoholism and beyond.
Collapse
Affiliation(s)
- H Le-Niculescu
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - N J Case
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - L Hulvershorn
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - S D Patel
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
- Indianapolis VA Medical Center, Indianapolis, IN, USA
| | - D Bowker
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - J Gupta
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - R Bell
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - H J Edenberg
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - M T Tsuang
- Department of Psychiatry, UC San Diego, La Jolla, CA, USA
| | - R Kuczenski
- Department of Psychiatry, UC San Diego, La Jolla, CA, USA
| | - M A Geyer
- Department of Psychiatry, UC San Diego, La Jolla, CA, USA
| | - Z A Rodd
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
| | - A B Niculescu
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
- Indianapolis VA Medical Center, Indianapolis, IN, USA
| |
Collapse
|
22
|
Early life stress paradigms in rodents: potential animal models of depression? Psychopharmacology (Berl) 2011; 214:131-40. [PMID: 21086114 DOI: 10.1007/s00213-010-2096-0] [Citation(s) in RCA: 130] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2010] [Accepted: 11/03/2010] [Indexed: 12/13/2022]
Abstract
RATIONALE While human depressive illness is indeed uniquely human, many of its symptoms may be modeled in rodents. Based on human etiology, the assumption has been made that depression-like behavior in rats and mice can be modulated by some of the powerful early life programming effects that are known to occur after manipulations in the first weeks of life. OBJECTIVE Here we review the evidence that is available in literature for early life manipulation as risk factors for the development of depression-like symptoms such as anhedonia, passive coping strategies, and neuroendocrine changes. Early life paradigms that were evaluated include early handling, separation, and deprivation protocols, as well as enriched and impoverished environments. We have also included a small number of stress-related pharmacological models. RESULTS We find that for most early life paradigms per se, the actual validity for depression is limited. A number of models have not been tested with respect to classical depression-like behaviors, while in many cases, the outcome of such experiments is variable and depends on strain and additional factors. CONCLUSION Because programming effects confer vulnerability rather than disease, a number of paradigms hold promise for usefulness in depression research, in combination with the proper genetic background and adult life challenges.
Collapse
|
23
|
Gao P, Ishige A, Murakami Y, Nakata H, Oka JI, Munakata K, Yamamoto M, Nishimura K, Watanabe K. Maternal stress affects postnatal growth and the pituitary expression of prolactin in mouse offspring. J Neurosci Res 2011; 89:329-40. [PMID: 21259320 DOI: 10.1002/jnr.22550] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2010] [Revised: 10/14/2010] [Accepted: 10/14/2010] [Indexed: 01/12/2023]
Abstract
Maternal stress exerts long-lasting psychiatric and somatic on offspring, which persist into adulthood. However, the effect of maternal stress on the postnatal growth of pups has not been widely reported. In this study, we found that maternal immobilization stress (IS) during lactation resulted in low body weight of male mouse offspring, which persisted after weaning. Despite free access to chow, IS induced maternal malnutrition and decreased the serum insulin-like growth factor-1 (IGF-1) levels in the mothers and in the pups. mRNA expression analysis of anterior pituitary hormones in the pups revealed that growth hormone (GH) and prolactin (PRL), but no other hormones, were decreased by IS. Expression of the pituitary transcription factor PIT1 and isoforms of PITX2, which are essential for the development and function of GH-producing somatotropes and PRL-producing lactotropes, was decreased, whereas that of PROP1, which is critical for the earlier stages of pituitary development, was unchanged. Immunohistochemistry also showed a decrease in pituitary PRL protein expression. These results suggest that stress in a postpartum mother has persistent effects on the body weight of the offspring. Reduced PRL expression in the offspring's pituitary gland may play a role in these effects.
Collapse
Affiliation(s)
- Pengfei Gao
- Center for Kampo Medicine, Keio University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Hoyle D, Juhasz G, Aso E, Chase D, del Rio J, Fabre V, Hamon M, Lanfumey L, Lesch KP, Maldonado R, Serra MA, Sharp T, Tordera R, Toro C, Deakin JFW. Shared changes in gene expression in frontal cortex of four genetically modified mouse models of depression. Eur Neuropsychopharmacol 2011; 21:3-10. [PMID: 21030216 DOI: 10.1016/j.euroneuro.2010.09.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2010] [Revised: 09/21/2010] [Accepted: 09/24/2010] [Indexed: 12/01/2022]
Abstract
This study aimed to identify whether genetic manipulation of four systems implicated in the pathogenesis of depression converge on shared molecular processes underpinning depression-like behaviour in mice. Altered 5HT function was modelled using the 5-HT transporter knock out mouse, impaired glucocorticoid receptor (GR) function using an antisense-induced knock down mouse, disrupted glutamate function using a heterozygous KO of the vesicular glutamate transporter 1 gene, and impaired cannabinoid signalling using the cannabinoid 1 receptor KO mouse. All 4 four genetically modified mice were previously shown to show exaggerated helpless behaviour compared to wild-type controls and variable degrees of anxiety and anhedonic behaviour. mRNA was extracted from frontal cortex and hybridised to Illumina microarrays. Combined contrast analysis was used to identify genes showing different patterns of up- and down-regulation across the 4 models. 1823 genes were differentially regulated. They were over-represented in gene ontology categories of metabolism, protein handling and synapse. In each model compared to wild-type mice of the same genetic background, a number of genes showed increased expression changes of >10%, other genes showed decreases in each model. Most of the genes showed mixed effects. Several previous array findings were replicated. The results point to cellular stress and changes in post-synaptic remodelling as final common mechanisms of depression and resilience.
Collapse
Affiliation(s)
- D Hoyle
- Faculty of Life Sciences, University of Manchester, Manchester, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Matzke AJM, Weiger TM, Matzke M. Ion channels at the nucleus: electrophysiology meets the genome. MOLECULAR PLANT 2010; 3:642-52. [PMID: 20410254 PMCID: PMC2910552 DOI: 10.1093/mp/ssq013] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2010] [Accepted: 03/12/2010] [Indexed: 05/21/2023]
Abstract
The nuclear envelope is increasingly viewed from an electrophysiological perspective by researchers interested in signal transduction pathways that influence gene transcription and other processes in the nucleus. Here, we describe evidence for ion channels and transporters in the nuclear membranes and for possible ion gating by the nuclear pores. We argue that a systems-level understanding of cellular regulation is likely to require the assimilation of nuclear electrophysiology into molecular and biochemical signaling pathways.
Collapse
Affiliation(s)
- Antonius J M Matzke
- Gregor Mendel Institute of Molecular Plant Biology, Austrian Academy of Sciences, Dr. Bohr-Gasse 3, A-1030 Vienna, Austria.
| | | | | |
Collapse
|
26
|
Albrecht D. Physiological and pathophysiological functions of different angiotensins in the brain. Br J Pharmacol 2010. [DOI: 10.1111/j.1476-5381.2010.00648.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
|