351
|
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: 225] [Impact Index Per Article: 32.1] [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
|
352
|
Weber MD, Godbout JP, Sheridan JF. Repeated Social Defeat, Neuroinflammation, and Behavior: Monocytes Carry the Signal. Neuropsychopharmacology 2017; 42:46-61. [PMID: 27319971 PMCID: PMC5143478 DOI: 10.1038/npp.2016.102] [Citation(s) in RCA: 198] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 04/28/2016] [Accepted: 05/27/2016] [Indexed: 02/06/2023]
Abstract
Mounting evidence indicates that proinflammatory signaling in the brain affects mood, cognition, and behavior and is linked with the etiology of psychiatric disorders, including anxiety and depression. The purpose of this review is to focus on stress-induced bidirectional communication pathways between the central nervous system (CNS) and peripheral immune system that converge to promote a heightened neuroinflammatory environment. These communication pathways involve sympathetic outflow from the brain to the peripheral immune system that biases hematopoietic stem cells to differentiate into a glucocorticoid-resistant and primed myeloid lineage immune cell. In conjunction, microglia-dependent neuroinflammatory events promote myeloid cell trafficking to the brain that reinforces stress-related behavior, and is argued to play a role in stress-related psychiatric disorders. We will discuss evidence implicating a key role for endothelial cells that comprise the blood-brain barrier in propagating peripheral-to-central immune communication. We will also discuss novel neuron-to-glia communication pathways involving endogenous danger signals that have recently been argued to facilitate neuroinflammation under various conditions, including stress. These findings help elucidate the complex communication that occurs in response to stress and highlight novel therapeutic targets against the development of stress-related psychiatric disorders.
Collapse
Affiliation(s)
- Michael D Weber
- Division of Biosciences, The Ohio State University, Columbus, OH, USA,Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA,Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA,Division of Biosciences, The Ohio State University, 223 IBMR Building, 305 W 12th Avenue, 460 Medical Center Drive, Columbus, OH 43210, USA, Tel: 614-293-3392, Fax: 614-292-6087, E-mail:
| | - Jonathan P Godbout
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA,Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA,Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - John F Sheridan
- Division of Biosciences, The Ohio State University, Columbus, OH, USA,Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA,Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
353
|
Haim A, Julian D, Albin-Brooks C, Brothers HM, Lenz KM, Leuner B. A survey of neuroimmune changes in pregnant and postpartum female rats. Brain Behav Immun 2017; 59:67-78. [PMID: 27686844 DOI: 10.1016/j.bbi.2016.09.026] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 09/12/2016] [Accepted: 09/22/2016] [Indexed: 12/13/2022] Open
Abstract
During pregnancy and the postpartum period, the adult female brain is remarkably plastic exhibiting modifications of neurons, astrocytes and oligodendrocytes. However, little is known about how microglia, the brain's innate immune cells, are altered during this time. In the current studies, microglial density, number and morphological phenotype were analyzed within multiple regions of the maternal brain that are known to show neural plasticity during the peripartum period and/or regulate peripartum behavioral changes. Our results show a significant reduction in microglial density during late pregnancy and the early-mid postpartum period in the basolateral amygdala, medial prefrontal cortex, nucleus accumbens shell and dorsal hippocampus. In addition, microglia numbers were reduced postpartum in all four brain regions, and these reductions occurred primarily in microglia with a thin, ramified morphology. Across the various measures, microglia in the motor cortex were unaffected by reproductive status. The peripartum decrease in microglia may be a consequence of reduced proliferation as there were fewer numbers of proliferating microglia, and no changes in apoptotic microglia, in the postpartum hippocampus. Finally, hippocampal concentrations of the cytokines interleukin (IL)-6 and IL-10 were increased postpartum. Together, these data point to a shift in the maternal neuroimmune environment during the peripartum period that could contribute to neural and behavioral plasticity occurring during the transition to motherhood.
Collapse
Affiliation(s)
- Achikam Haim
- Department of Neuroscience, The Ohio State University, 333 West 10th Avenue, Columbus, OH 43210, USA
| | - Dominic Julian
- Department of Psychology, The Ohio State University, 1835 Neil Avenue, Columbus, OH 43210, USA
| | | | - Holly M Brothers
- Department of Psychology, The Ohio State University, 1835 Neil Avenue, Columbus, OH 43210, USA
| | - Kathryn M Lenz
- Department of Neuroscience, The Ohio State University, 333 West 10th Avenue, Columbus, OH 43210, USA; Department of Psychology, The Ohio State University, 1835 Neil Avenue, Columbus, OH 43210, USA; Behavioral Neuroendocrinology Group, The Ohio State University, Columbus, OH 43210, USA
| | - Benedetta Leuner
- Department of Neuroscience, The Ohio State University, 333 West 10th Avenue, Columbus, OH 43210, USA; Department of Psychology, The Ohio State University, 1835 Neil Avenue, Columbus, OH 43210, USA; Behavioral Neuroendocrinology Group, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
354
|
Dinan TG, Cryan JF. Microbes, Immunity, and Behavior: Psychoneuroimmunology Meets the Microbiome. Neuropsychopharmacology 2017; 42:178-192. [PMID: 27319972 PMCID: PMC5143479 DOI: 10.1038/npp.2016.103] [Citation(s) in RCA: 138] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 05/26/2016] [Accepted: 06/13/2016] [Indexed: 02/07/2023]
Abstract
There is now a large volume of evidence to support the view that the immune system is a key communication pathway between the gut and brain, which plays an important role in stress-related psychopathologies and thus provides a potentially fruitful target for psychotropic intervention. The gut microbiota is a complex ecosystem with a diverse range of organisms and a sophisticated genomic structure. Bacteria within the gut are estimated to weigh in excess of 1 kg in the adult human and the microbes within not only produce antimicrobial peptides, short chain fatty acids, and vitamins, but also most of the common neurotransmitters found in the human brain. That the microbial content of the gut plays a key role in immune development is now beyond doubt. Early disruption of the host-microbe interplay can have lifelong consequences, not just in terms of intestinal function but in distal organs including the brain. It is clear that the immune system and nervous system are in continuous communication in order to maintain a state of homeostasis. Significant gaps in knowledge remain about the effect of the gut microbiota in coordinating the immune-nervous systems dialogue. However, studies using germ-free animals, infective models, prebiotics, probiotics, and antibiotics have increased our understanding of the interplay. Early life stress can have a lifelong impact on the microbial content of the intestine and permanently alter immune functioning. That early life stress can also impact adult psychopathology has long been appreciated in psychiatry. The challenge now is to fully decipher the molecular mechanisms that link the gut microbiota, immune, and central nervous systems in a network of communication that impacts behavior patterns and psychopathology, to eventually translate these findings to the human situation both in health and disease. Even at this juncture, there is evidence to pinpoint key sites of communication where gut microbial interventions either with drugs or diet or perhaps fecal microbiota transplantation may positively impact mental health.
Collapse
Affiliation(s)
- Timothy G Dinan
- APC Microbiome Institute, University College Cork, Cork, Ireland
- Department of Psychiatry & Neurobehavioural Sciences, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Institute, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| |
Collapse
|
355
|
Colle R, de Larminat D, Rotenberg S, Hozer F, Hardy P, Verstuyft C, Fève B, Corruble E. Pioglitazone could induce remission in major depression: a meta-analysis. Neuropsychiatr Dis Treat 2017; 13:9-16. [PMID: 28031713 PMCID: PMC5182046 DOI: 10.2147/ndt.s121149] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Pioglitazone, a selective agonist of the nuclear transcription factor peroxisome proliferator-activated receptor-gamma (PPAR-γ), prescribed for the treatment of type 2 diabetes, could have antidepressant properties. However, its potential to induce remission of major depressive episodes, the optimal clinical target for an antidepressant drug, is a matter of concern. Indeed, only one out of four double-blind randomized controlled trials show higher remission rates with pioglitazone than with control treatments. Hence, the main aim of this study was to perform a meta-analysis of the efficacy of pioglitazone for the treatment of MDE, focusing on remission rates. METHODS Four double-blind randomized controlled trials, comprising 161 patients with an MDE, were included in this meta-analysis. Pioglitazone was studied either alone (one study) or as add-on therapy to conventional treatments (antidepressant drugs or lithium salts). It was compared either to placebo (three studies) or to metformin (one study). Remission was defined by a Hamilton Depression Rating Scale score <8 after treatment. RESULTS Pioglitazone could induce higher remission rates than control treatments (27% versus 10%, I2=17.3%, fixed-effect model: odds ratio [OR] =3.3, 95% confidence interval [95% CI; 1.4; 7.8], P=0.008). The OR was even higher in the subgroup of patients with major depressive disorder (n=80; 23% versus 8%, I2=0.0%; fixed-effect model: OR =5.9, 95% CI [1.6; 22.4], P=0.009) and in the subgroup of patients without metabolic comorbidities (n=84; 33% versus 10%, I2=0.0%; fixed-effect model: OR =5.1, 95% CI [1.5; 17.9], P=0.01). As compared to control treatments, results suggest six patients would need to be treated with pioglitazone in order to achieve the possibility of one more remission. CONCLUSION Pioglitazone, either alone or as add-on therapy to conventional treatments, could induce remission of MDE, suggesting that drugs with PPAR-γ agonist properties may be true and clinically relevant antidepressants, even in patients without metabolic comorbidities.
Collapse
Affiliation(s)
- Romain Colle
- Psychiatry Department, Hôpital Bicêtre, INSERM, UMR S1178, University Paris-Sud, Assistance Publique-Hôpitaux de Paris, Le Kremlin Bicêtre, France
| | - Delphine de Larminat
- Psychiatry Department, Hôpital Bicêtre, INSERM, UMR S1178, University Paris-Sud, Assistance Publique-Hôpitaux de Paris, Le Kremlin Bicêtre, France
| | - Samuel Rotenberg
- Psychiatry Department, Hôpital Bicêtre, INSERM, UMR S1178, University Paris-Sud, Assistance Publique-Hôpitaux de Paris, Le Kremlin Bicêtre, France
| | - Franz Hozer
- Psychiatry Department, Hôpital Bicêtre, INSERM, UMR S1178, University Paris-Sud, Assistance Publique-Hôpitaux de Paris, Le Kremlin Bicêtre, France
| | - Patrick Hardy
- Psychiatry Department, Hôpital Bicêtre, INSERM, UMR S1178, University Paris-Sud, Assistance Publique-Hôpitaux de Paris, Le Kremlin Bicêtre, France
| | - Céline Verstuyft
- Molecular Genetic, Pharmacogenetics and Hormonology Department, Hôpital Bicêtre, INSERM UMR_S1184, Centre IMVA, University Paris-Sud, Assistance Publique-Hôpitaux de Paris, Le Kremlin Bicêtre, France
| | - Bruno Fève
- Endocrinology Department, INSERM UMR_S938, Hôpital Saint-Antoine, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire ICAN, Sorbonne Universités, Université Pierre et Marie Curie, Assistance Publique des Hôpitaux de Paris, Paris, France
| | - Emmanuelle Corruble
- Psychiatry Department, Hôpital Bicêtre, INSERM, UMR S1178, University Paris-Sud, Assistance Publique-Hôpitaux de Paris, Le Kremlin Bicêtre, France
| |
Collapse
|
356
|
Glial and Neuroimmune Mechanisms as Critical Modulators of Drug Use and Abuse. Neuropsychopharmacology 2017; 42:156-177. [PMID: 27402494 PMCID: PMC5143481 DOI: 10.1038/npp.2016.121] [Citation(s) in RCA: 187] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 06/24/2016] [Accepted: 06/28/2016] [Indexed: 12/26/2022]
Abstract
Drugs of abuse cause persistent alterations in synaptic plasticity that may underlie addiction behaviors. Evidence suggests glial cells have an essential and underappreciated role in the development and maintenance of drug abuse by influencing neuronal and synaptic functions in multifaceted ways. Microglia and astrocytes perform critical functions in synapse formation and refinement in the developing brain, and there is growing evidence that disruptions in glial function may be implicated in numerous neurological disorders throughout the lifespan. Linking evidence of function in health and under pathological conditions, this review will outline the glial and neuroimmune mechanisms that may contribute to drug-abuse liability, exploring evidence from opioids, alcohol, and psychostimulants. Drugs of abuse can activate microglia and astrocytes through signaling at innate immune receptors, which in turn influence neuronal function not only through secretion of soluble factors (eg, cytokines and chemokines) but also potentially through direct remodeling of the synapses. In sum, this review will argue that neural-glial interactions represent an important avenue for advancing our understanding of substance abuse disorders.
Collapse
|
357
|
Therapeutic Implications of Brain-Immune Interactions: Treatment in Translation. Neuropsychopharmacology 2017; 42:334-359. [PMID: 27555382 PMCID: PMC5143492 DOI: 10.1038/npp.2016.167] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 07/22/2016] [Accepted: 08/17/2016] [Indexed: 02/06/2023]
Abstract
A wealth of data has been amassed that details a complex, yet accessible, series of pathways by which the immune system, notably inflammation, can influence the brain and behavior. These data have opened the window to a diverse array of novel targets whose potential efficacy is tied to specific neurotransmitters and neurocircuits as well as specific behaviors. What is clear is that the impact of inflammation on the brain cuts across psychiatric disorders and engages dopaminergic and glutamatergic pathways that regulate motivation and motor activity as well as the sensitivity to threat. Given the ability to identify patient populations with increased inflammation, the precision of interventions can be further tuned, in conjunction with the ability to establish target engagement in the brain through the use of multiple neuroimaging strategies. After a brief overview of the mechanisms by which inflammation affects the brain and behavior, this review examines the extant literature on the efficacy of anti-inflammatory treatments, while forging guidelines for future intelligent clinical trial design. An examination of the most promising therapeutic strategies is also provided, along with some of the most exciting clinical trials that are currently being planned or underway.
Collapse
|
358
|
Birur B, Shelton RC, Amrock EM, Li L. Response to: Commentary: Sex Differences in the Peripheral Immune System in Patients with Depression. Front Psychiatry 2017; 8:231. [PMID: 29176954 PMCID: PMC5686389 DOI: 10.3389/fpsyt.2017.00231] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 10/30/2017] [Indexed: 11/13/2022] Open
Affiliation(s)
- Badari Birur
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Richard C Shelton
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Ellen M Amrock
- Birmingham VA Medical Center (VHA), Birmingham, AL, United States
| | - Li Li
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
359
|
Haroon E, Miller AH. Inflammation Effects on Brain Glutamate in Depression: Mechanistic Considerations and Treatment Implications. Curr Top Behav Neurosci 2017; 31:173-198. [PMID: 27830574 DOI: 10.1007/7854_2016_40] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
There has been increasing interest in the role of glutamate in mood disorders, especially given the profound effect of the glutamate receptor antagonist ketamine in improving depressive symptoms in patients with treatment-resistant depression. One pathway by which glutamate alterations may occur in mood disorders involves inflammation. Increased inflammation has been observed in a significant subgroup of patients with mood disorders, and inflammatory cytokines have been shown to influence glutamate metabolism through effects on astrocytes and microglia. In addition, the administration of the inflammatory cytokine interferon-alpha has been shown to increase brain glutamate in the basal ganglia and dorsal anterior cingulate cortex as measured by magnetic resonance spectroscopy (MRS). Moreover, MRS studies in patients with major depressive disorder have revealed that increased markers of inflammation including C-reactive protein correlate with increased basal ganglia glutamate, which in turn was associated with anhedonia and psychomotor retardation. Finally, human and laboratory animal studies have shown that the response to glutamate antagonists such as ketamine is predicted by increased inflammatory cytokines. Taken together, these data make a strong case that inflammation may influence glutamate metabolism to alter behavior, leading to depressive symptoms including anhedonia and psychomotor slowing.
Collapse
Affiliation(s)
- Ebrahim Haroon
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, 1365-B Clifton Road., 5th Floor, B5101, Atlanta, GA, 30322, USA
| | - Andrew H Miller
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, 1365-B Clifton Road., 5th Floor, B5101, Atlanta, GA, 30322, USA.
| |
Collapse
|
360
|
Toepfer P, Heim C, Entringer S, Binder E, Wadhwa P, Buss C. Oxytocin pathways in the intergenerational transmission of maternal early life stress. Neurosci Biobehav Rev 2016; 73:293-308. [PMID: 28027955 DOI: 10.1016/j.neubiorev.2016.12.026] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 12/08/2016] [Accepted: 12/17/2016] [Indexed: 12/17/2022]
Abstract
Severe stress in early life, such as childhood abuse and neglect, constitutes a major risk factor in the etiology of psychiatric disorders and somatic diseases. Importantly, these long-term effects may impact the next generation. The intergenerational transmission of maternal early life stress (ELS) may occur via pre-and postnatal pathways, such as alterations in maternal-fetal-placental stress physiology, maternal depression during pregnancy and postpartum, as well as impaired mother-offspring interactions. The neuropeptide oxytocin (OT) has gained considerable attention for its role in modulating all of these assumed transmission pathways. Moreover, central and peripheral OT signaling pathways are highly sensitive to environmental exposures and may be compromised by ELS with implications for these putative transmission mechanisms. Together, these data suggest that OT pathways play an important role in the intergenerational transmission of maternal ELS in humans. By integrating recent studies on gene-environment interactions and epigenetic modifications in OT pathway genes, the present review aims to develop a conceptual framework of intergenerational transmission of maternal ELS that emphasizes the role of OT.
Collapse
Affiliation(s)
- Philipp Toepfer
- Institute of Medical Psychology, Charité - Universitätsmedizin Berlin, Luisenstraße 57, 10117, Berlin, Germany
| | - Christine Heim
- Institute of Medical Psychology, Charité - Universitätsmedizin Berlin, Luisenstraße 57, 10117, Berlin, Germany; Department of Biobehavioral Health, Penn State University, 219 Biobehavioral Health Building University Park, PA, 16802, USA
| | - Sonja Entringer
- Institute of Medical Psychology, Charité - Universitätsmedizin Berlin, Luisenstraße 57, 10117, Berlin, Germany; UC Irvine Development, Health and Disease Research Program, 333 The City Blvd. W, Suite 810, Orange, CA, 92868, USA
| | - Elisabeth Binder
- Max-Planck Institute of Psychiatry, Kraepelinstr. 2-10, 80804, Munich, Germany; Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, School of Medicine Atlanta, GA, 30307, USA
| | - Pathik Wadhwa
- UC Irvine Development, Health and Disease Research Program, 333 The City Blvd. W, Suite 810, Orange, CA, 92868, USA
| | - Claudia Buss
- Institute of Medical Psychology, Charité - Universitätsmedizin Berlin, Luisenstraße 57, 10117, Berlin, Germany; UC Irvine Development, Health and Disease Research Program, 333 The City Blvd. W, Suite 810, Orange, CA, 92868, USA.
| |
Collapse
|
361
|
Pfau ML, Purushothaman I, Feng J, Golden SA, Aleyasin H, Lorsch ZS, Cates HM, Flanigan ME, Menard C, Heshmati M, Wang Z, Ma'ayan A, Shen L, Hodes GE, Russo SJ. Integrative Analysis of Sex-Specific microRNA Networks Following Stress in Mouse Nucleus Accumbens. Front Mol Neurosci 2016; 9:144. [PMID: 28066174 PMCID: PMC5179560 DOI: 10.3389/fnmol.2016.00144] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 11/28/2016] [Indexed: 12/21/2022] Open
Abstract
Adult women are twice as likely as men to suffer from affective and anxiety disorders, although the mechanisms underlying heightened female stress susceptibility are incompletely understood. Recent findings in mouse Nucleus Accumbens (NAc) suggest a role for DNA methylation-driven sex differences in genome-wide transcriptional profiles. However, the role of another epigenetic process-microRNA (miR) regulation-has yet to be explored. We exposed male and female mice to Subchronic Variable Stress (SCVS), a stress paradigm that produces depression-like behavior in female, but not male, mice, and performed next generation mRNA and miR sequencing on NAc tissue. We applied a combination of differential expression, miR-mRNA network and functional enrichment analyses to characterize the transcriptional and post-transcriptional landscape of sex differences in NAc stress response. We find that male and female mice exhibit largely non-overlapping miR and mRNA profiles following SCVS. The two sexes also show enrichment of different molecular pathways and functions. Collectively, our results suggest that males and females mount fundamentally different transcriptional and post-transcriptional responses to SCVS and engage sex-specific molecular processes following stress. These findings have implications for the pathophysiology and treatment of stress-related disorders in women.
Collapse
Affiliation(s)
- Madeline L Pfau
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount SinaiNew York, NY, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount SinaiNew York, NY, USA
| | - Immanuel Purushothaman
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai New York, NY, USA
| | - Jian Feng
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai New York, NY, USA
| | - Sam A Golden
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount SinaiNew York, NY, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount SinaiNew York, NY, USA
| | - Hossein Aleyasin
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai New York, NY, USA
| | - Zachary S Lorsch
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount SinaiNew York, NY, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount SinaiNew York, NY, USA
| | - Hannah M Cates
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount SinaiNew York, NY, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount SinaiNew York, NY, USA
| | - Meghan E Flanigan
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount SinaiNew York, NY, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount SinaiNew York, NY, USA
| | - Caroline Menard
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai New York, NY, USA
| | - Mitra Heshmati
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount SinaiNew York, NY, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount SinaiNew York, NY, USA
| | - Zichen Wang
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount SinaiNew York, NY, USA; Department of Pharmacology and Systems Therapeutics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount SinaiNew York, NY, USA
| | - Avi Ma'ayan
- Department of Pharmacology and Systems Therapeutics, BD2K-LINCS Data Coordination and Integration Center, Icahn School of Medicine at Mount Sinai New York, NY, USA
| | - Li Shen
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai New York, NY, USA
| | - Georgia E Hodes
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai New York, NY, USA
| | - Scott J Russo
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai New York, NY, USA
| |
Collapse
|
362
|
Wohleb ES. Neuron-Microglia Interactions in Mental Health Disorders: "For Better, and For Worse". Front Immunol 2016; 7:544. [PMID: 27965671 PMCID: PMC5126117 DOI: 10.3389/fimmu.2016.00544] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 11/16/2016] [Indexed: 12/13/2022] Open
Abstract
Persistent cognitive and behavioral symptoms that characterize many mental health disorders arise from impaired neuroplasticity in several key corticolimbic brain regions. Recent evidence suggests that reciprocal neuron–microglia interactions shape neuroplasticity during physiological conditions, implicating microglia in the neurobiology of mental health disorders. Neuron–microglia interactions are modulated by several molecular and cellular pathways, and dysregulation of these pathways often have neurobiological consequences, including aberrant neuronal responses and microglia activation. Impaired neuron-microglia interactions are implicated in mental health disorders because rodent stress models lead to concomitant neuronal dystrophy and alterations in microglia morphology and function. In this context, functional changes in microglia may be indicative of an immune state termed parainflammation in which tissue-resident macrophages (i.e., microglia) respond to malfunctioning cells by initiating modest inflammation in an attempt to restore homeostasis. Thus, aberrant neuronal activity and release of damage-associated signals during repeated stress exposure may contribute to functional changes in microglia and resultant parainflammation. Furthermore, accumulating evidence shows that uncoupling neuron–microglia interactions may contribute to altered neuroplasticity and associated anxiety- or depressive-like behaviors. Additional work shows that microglia have varied phenotypes in specific brain regions, which may underlie divergent neuroplasticity observed in corticolimbic structures following stress exposure. These findings indicate that neuron–microglia interactions are critical mediators of the interface between adaptive, homeostatic neuronal function and the neurobiology of mental health disorders.
Collapse
Affiliation(s)
- Eric S Wohleb
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA; Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
363
|
Arango-Lievano M, Peguet C, Catteau M, Parmentier ML, Wu S, Chao MV, Ginsberg SD, Jeanneteau F. Deletion of Neurotrophin Signaling through the Glucocorticoid Receptor Pathway Causes Tau Neuropathology. Sci Rep 2016; 6:37231. [PMID: 27849045 PMCID: PMC5110980 DOI: 10.1038/srep37231] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 10/26/2016] [Indexed: 01/29/2023] Open
Abstract
Glucocorticoid resistance is a risk factor for Alzheimer's disease (AD). Molecular and cellular mechanisms of glucocorticoid resistance in the brain have remained unknown and are potential therapeutic targets. Phosphorylation of glucocorticoid receptors (GR) by brain-derived neurotrophic factor (BDNF) signaling integrates both pathways for remodeling synaptic structure and plasticity. The goal of this study is to test the role of the BDNF-dependent pathway on glucocorticoid signaling in a mouse model of glucocorticoid resistance. We report that deletion of GR phosphorylation at BDNF-responding sites and downstream signaling via the MAPK-phosphatase DUSP1 triggers tau phosphorylation and dendritic spine atrophy in mouse cortex. In human cortex, DUSP1 protein expression correlates with tau phosphorylation, synaptic defects and cognitive decline in subjects diagnosed with AD. These findings provide evidence for a causal role of BDNF-dependent GR signaling in tau neuropathology and indicate that DUSP1 is a potential target for therapeutic interventions.
Collapse
Affiliation(s)
- Margarita Arango-Lievano
- Inserm, U1191, Institute of Functional Genomics, F-34000 Montpellier, France
- CNRS, UMR-5203, F-34000 Montpellier, France
- Université de Montpellier, F-34000 Montpellier, France
| | - Camille Peguet
- Inserm, U1191, Institute of Functional Genomics, F-34000 Montpellier, France
- CNRS, UMR-5203, F-34000 Montpellier, France
- Université de Montpellier, F-34000 Montpellier, France
| | - Matthias Catteau
- Inserm, U1191, Institute of Functional Genomics, F-34000 Montpellier, France
- CNRS, UMR-5203, F-34000 Montpellier, France
- Université de Montpellier, F-34000 Montpellier, France
| | - Marie-Laure Parmentier
- Inserm, U1191, Institute of Functional Genomics, F-34000 Montpellier, France
- CNRS, UMR-5203, F-34000 Montpellier, France
- Université de Montpellier, F-34000 Montpellier, France
| | - Synphen Wu
- Skirball Institute of biomolecular medicine, New York University Langone Medical Center, New York, NY 10016, USA
| | - Moses V Chao
- Skirball Institute of biomolecular medicine, New York University Langone Medical Center, New York, NY 10016, USA
| | - Stephen D. Ginsberg
- Center for Dementia Research, Nathan Kline Institute, Departments of Psychiatry, Neuroscience & Physiology, New York University Langone Medical Center, Orangeburg, NY 10962, USA
| | - Freddy Jeanneteau
- Inserm, U1191, Institute of Functional Genomics, F-34000 Montpellier, France
- CNRS, UMR-5203, F-34000 Montpellier, France
- Université de Montpellier, F-34000 Montpellier, France
| |
Collapse
|
364
|
Aizenstein HJ, Baskys A, Boldrini M, Butters MA, Diniz BS, Jaiswal MK, Jellinger KA, Kruglov LS, Meshandin IA, Mijajlovic MD, Niklewski G, Pospos S, Raju K, Richter K, Steffens DC, Taylor WD, Tene O. Vascular depression consensus report - a critical update. BMC Med 2016; 14:161. [PMID: 27806704 PMCID: PMC5093970 DOI: 10.1186/s12916-016-0720-5] [Citation(s) in RCA: 145] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Accepted: 10/14/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Vascular depression is regarded as a subtype of late-life depression characterized by a distinct clinical presentation and an association with cerebrovascular damage. Although the term is commonly used in research settings, widely accepted diagnostic criteria are lacking and vascular depression is absent from formal psychiatric manuals such as the Diagnostic and Statistical Manual of Mental Disorders, 5th edition - a fact that limits its use in clinical settings. Magnetic resonance imaging (MRI) techniques, showing a variety of cerebrovascular lesions, including extensive white matter hyperintensities, subcortical microvascular lesions, lacunes, and microinfarcts, in patients with late life depression, led to the introduction of the term "MRI-defined vascular depression". DISCUSSION This diagnosis, based on clinical and MRI findings, suggests that vascular lesions lead to depression by disruption of frontal-subcortical-limbic networks involved in mood regulation. However, despite multiple MRI approaches to shed light on the spatiotemporal structural changes associated with late life depression, the causal relationship between brain changes, related lesions, and late life depression remains controversial. While postmortem studies of elderly persons who died from suicide revealed lacunes, small vessel, and Alzheimer-related pathologies, recent autopsy data challenged the role of these lesions in the pathogenesis of vascular depression. Current data propose that the vascular depression connotation should be reserved for depressed older patients with vascular pathology and evident cerebral involvement. Based on current knowledge, the correlations between intra vitam neuroimaging findings and their postmortem validity as well as the role of peripheral markers of vascular disease in late life depression are discussed. CONCLUSION The multifold pathogenesis of vascular depression as a possible subtype of late life depression needs further elucidation. There is a need for correlative clinical, intra vitam structural and functional MRI as well as postmortem MRI and neuropathological studies in order to confirm the relationship between clinical symptomatology and changes in specific brain regions related to depression. To elucidate the causal relationship between regional vascular brain changes and vascular depression, animal models could be helpful. Current treatment options include a combination of vasoactive drugs and antidepressants, but the outcomes are still unsatisfying.
Collapse
Affiliation(s)
- Howard J Aizenstein
- Department of Psychiatry, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Andrius Baskys
- Memory Disorders Clinic, Riverside Psychiatric Medical Group, Riverside, CA, USA
| | - Maura Boldrini
- Department of Psychiatry, Columbia University, New York, NY, USA.,Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, NY, USA
| | - Meryl A Butters
- Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, PA, USA
| | - Breno S Diniz
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Manoj Kumar Jaiswal
- Department of Psychiatry, Columbia University, New York, NY, USA.,Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, Columbia University, New York, NY, USA
| | - Kurt A Jellinger
- Institute of Clinical Neurobiology, Alberichgasse 5/13, Vienna, A-1150, Austria.
| | - Lev S Kruglov
- Department of Geriatric Psychiatry of the St. Petersburg Psychoneurological Research Institute named after V. M. Bekhterev, Medical Faculty of St. Petersburg University, St. Petersburg, Russia
| | - Ivan A Meshandin
- Clinical Department, Scientific and Practical Center of Psychoneurology named after V. M. Soloviev, St. Petersburg, Russia
| | - Milija D Mijajlovic
- Neurology Clinic, Clinical Center of Serbia, School of Medicine University of Belgrade, Belgrade, Serbia
| | - Guenter Niklewski
- University Clinic for Psychiatry and Psychotherapy, Paracelsus Private Medical University, Nuremberg, Germany
| | - Sarah Pospos
- Memory Disorders Clinic, Riverside Psychiatric Medical Group, Riverside, CA, USA
| | - Keerthy Raju
- Consultant in Old Age Psychiatry, Cheshire and Wirral Partnership NHS Foundation Trust, Chester, UK
| | - Kneginja Richter
- University Clinic for Psychiatry and Psychotherapy, Paracelsus Private Medical University, Nuremberg, Germany.,Faculty for Social Sciences, Technical University of Nuremberg Georg Simon Ohm, Nuremberg, Germany
| | - David C Steffens
- Department of Psychiatry, University of Connecticut Health Center, Farmington, CT, USA
| | - Warren D Taylor
- Department of Psychiatry, The Center for Cognitive Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Veterans Affairs Medical Center, The Geriatric Research, Education, and Clinical Center (GRECC), Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Oren Tene
- Departments of Neurology and Psychiatry, Tel Aviv Medical Center, Tel Aviv, Israel.,Tel Aviv University, Sackler Faculty of Medicine, Tel Aviv, Israel
| |
Collapse
|
365
|
Jeltsch-David H, Muller S. Autoimmunity, neuroinflammation, pathogen load: A decisive crosstalk in neuropsychiatric SLE. J Autoimmun 2016; 74:13-26. [DOI: 10.1016/j.jaut.2016.04.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Revised: 04/21/2016] [Accepted: 04/24/2016] [Indexed: 12/23/2022]
|
366
|
Milosevic A, Liebmann T, Knudsen M, Schintu N, Svenningsson P, Greengard P. Cell- and region-specific expression of depression-related protein p11 (S100a10) in the brain. J Comp Neurol 2016; 525:955-975. [PMID: 27616678 PMCID: PMC5222728 DOI: 10.1002/cne.24113] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 08/03/2016] [Accepted: 08/09/2016] [Indexed: 12/20/2022]
Abstract
P11 (S100a10), a member of the S100 family of proteins, has widespread distribution in the vertebrate body, including in the brain, where it has a key role in membrane trafficking, vesicle secretion, and endocytosis. Recently, our laboratory has shown that a constitutive knockout of p11 (p11-KO) in mice results in a depressive-like phenotype. Furthermore, p11 has been implicated in major depressive disorder (MDD) and in the actions of antidepressants. Since depression affects multiple brain regions, and the role of p11 has only been determined in a few of these areas, a detailed analysis of p11 expression in the brain is warranted. Here we demonstrate that, although widespread in the brain, p11 expression is restricted to distinct regions, and specific neuronal and nonneuronal cell types. Furthermore, we provide comprehensive mapping of p11 expression using in situ hybridization, immunocytochemistry, and whole-tissue volume imaging. Overall, expression spans multiple brain regions, structures, and cell types, suggesting a complex role of p11 in depression. J. Comp. Neurol. 525:955-975, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ana Milosevic
- Laboratory of Molecular and Cellular Neuroscience, Rockefeller University, New York, New York, USA
| | - Thomas Liebmann
- Laboratory of Molecular and Cellular Neuroscience, Rockefeller University, New York, New York, USA
| | - Margarete Knudsen
- Laboratory of Molecular and Cellular Neuroscience, Rockefeller University, New York, New York, USA
| | - Nicoletta Schintu
- Section for Translational Neuropharmacology, Department of Clinical Neuroscience, CMM L8:01, Karolinska Institutet, 17176 Stockholm, Sweden
| | - Per Svenningsson
- Section for Translational Neuropharmacology, Department of Clinical Neuroscience, CMM L8:01, Karolinska Institutet, 17176 Stockholm, Sweden
| | - Paul Greengard
- Laboratory of Molecular and Cellular Neuroscience, Rockefeller University, New York, New York, USA
| |
Collapse
|
367
|
Carboni L, Nguyen TP, Caberlotto L. Systems biology integration of proteomic data in rodent models of depression reveals involvement of the immune response and glutamatergic signaling. Proteomics Clin Appl 2016; 10:1254-1263. [DOI: 10.1002/prca.201500149] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 08/31/2016] [Accepted: 09/07/2016] [Indexed: 01/29/2023]
Affiliation(s)
- Lucia Carboni
- Department of Pharmacy and Biotechnology; Alma Mater Studiorum University of Bologna; Bologna Italy
| | | | - Laura Caberlotto
- Centre for Computational and Systems Biology (COSBI); The Microsoft Research-University of Trento; Trento Italy
- Aptuit (Verona); Verona Italy
| |
Collapse
|
368
|
Huang T, Zhou F, Wang-Johanning F, Nan K, Wei Y. Depression accelerates the development of gastric cancer through reactive oxygen species‑activated ABL1 (Review). Oncol Rep 2016; 36:2435-2443. [PMID: 27666407 DOI: 10.3892/or.2016.5127] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 09/05/2016] [Indexed: 11/06/2022] Open
Abstract
Depression is a common symptom among gastric cancer (GC) patients and serves as a potential indication of poor prognosis and advanced cancer clinical stage. However, the molecular mechanism of depression‑associated poor prognoses of GC patients remains unclear. Recent studies have revealed that GC patients with depression are under high levels of oxidative stress (OS) status that is accompanied by the dysfunction of numerous proto‑oncogenes, including the ABL proto‑oncogene 1 (ABL1), which is a non‑receptor tyrosine kinase. Recent evidence indicates that ABL1 was dysregulated in both major depressive disorder (MDD) and cancer patients with depression, and high levels of reactive oxygen species (ROS) can lead to the activation of ABL1 in response to OS and that activated ABL1 subsequently contributes to development of GC via interactions with the downstream targets and corresponding signaling pathways. In this review, we examine the evidence to illuminate the molecular mechanism of ABL1 in the progression of GC patients with depression and identify out new and effective methods for the initial and long‑term treatment of GC.
Collapse
Affiliation(s)
- Tianhe Huang
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Fuling Zhou
- Department of Clinical Hematology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | | | - Kejun Nan
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yongchang Wei
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
369
|
Montesinos J, Alfonso-Loeches S, Guerri C. Impact of the Innate Immune Response in the Actions of Ethanol on the Central Nervous System. Alcohol Clin Exp Res 2016; 40:2260-2270. [PMID: 27650785 DOI: 10.1111/acer.13208] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 08/02/2016] [Indexed: 12/25/2022]
Abstract
The innate immune response in the central nervous system (CNS) participates in both synaptic plasticity and neural damage. Emerging evidence from human and animal studies supports the role of the neuroimmune system response in many actions of ethanol (EtOH) on the CNS. Research studies have shown that alcohol stimulates brain immune cells, microglia, and astrocytes, by activating innate immune receptors Toll-like receptors (TLRs) and NOD-like receptors (inflammasome NLRs) triggering signaling pathways, which culminate in the production of pro-inflammatory cytokines and chemokines that lead to neuroinflammation. This review focuses on evidence that indicates the participation of TLRs and the inflammasome NLRs signaling response in many effects of EtOH on the CNS, such as neuroinflammation associated with brain damage, cognitive and behavioral dysfunction, and adolescent brain development alterations. It also reviews findings that indicate the role of TLR4-dependent signaling immune molecules in alcohol consumption, reward, and addiction. The research data suggest that overactivation of TLR4 or NLRs increases pro-inflammatory cytokines and mediators to cause neural damage in the cerebral cortex and hippocampus, while modest TLR4 activation, along with the generation of certain cytokines and chemokines in specific brain areas (e.g., amygdala, ventral tegmental area), modulate neurotransmission, alcohol drinking, and alcohol rewards. Elimination of TLR4 and NLRP3 abolishes many neuroimmune effects of EtOH. Despite much progress being made in this area, there are some research gaps and unanswered questions that this review discusses. Finally, potential therapies that target neuroimmune pathways to treat neuropathological and behavioral consequences of alcohol abuse are also evaluated.
Collapse
Affiliation(s)
- Jorge Montesinos
- Department of Molecular and Cellular Pathology of Alcohol, Príncipe Felipe Research Center, Valencia, Spain
| | - Silvia Alfonso-Loeches
- Department of Molecular and Cellular Pathology of Alcohol, Príncipe Felipe Research Center, Valencia, Spain
| | - Consuelo Guerri
- Department of Molecular and Cellular Pathology of Alcohol, Príncipe Felipe Research Center, Valencia, Spain.
| |
Collapse
|
370
|
Sharma A. Editorial (Thematic Selection: Inflammatory and Immune Responses in Depression). Curr Neuropharmacol 2016; 14:663-4. [PMID: 27640516 PMCID: PMC5050396 DOI: 10.2174/1570159x1407160826191654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Affiliation(s)
- Abhay Sharma
- CSIR-Institute of Genomics and Integrative Biology Council of Scientific and Industrial Research Sukhdev Vihar, Mathura Road New Delhi 110025
| |
Collapse
|
371
|
Abstract
Major depressive disorder (MDD) is a debilitating disease that is characterized by depressed mood, diminished interests, impaired cognitive function and vegetative symptoms, such as disturbed sleep or appetite. MDD occurs about twice as often in women than it does in men and affects one in six adults in their lifetime. The aetiology of MDD is multifactorial and its heritability is estimated to be approximately 35%. In addition, environmental factors, such as sexual, physical or emotional abuse during childhood, are strongly associated with the risk of developing MDD. No established mechanism can explain all aspects of the disease. However, MDD is associated with alterations in regional brain volumes, particularly the hippocampus, and with functional changes in brain circuits, such as the cognitive control network and the affective-salience network. Furthermore, disturbances in the main neurobiological stress-responsive systems, including the hypothalamic-pituitary-adrenal axis and the immune system, occur in MDD. Management primarily comprises psychotherapy and pharmacological treatment. For treatment-resistant patients who have not responded to several augmentation or combination treatment attempts, electroconvulsive therapy is the treatment with the best empirical evidence. In this Primer, we provide an overview of the current evidence of MDD, including its epidemiology, aetiology, pathophysiology, diagnosis and treatment.
Collapse
Affiliation(s)
- Christian Otte
- Department of Psychiatry and Psychotherapy, Charité University Medical Center, Campus Benjamin Franklin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Stefan M Gold
- Department of Psychiatry and Psychotherapy, Charité University Medical Center, Campus Benjamin Franklin, Hindenburgdamm 30, 12203 Berlin, Germany
- Institute of Neuroimmunology and Multiple Sclerosis (INIMS), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Brenda W Penninx
- Department of Psychiatry, VU University Medical Center, Amsterdam, The Netherlands
| | - Carmine M Pariante
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Amit Etkin
- Department of Psychiatry and Behavioural Sciences, Stanford University School of Medicine, Palo Alto, California, USA
| | - Maurizio Fava
- Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - David C Mohr
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Alan F Schatzberg
- Department of Psychiatry and Behavioural Sciences, Stanford University School of Medicine, Palo Alto, California, USA
| |
Collapse
|
372
|
GC-MS-based metabolomic study on the antidepressant-like effects of diterpene ginkgolides in mouse hippocampus. Behav Brain Res 2016; 314:116-24. [PMID: 27498146 DOI: 10.1016/j.bbr.2016.08.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 07/28/2016] [Accepted: 08/01/2016] [Indexed: 12/29/2022]
Abstract
Ginkgo biloba extract (GBE), including EGb-761, have been suggested to have antidepressant activity based on previous behavioral and biochemical analyses. However, because GBE contain many constituents, the mechanisms underlying this suggested antidepressant activity are unclear. Here, we investigated the antidepressant-like effects of diterpene ginkgolides (DG), an important class of constituents in GBE, and studied their effects in the mouse hippocampus using a GC-MS-based metabolomics approach. Mice were randomly divided into five groups and injected daily until testing with 0.9% NaCl solution, one of three doses of DG (4.06, 12.18, and 36.54mg/kg), or venlafaxine. Sucrose preference (SPT) and tail suspension (TST) tests were then performed to evaluate depressive-like behaviors in mice. DG (12.18 and 36.54mg/kg) and venlafaxine (VLX) administration significantly increased hedonic behavior in mice in the SPT. DG (12.18mg/kg) treatment also shortened immobility time in the TST, suggestive of antidepressant-like effects. Significant differences in the metabolic profile in the DG (12.18mg/kg) compared with the control or VLX group indicative of an antidepressant-like effect were observed using multivariate analysis. Eighteen differential hippocampal metabolites were identified that discriminated the DG (12.18mg/kg) and control groups. These biochemical changes involved neurotransmitter metabolism, oxidative stress, glutathione metabolism, lipid metabolism, energy metabolism, and kynurenic acid, providing clues to the therapeutic mechanisms of DG. Thus, this study showed that DG has antidepressant-like activities in mice and shed light on the biological mechanisms underlying the effects of diterpene ginkgolides on behavior, providing an important drug candidate for the treatment of depression.
Collapse
|
373
|
Mast cell activation disease and the modern epidemic of chronic inflammatory disease. Transl Res 2016; 174:33-59. [PMID: 26850903 DOI: 10.1016/j.trsl.2016.01.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Revised: 01/07/2016] [Accepted: 01/08/2016] [Indexed: 12/18/2022]
Abstract
A large and growing portion of the human population, especially in developed countries, suffers 1 or more chronic, often quite burdensome ailments which either are overtly inflammatory in nature or are suspected to be of inflammatory origin, but for which investigations to date have failed to identify specific causes, let alone unifying mechanisms underlying the multiple such ailments that often afflict such patients. Relatively recently described as a non-neoplastic cousin of the rare hematologic disease mastocytosis, mast cell (MC) activation syndrome-suspected to be of greatly heterogeneous, complex acquired clonality in many cases-is a potential underlying/unifying explanation for a diverse assortment of inflammatory ailments. A brief review of MC biology and how aberrant primary MC activation might lead to such a vast range of illness is presented.
Collapse
|
374
|
Becker KJ. Strain-Related Differences in the Immune Response: Relevance to Human Stroke. Transl Stroke Res 2016; 7:303-12. [PMID: 26860504 PMCID: PMC4929040 DOI: 10.1007/s12975-016-0455-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 01/31/2016] [Accepted: 02/02/2016] [Indexed: 02/07/2023]
Abstract
There are significant differences in the immune response and in the susceptibility to autoimmune diseases among rodent strains. It would thus be expected that the contribution of the immune response to cerebral ischemic injury would also differ among rodent strains. More importantly, there are significant differences between the immune responses of rodents and humans. All of these factors are likely to impact the successful translation of immunomodulatory therapies from experimental rodent models to patients with stroke.
Collapse
Affiliation(s)
- Kyra J Becker
- Department of Neurology, University of Washington School of Medicine, Harborview Medical Center, 325 9th Ave, Box 359775, Seattle, WA, 98104-2499, USA.
| |
Collapse
|
375
|
New translational perspectives for blood-based biomarkers of PTSD: From glucocorticoid to immune mediators of stress susceptibility. Exp Neurol 2016; 284:133-140. [PMID: 27481726 DOI: 10.1016/j.expneurol.2016.07.024] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 07/27/2016] [Accepted: 07/28/2016] [Indexed: 01/08/2023]
Abstract
Although biological systems have evolved to promote stress-resilience, there is variation in stress-responses. Understanding the biological basis of such individual differences has implications for understanding Posttraumatic Stress Disorder (PTSD) etiology, which is a maladaptive response to trauma occurring only in a subset of vulnerable individuals. PTSD involves failure to reinstate physiological homeostasis after traumatic events and is due to either intrinsic or trauma-related alterations in physiological systems across the body. Master homeostatic regulators that circulate and operate throughout the organism, such as stress hormones (e.g., glucocorticoids) and immune mediators (e.g., cytokines), are at the crossroads of peripheral and central susceptibility pathways and represent promising functional biomarkers of stress-response and target for novel therapeutics.
Collapse
|
376
|
Wohleb ES, Franklin T, Iwata M, Duman RS. Integrating neuroimmune systems in the neurobiology of depression. Nat Rev Neurosci 2016; 17:497-511. [PMID: 27277867 DOI: 10.1038/nrn.2016.69] [Citation(s) in RCA: 419] [Impact Index Per Article: 52.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Data from clinical and preclinical studies indicate that immune dysregulation, specifically of inflammatory processes, is associated with symptoms of major depressive disorder (MDD). In particular, increased levels of circulating pro-inflammatory cytokines and concomitant activation of brain-resident microglia can lead to depressive behavioural symptoms. Repeated exposure to psychological stress has a profound impact on peripheral immune responses and perturbs the function of brain microglia, which may contribute to neurobiological changes underlying MDD. Here, we review these findings and discuss ongoing studies examining neuroimmune mechanisms that influence neuronal activity as well as synaptic plasticity. Interventions targeting immune-related cellular and molecular pathways may benefit subsets of MDD patients with immune dysregulation.
Collapse
Affiliation(s)
- Eric S Wohleb
- Departments of Psychiatry and Neurobiology, Yale University School of Medicine, New Haven, Connecticut 06519, USA
| | - Tina Franklin
- Departments of Psychiatry and Neurobiology, Yale University School of Medicine, New Haven, Connecticut 06519, USA
| | - Masaaki Iwata
- Division of Neuropsychiatry, Department of Brain and Neurosciences, Tottori University Faculty of Medicine, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
| | - Ronald S Duman
- Departments of Psychiatry and Neurobiology, Yale University School of Medicine, New Haven, Connecticut 06519, USA
| |
Collapse
|
377
|
Murrough JW, Abdallah CG, Anticevic A, Collins KA, Geha P, Averill LA, Schwartz J, DeWilde KE, Averill C, Jia-Wei Yang G, Wong E, Tang CY, Krystal JH, Iosifescu DV, Charney DS. Reduced global functional connectivity of the medial prefrontal cortex in major depressive disorder. Hum Brain Mapp 2016; 37:3214-23. [PMID: 27144347 DOI: 10.1002/hbm.23235] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 03/28/2016] [Accepted: 04/19/2016] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Major depressive disorder is a disabling neuropsychiatric condition that is associated with disrupted functional connectivity across brain networks. The precise nature of altered connectivity, however, remains incompletely understood. The current study was designed to examine the coherence of large-scale connectivity in depression using a recently developed technique termed global brain connectivity. METHODS A total of 82 subjects, including medication-free patients with major depression (n = 57) and healthy volunteers (n = 25) underwent functional magnetic resonance imaging with resting data acquisition for functional connectivity analysis. Global brain connectivity was computed as the mean of each voxel's time series correlation with every other voxel and compared between study groups. Relationships between global connectivity and depressive symptom severity measured using the Montgomery-Åsberg Depression Rating Scale were examined by means of linear correlation. RESULTS Relative to the healthy group, patients with depression evidenced reduced global connectivity bilaterally within multiple regions of medial and lateral prefrontal cortex. The largest between-group difference was observed within the right subgenual anterior cingulate cortex, extending into ventromedial prefrontal cortex bilaterally (Hedges' g = -1.48, P < 0.000001). Within the depressed group, patients with the lowest connectivity evidenced the highest symptom severity within ventromedial prefrontal cortex (r = -0.47, P = 0.0005). CONCLUSIONS Patients with major depressive evidenced abnormal large-scale functional coherence in the brain that was centered within the subgenual cingulate cortex, and medial prefrontal cortex more broadly. These data extend prior studies of connectivity in depression and demonstrate that functional disconnection of the medial prefrontal cortex is a key pathological feature of the disorder. Hum Brain Mapp 37:3214-3223, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- James W Murrough
- Mood and Anxiety Disorders Program, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York.,Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Chadi G Abdallah
- Clinical Neuroscience Division, VA National Center for PTSD, West Haven, Connecticut.,Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Alan Anticevic
- Clinical Neuroscience Division, VA National Center for PTSD, West Haven, Connecticut.,Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Katherine A Collins
- Mood and Anxiety Disorders Program, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Paul Geha
- Clinical Neuroscience Division, VA National Center for PTSD, West Haven, Connecticut.,Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Lynnette A Averill
- Clinical Neuroscience Division, VA National Center for PTSD, West Haven, Connecticut.,Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Jaclyn Schwartz
- Mood and Anxiety Disorders Program, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Kaitlin E DeWilde
- Mood and Anxiety Disorders Program, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Christopher Averill
- Clinical Neuroscience Division, VA National Center for PTSD, West Haven, Connecticut.,Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | | | - Edmund Wong
- Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Cheuk Y Tang
- Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - John H Krystal
- Clinical Neuroscience Division, VA National Center for PTSD, West Haven, Connecticut.,Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| | - Dan V Iosifescu
- Mood and Anxiety Disorders Program, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York.,Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Dennis S Charney
- Mood and Anxiety Disorders Program, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York.,Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
378
|
Hodes GE, Ménard C, Russo SJ. Integrating Interleukin-6 into depression diagnosis and treatment. Neurobiol Stress 2016; 4:15-22. [PMID: 27981186 PMCID: PMC5146277 DOI: 10.1016/j.ynstr.2016.03.003] [Citation(s) in RCA: 173] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 03/24/2016] [Accepted: 03/25/2016] [Indexed: 12/13/2022] Open
Abstract
There is growing evidence of a relationship between inflammation and psychiatric illness. In particular, the cytokine Interleukin-6 (IL-6) has been linked to stress-related disorders such as depression and anxiety. Here we discuss evidence from preclinical and clinical studies examining the role of IL-6 in mood disorders. We focus on the functional role of peripheral and central release of IL-6 on the development of stress susceptibility and depression-associated behavior. By examining the contribution of both peripheral and central IL-6 to manifestations of stress-related symptomatology, we hope to broaden the way the field thinks about diagnosing and treating mood disorders.
Collapse
Affiliation(s)
- Georgia E Hodes
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Caroline Ménard
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Scott J Russo
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
379
|
Rea K, Dinan TG, Cryan JF. The microbiome: A key regulator of stress and neuroinflammation. Neurobiol Stress 2016; 4:23-33. [PMID: 27981187 PMCID: PMC5146205 DOI: 10.1016/j.ynstr.2016.03.001] [Citation(s) in RCA: 303] [Impact Index Per Article: 37.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 03/03/2016] [Accepted: 03/03/2016] [Indexed: 02/06/2023] Open
Abstract
There is a growing emphasis on the relationship between the complexity and diversity of the microorganisms that inhabit our gut (human gastrointestinal microbiota) and health/disease, including brain health and disorders of the central nervous system. The microbiota-gut-brain axis is a dynamic matrix of tissues and organs including the brain, glands, gut, immune cells and gastrointestinal microbiota that communicate in a complex multidirectional manner to maintain homeostasis. Changes in this environment can lead to a broad spectrum of physiological and behavioural effects including hypothalamic-pituitary-adrenal (HPA) axis activation, and altered activity of neurotransmitter systems and immune function. While an appropriate, co-ordinated physiological response, such as an immune or stress response are necessary for survival, a dysfunctional response can be detrimental to the host contributing to the development of a number of CNS disorders. In this review, the involvement of the gastrointestinal microbiota in stress-mediated and immune-mediated modulation of neuroendocrine, immune and neurotransmitter systems and the consequential behaviour is considered. We also focus on the mechanisms by which commensal gut microbiota can regulate neuroinflammation and further aim to exploit our understanding of their role in stress-related disorders as a consequence of neuroinflammatory processes.
Collapse
Affiliation(s)
- Kieran Rea
- APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Timothy G Dinan
- APC Microbiome Institute, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Institute, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| |
Collapse
|
380
|
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: 0.4] [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]
|
381
|
Bhattacharya A, Drevets WC. Role of Neuro-Immunological Factors in the Pathophysiology of Mood Disorders: Implications for Novel Therapeutics for Treatment Resistant Depression. Curr Top Behav Neurosci 2016; 31:339-356. [PMID: 27677784 DOI: 10.1007/7854_2016_43] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Mood disorders are associated with persistently high rates of morbidity and mortality, despite the widespread availability of antidepressant treatments. One limitation to extant therapeutic options has been that nearly all approved antidepressant pharmacotherapies exert a similar primary action of blocking monoamine transporters, and few options exist for transitioning treatment resistant patients to alternatives with distinct mechanisms. An emerging area of science that promises novel pathways to antidepressant and mood-stabilizing therapies has followed from evidence that immunological factors play major roles in the pathophysiology of at least some mood disorder subtypes. Here we review evidence that the compounds that reduce the release or signaling of neuroactive cytokines, particularly IL-1β, IL-6, and TNF-α, can exert antidepressant effects in subgroups of depressed patients who are identified by blood-based biomarkers associated with inflammation. Within this context we discuss the role of microglia in central neuroinflammation, and the interaction between the peripheral immune system and the central synaptic microenvironment during and after neuroinflammation. Finally we review data using preclinical neuroinflammation models that produce depression-like behaviors in experimental animals to guide the discovery of novel neuro-immune drug targets.
Collapse
Affiliation(s)
- Anindya Bhattacharya
- Neuroscience Drug Discovery, Janssen Research & Development, LLC, Pharmaceutical Companies of Johnson & Johnson, 3210 Merryfield Row, San Diego, CA, 92121, USA.
| | - Wayne C Drevets
- Neuroscience, Janssen Research & Development, LLC, Titusville, NJ, 08560, USA
| |
Collapse
|
382
|
Sharma A. Systems Genomics Support for Immune and Inflammation Hypothesis of Depression. Curr Neuropharmacol 2016; 14:749-58. [PMID: 26733279 PMCID: PMC5050401 DOI: 10.2174/1570159x14666160106155331] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Revised: 11/02/2015] [Accepted: 11/09/2015] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Immune system plays an important role in brain development and function. With the discovery of increased circulating inflammatory cytokine levels in depression over two decades ago, evidence implicating immune system alterations in the disease has increasingly accumulated. OBJECTIVE To assess the underlying etiology and pathophysiology, a brief overview of the hypothesis free genomic, transcriptomic and proteomic studies in depression is presented here in order to specifically examine if the immune and inflammation hypothesis of depression is supported. RESULTS It is observed that genes identified in genome-wide association studies, and genes showing differential expression in transcriptomic studies in human depression do separately overrepresent processes related to both development as well as functioning of the immune system, and inflammatory response. These processes are also enriched in differentially expressed genes reported in animal models of antidepressant treatment. It is further noted that some of the genes identified in genome sequencing and proteomic analyses in human depression, and transcriptomic studies in chronic social defeat stress, an established animal model of depression, relate to immune and inflammatory pathways. CONCLUSION In conclusion, integrative genomics evidence supports the immune and inflammation hypothesis of depression.
Collapse
Affiliation(s)
- Abhay Sharma
- CSIR-Institute of Genomics and Integrative Biology, Council of Scientific and Industrial Research, Sukhdev Vihar, Mathura Road, New Delhi 110025, India
| |
Collapse
|
383
|
Wu S, Gao Q, Zhao P, Gao Y, Xi Y, Wang X, Liang Y, Shi H, Ma Y. Sulforaphane produces antidepressant- and anxiolytic-like effects in adult mice. Behav Brain Res 2015; 301:55-62. [PMID: 26721468 DOI: 10.1016/j.bbr.2015.12.030] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 12/16/2015] [Accepted: 12/17/2015] [Indexed: 02/06/2023]
Abstract
Increasing evidence suggests that depression is accompanied by dysregulation of neuroimmune system. Sulforaphane (SFN) is a natural compound with antioxidative, anti-inflammatory and neuroprotective activities. The present study aims to investigate the effects of SFN on depressive- and anxiety-like behaviors as well as potential neuroimmune mechanisms in mice. Repeated SFN administration (10mg/kg, i.p.) significantly decreased the immobility time in the forced swimming test (FST), tail suspension test (TST), and latency time to feeding in the novelty suppressed feeding test (NSF), and increased the time in the central zone in the open field test (OPT). Using the chronic mild stress (CMS) paradigm, we confirmed that repeated SFN (10mg/kg, i.p.) administration significantly increased sucrose preference in the sucrose preference test (SPT), and immobility time in the FST and TST of mice subjected to CMS. Also, SFN treatment significantly reversed anxiety-like behaviors (assessed by the OPT and NSF) of chronically stressed mice. Finally, ELISA analysis showed that SFN administration blocked the increase in the serum levels of corticosterone (CORT), adrenocorticotropic hormone (ACTH), interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) in chronically stressed mice. In summary, these findings demonstrated that SFN has antidepressant- and anxiolytic-like activities in stressed mice model of depression, which likely occurs by inhibiting the hypothalamic-pituitary-adrenal (HPA) axis and inflammatory response to stress. These data support further exploration for developing SFN as a novel agent to treat depression and anxiety disorders.
Collapse
Affiliation(s)
- Shuhui Wu
- Department of Pathogenic Biology and Immunology, School of Basic Medicine, Hebei College of Traditional Chinese Medicine, Shijiazhuang 050000, China
| | - Qiang Gao
- Department of Nutrition, Hebei Medical University, Shijiazhuang 050017, China
| | - Pei Zhao
- Clinical laboratory, Hebei General Hospital, Shijiazhuang 050000, China
| | - Yuan Gao
- Department of Biochemistry and Molecular Biology, Hebei Medical University, Shijiazhuang 050017, China
| | - Yanjie Xi
- Undergraduate of College of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Xiaoting Wang
- Undergraduate of College of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Ying Liang
- Undergraduate of College of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Haishui Shi
- Department of Biochemistry and Molecular Biology, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Medical Biotechnology, Hebei Medical University, Shijiazhuang 050017, China.
| | - Yuxia Ma
- Department of Nutrition, Hebei Medical University, Shijiazhuang 050017, China.
| |
Collapse
|
384
|
Sapolsky RM. Stress and the brain: individual variability and the inverted-U. Nat Neurosci 2015; 18:1344-6. [DOI: 10.1038/nn.4109] [Citation(s) in RCA: 224] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|