151
|
Salvetat N, Chimienti F, Cayzac C, Dubuc B, Checa-Robles F, Dupre P, Mereuze S, Patel V, Genty C, Lang JP, Pujol JF, Courtet P, Weissmann D. Phosphodiesterase 8A to discriminate in blood samples depressed patients and suicide attempters from healthy controls based on A-to-I RNA editing modifications. Transl Psychiatry 2021; 11:255. [PMID: 33931591 PMCID: PMC8087806 DOI: 10.1038/s41398-021-01377-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 03/19/2021] [Accepted: 04/06/2021] [Indexed: 12/12/2022] Open
Abstract
Mental health issues, including major depressive disorder, which can lead to suicidal behavior, are considered by the World Health Organization as a major threat to global health. Alterations in neurotransmitter signaling, e.g., serotonin and glutamate, or inflammatory response have been linked to both MDD and suicide. Phosphodiesterase 8A (PDE8A) gene expression is significantly decreased in the temporal cortex of major depressive disorder (MDD) patients. PDE8A specifically hydrolyzes adenosine 3',5'-cyclic monophosphate (cAMP), which is a key second messenger involved in inflammation, cognition, and chronic antidepressant treatment. Moreover, alterations of RNA editing in PDE8A mRNA has been described in the brain of depressed suicide decedents. Here, we investigated PDE8A A-to-I RNA editing-related modifications in whole blood of depressed patients and suicide attempters compared to age-matched and sex-matched healthy controls. We report significant alterations of RNA editing of PDE8A in the blood of depressed patients and suicide attempters with major depression, for which the suicide attempt took place during the last month before sample collection. The reported RNA editing modifications in whole blood were similar to the changes observed in the brain of suicide decedents. Furthermore, analysis and combinations of different edited isoforms allowed us to discriminate between suicide attempters and control groups. Altogether, our results identify PDE8A as an immune response-related marker whose RNA editing modifications translate from brain to blood, suggesting that monitoring RNA editing in PDE8A in blood samples could help to evaluate depressive state and suicide risk.
Collapse
Affiliation(s)
- Nicolas Salvetat
- grid.4444.00000 0001 2112 9282ALCEDIAG/Sys2Diag, CNRS UMR 9005, Parc Euromédecine, Cap Delta, 1682 Rue de la Valsière, Montpellier, 34184 France
| | - Fabrice Chimienti
- grid.4444.00000 0001 2112 9282ALCEDIAG/Sys2Diag, CNRS UMR 9005, Parc Euromédecine, Cap Delta, 1682 Rue de la Valsière, Montpellier, 34184 France
| | - Christopher Cayzac
- grid.4444.00000 0001 2112 9282ALCEDIAG/Sys2Diag, CNRS UMR 9005, Parc Euromédecine, Cap Delta, 1682 Rue de la Valsière, Montpellier, 34184 France
| | - Benjamin Dubuc
- grid.4444.00000 0001 2112 9282ALCEDIAG/Sys2Diag, CNRS UMR 9005, Parc Euromédecine, Cap Delta, 1682 Rue de la Valsière, Montpellier, 34184 France
| | - Francisco Checa-Robles
- grid.4444.00000 0001 2112 9282ALCEDIAG/Sys2Diag, CNRS UMR 9005, Parc Euromédecine, Cap Delta, 1682 Rue de la Valsière, Montpellier, 34184 France
| | - Pierrick Dupre
- grid.4444.00000 0001 2112 9282ALCEDIAG/Sys2Diag, CNRS UMR 9005, Parc Euromédecine, Cap Delta, 1682 Rue de la Valsière, Montpellier, 34184 France
| | - Sandie Mereuze
- grid.4444.00000 0001 2112 9282ALCEDIAG/Sys2Diag, CNRS UMR 9005, Parc Euromédecine, Cap Delta, 1682 Rue de la Valsière, Montpellier, 34184 France
| | - Vipul Patel
- grid.4444.00000 0001 2112 9282ALCEDIAG/Sys2Diag, CNRS UMR 9005, Parc Euromédecine, Cap Delta, 1682 Rue de la Valsière, Montpellier, 34184 France
| | - Catherine Genty
- Department of Emergency Psychiatry and Acute Care, University Hospital/INSERM U1061, 191 Av. du Doyen Gaston Giraud, Montpellier, 34295 France
| | - Jean-Philippe Lang
- grid.4444.00000 0001 2112 9282ALCEDIAG/Sys2Diag, CNRS UMR 9005, Parc Euromédecine, Cap Delta, 1682 Rue de la Valsière, Montpellier, 34184 France
| | - Jean-François Pujol
- grid.4444.00000 0001 2112 9282ALCEDIAG/Sys2Diag, CNRS UMR 9005, Parc Euromédecine, Cap Delta, 1682 Rue de la Valsière, Montpellier, 34184 France
| | - Philippe Courtet
- Department of Emergency Psychiatry and Acute Care, University Hospital/INSERM U1061, 191 Av. du Doyen Gaston Giraud, Montpellier, 34295 France
| | - Dinah Weissmann
- ALCEDIAG/Sys2Diag, CNRS UMR 9005, Parc Euromédecine, Cap Delta, 1682 Rue de la Valsière, Montpellier, 34184, France.
| |
Collapse
|
152
|
Ivanets NN, Svistunov AA, Chubarev VN, Kinkulkina MA, Tikhonova YG, Syzrantsev NS, Sologova SS, Ignatyeva NV, Mutig K, Tarasov VV. Can Molecular Biology Propose Reliable Biomarkers for Diagnosing Major Depression? Curr Pharm Des 2021; 27:305-318. [PMID: 33234092 DOI: 10.2174/1381612826666201124110437] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 08/16/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Modern medicine has provided considerable knowledge of the pathophysiology of mental disorders at the body, systemic, organ and neurochemical levels of the biological organization of the body. Modern clinical diagnostics of depression have some problems, that is why psychiatric society makes use of diagnostics and taxonomy of different types of depression by implemention of modern molecular biomarkers in diagnostic procedures. But up to now, there are no reliable biomarkers of major depressive disorder (MDD) and other types of depression. OBJECTIVE The purpose of this review is to find fundamentals in pathological mechanisms of depression, which could be a basis for development of molecular and genetic biomarkers, being the most feasible for clinical use. METHOD This review summarizes the published data using PubMed, Science Direct, Google Scholar and Scopus. RESULTS In this review, we summarized and discussed findings in molecular biology, genetics, neuroplasticity, neurotransmitters, and neuroimaging that could increase our understanding of the biological foundations of depression and show new directions for the development of reliable biomarkers. We did not find any molecular and genetic biomarker approved for the clinic. But the Genome-Wide Association Study method promises some progress in the development of biomarkers based on SNP in the future. Epigenetic factors also are a promising target for biomarkers. We have found some differences in the etiology of different types of atypical and melancholic depression. This knowledge could be the basis for development of biomarkers for clinical practice in diagnosis, prognosis and selection of treatment. CONCLUSION Depression is not a monoetiological disease. Many pathological mechanisms are involved in depression, thus up to now, there is no approved and reliable biomarker for diagnosis, prognosis and correction of treatment of depression. The structural and functional complexity of the brain, the lack of invasive technology, poor correlations between genetic and clinical manifestation of depression, imperfect psychiatric classification and taxonomy of subtypes of disease are the main causes of this situation. One of the possible ways to come over this situation can be to pay attention to the trigger mechanism of disease and its subtypes. Researchers and clinicians should focus their efforts on searching the trigger mechanism of depression and different types of it . HPA axis can be a candidate for such trigger in depression caused by stress, because it influences the main branches of disease: neuroinflammation, activity of biogenic amines, oxidative and nitrosative stress, epigenetic factors, metabolomics, etc. But before we shall find any trigger mechanism, we need to create complex biomarkers reflecting genetic, epigenetic, metabolomics and other pathological changes in different types of depression. Recently the most encouraging results have been obtained from genetics and neuroimaging. Continuing research in these areas should be forced by using computational, statistical and systems biology approaches, which can allow to obtain more knowledge about the neurobiology of depression. In order to obtain clinically useful tests, search for biomarkers should use appropriate research methodologies with increasing samples and identifying more homogeneous groups of depressed patients.
Collapse
Affiliation(s)
- Nikolay N Ivanets
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119991, Russian Federation
| | - Andrey A Svistunov
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119991, Russian Federation
| | - Vladimir N Chubarev
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119991, Russian Federation
| | - Marina A Kinkulkina
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119991, Russian Federation
| | - Yuliya G Tikhonova
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119991, Russian Federation
| | - Nikita S Syzrantsev
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119991, Russian Federation
| | - Susanna S Sologova
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119991, Russian Federation
| | - Nelly V Ignatyeva
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119991, Russian Federation
| | - Kerim Mutig
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119991, Russian Federation
| | - Vadim V Tarasov
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119991, Russian Federation
| |
Collapse
|
153
|
Namba MD, Leyrer-Jackson JM, Nagy EK, Olive MF, Neisewander JL. Neuroimmune Mechanisms as Novel Treatment Targets for Substance Use Disorders and Associated Comorbidities. Front Neurosci 2021; 15:650785. [PMID: 33935636 PMCID: PMC8082184 DOI: 10.3389/fnins.2021.650785] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/24/2021] [Indexed: 12/12/2022] Open
Abstract
Recent studies examining the neurobiology of substance abuse have revealed a significant role of neuroimmune signaling as a mechanism through which drugs of abuse induce aberrant changes in synaptic plasticity and contribute to substance abuse-related behaviors. Immune signaling within the brain and the periphery critically regulates homeostasis of the nervous system. Perturbations in immune signaling can induce neuroinflammation or immunosuppression, which dysregulate nervous system function including neural processes associated with substance use disorders (SUDs). In this review, we discuss the literature that demonstrates a role of neuroimmune signaling in regulating learning, memory, and synaptic plasticity, emphasizing specific cytokine signaling within the central nervous system. We then highlight recent preclinical studies, within the last 5 years when possible, that have identified immune mechanisms within the brain and the periphery associated with addiction-related behaviors. Findings thus far underscore the need for future investigations into the clinical potential of immunopharmacology as a novel approach toward treating SUDs. Considering the high prevalence rate of comorbidities among those with SUDs, we also discuss neuroimmune mechanisms of common comorbidities associated with SUDs and highlight potentially novel treatment targets for these comorbid conditions. We argue that immunopharmacology represents a novel frontier in the development of new pharmacotherapies that promote long-term abstinence from drug use and minimize the detrimental impact of SUD comorbidities on patient health and treatment outcomes.
Collapse
Affiliation(s)
- Mark D. Namba
- School of Life Sciences, Arizona State University, Tempe, AZ, United States
| | | | - Erin K. Nagy
- Department of Psychology, Arizona State University, Tempe, AZ, United States
| | - M. Foster Olive
- Department of Psychology, Arizona State University, Tempe, AZ, United States
| | | |
Collapse
|
154
|
Rosado AF, Rosa PB, Platt N, Pierone BC, Neis VB, Severo Rodrigues AL, Kaster MP, Kaufmann FN. Glibenclamide treatment prevents depressive-like behavior and memory impairment induced by chronic unpredictable stress in female mice. Behav Pharmacol 2021; 32:170-181. [PMID: 33079735 DOI: 10.1097/fbp.0000000000000599] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Glibenclamide is a second-generation sulfonylurea used in the treatment of Type 2 Diabetes Mellitus. The primary target of glibenclamide is ATP-sensitive potassium channels inhibition; however, other possible targets include the control of inflammation and blood-brain barrier permeability, which makes this compound potentially interesting for the management of brain-related disorders. Here, we showed that systemic treatment with glibenclamide (5 mg/kg, p.o., for 21 days) could prevent the behavioral despair and the cognitive dysfunction induced by chronic unpredictable stress (CUS) in mice. In nonhypoglycemic doses, glibenclamide attenuated the stress-induced weight loss, decreased adrenal weight, and prevented the increase in glucocorticoid receptors in the prefrontal cortex, suggesting an impact in hypothalamic-pituitary-adrenal (HPA) axis function. Additionally, we did not observe changes in Iba-1, NLRP3 and caspase-1 levels in the prefrontal cortex or hippocampus after CUS or glibenclamide treatment. Thus, this study suggests that chronic treatment with glibenclamide prevents the emotional and cognitive effects of chronic stress in female mice. On the other hand, the control of neuroinflammation and NLRP3 inflammasome pathway is not the major mechanism mediating these effects. The behavioral effects might be mediated, in part, by the normalization of glucocorticoid receptors and HPA axis.
Collapse
Affiliation(s)
- Axel Fogaça Rosado
- Department of Biochemistry, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Priscila Batista Rosa
- Department of Biochemistry, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Nicolle Platt
- Department of Biochemistry, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Bruna Caroline Pierone
- Department of Biochemistry, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Vivian Binder Neis
- Department of Biochemistry, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | | | - Manuella Pinto Kaster
- Department of Biochemistry, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Fernanda Neutzling Kaufmann
- Department of Biochemistry, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
- Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Center, Université Laval, Quebec City, Canada
| |
Collapse
|
155
|
Cannabinoid receptor 1 signalling modulates stress susceptibility and microglial responses to chronic social defeat stress. Transl Psychiatry 2021; 11:164. [PMID: 33723234 PMCID: PMC7961142 DOI: 10.1038/s41398-021-01283-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 01/28/2021] [Accepted: 02/18/2021] [Indexed: 01/31/2023] Open
Abstract
Psychosocial stress is one of the main environmental factors contributing to the development of psychiatric disorders. In humans and rodents, chronic stress is associated with elevated inflammatory responses, indicated by increased numbers of circulating myeloid cells and activation of microglia, the brain-resident immune cells. The endocannabinoid system (ECS) regulates neuronal and endocrine stress responses via the cannabinoid receptor 1 (CB1). CB1-deficient mice (Cnr1-/-) are highly sensitive to stress, but if this involves altered inflammatory responses is not known. To test this, we exposed Cnr1+/+ and Cnr1-/- mice to chronic social defeat stress (CSDS). Cnr1-/- mice were extremely sensitive to a standard protocol of CSDS, indicated by an increased mortality rate. Therefore, a mild CSDS protocol was established, which still induced a behavioural phenotype in susceptible Cnr1-/- mice. These mice also showed altered glucocorticoid levels after mild CSDS, suggesting dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis. Mild CSDS induced weak myelopoiesis in the periphery, but no recruitment of myeloid cells to the brain. In contrast, mild CSDS altered microglial activation marker expression and morphology in Cnr1-/- mice. These microglial changes correlated with the severity of the behavioural phenotype. Furthermore, microglia of Cnr1-/- mice showed increased expression of Fkbp5, an important regulator of glucocorticoid signalling. Overall, the results confirm that CB1 signalling protects the organism from the physical and emotional harm of social stress and implicate endocannabinoid-mediated modulation of microglia in the development of stress-related pathologies.
Collapse
|
156
|
Demin KA, Smagin DA, Kovalenko IL, Strekalova T, Galstyan DS, Kolesnikova TO, De Abreu MS, Galyamina AG, Bashirzade A, Kalueff AV. CNS genomic profiling in the mouse chronic social stress model implicates a novel category of candidate genes integrating affective pathogenesis. Prog Neuropsychopharmacol Biol Psychiatry 2021; 105:110086. [PMID: 32889031 DOI: 10.1016/j.pnpbp.2020.110086] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 08/17/2020] [Accepted: 08/26/2020] [Indexed: 01/23/2023]
Abstract
Despite high prevalence, medical impact and societal burden, anxiety, depression and other affective disorders remain poorly understood and treated. Clinical complexity and polygenic nature complicate their analyses, often revealing genetic overlap and cross-disorder heritability. However, the interplay or overlaps between disordered phenotypes can also be based on shared molecular pathways and 'crosstalk' mechanisms, which themselves may be genetically determined. We have earlier predicted (Kalueff et al., 2014) a new class of 'interlinking' brain genes that do not affect the disordered phenotypes per se, but can instead specifically determine their interrelatedness. To test this hypothesis experimentally, here we applied a well-established rodent chronic social defeat stress model, known to progress in C57BL/6J mice from the Anxiety-like stage on Day 10 to Depression-like stage on Day 20. The present study analyzed mouse whole-genome expression in the prefrontal cortex and hippocampus during the Day 10, the Transitional (Day 15) and Day 20 stages in this model. Our main question here was whether a putative the Transitional stage (Day 15) would reveal distinct characteristic genomic responses from Days 10 and 20 of the model, thus reflecting unique molecular events underlining the transformation or switch from anxiety to depression pathogenesis. Overall, while in the Day 10 (Anxiety) group both brain regions showed major genomic alterations in various neurotransmitter signaling pathways, the Day 15 (Transitional) group revealed uniquely downregulated astrocyte-related genes, and the Day 20 (Depression) group demonstrated multiple downregulated genes of cell adhesion, inflammation and ion transport pathways. Together, these results reveal a complex temporal dynamics of mouse affective phenotypes as they develop. Our genomic profiling findings provide first experimental support to the idea that novel brain genes (activated here only during the Transitional stage) may uniquely integrate anxiety and depression pathogenesis and, hence, determine the progression from one pathological state to another. This concept can potentially be extended to other brain conditions as well. This preclinical study also further implicates cilial and astrocytal mechanisms in the pathogenesis of affective disorders.
Collapse
Affiliation(s)
- Konstantin A Demin
- Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Dmitry A Smagin
- Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia
| | | | - Tatyana Strekalova
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands; Research Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - David S Galstyan
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Granov Russian Scientific Center of Radiology and Surgical Technologies, Ministry of Healthcare, St. Petersburg, Russia
| | - Tatyana O Kolesnikova
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Laboratory of Cell and Molecular Biology and Neurobiology, School of Biological and Medical Physics, Moscow Institute of Physics and Technology, Moscow, Russia
| | | | | | - Alim Bashirzade
- Scientific Research Institute of Physiology and Basic Medicine, Novosibirsk, Russia; Institute of Medicine and Psychology, Novosibirsk State University, Novosibirsk, Russia
| | - Allan V Kalueff
- School of Pharmacy, Southwest University, Chongqing, China; Ural Federal University, Ekaterinburg, Russia; Laboratory of Cell and Molecular Biology and Neurobiology, School of Biological and Medical Physics, Moscow Institute of Physics and Technology, Moscow, Russia.
| |
Collapse
|
157
|
Treatment-Resistant Depression Revisited: A Glimmer of Hope. J Pers Med 2021; 11:jpm11020155. [PMID: 33672126 PMCID: PMC7927134 DOI: 10.3390/jpm11020155] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/18/2021] [Accepted: 02/18/2021] [Indexed: 12/20/2022] Open
Abstract
Major Depressive Disorder (MDD) is a highly prevalent psychiatric disorder worldwide. It causes individual suffering, loss of productivity, increased health care costs and high suicide risk. Current pharmacologic interventions fail to produce at least partial response to approximately one third of these patients, and remission is obtained in approximately 30% of patients. This is known as Treatment-Resistant Depression (TRD). The burden of TRD exponentially increases the longer it persists, with a higher risk of impaired functional and social functioning, vast losses in quality of life and significant risk of somatic morbidity and suicidality. Different approaches have been suggested and utilized, but the results have not been encouraging. In this review article, we present new approaches to identify and correct potential causes of TRD, thereby reducing its prevalence and with it the overall burden of this disease entity. We will address potential contributory factors to TRD, most of which can be investigated in many laboratories as routine tests. We discuss endocrinological aberrations, notably, hypothalamic-pituitary-adrenal (HPA) axis dysregulation and thyroid and gonadal dysfunction. We address the role of Vitamin D in contributing to depression. Pharmacogenomic testing is being increasingly used to determine Single Nucleotide Polymorphisms in Cytochrome P450, Serotonin Transporter, COMT, folic acid conversion (MTHFR). As the role of immune system dysregulation is being recognized as potentially a major contributory factor to TRD, the measurement of C-reactive protein (CRP) and select immune biomarkers, where testing is available, can guide combination treatments with anti-inflammatory agents (e.g., selective COX-2 inhibitors) reversing treatment resistance. We focus on established and emerging test procedures, potential biomarkers and non-biologic assessments and interventions to apply personalized medicine to effectively manage treatment resistance in general and TRD specifically.
Collapse
|
158
|
Nishiguchi T, Iwata M, Kajitani N, Miura A, Matsuo R, Murakami S, Nakada Y, Pu S, Shimizu Y, Tsubakino T, Yamanashi T, Shinozaki G, Tsubota J, Shirayama Y, Watanabe K, Kaneko K. Stress increases blood beta-hydroxybutyrate levels and prefrontal cortex NLRP3 activity jointly in a rodent model. Neuropsychopharmacol Rep 2021; 41:159-167. [PMID: 33609086 PMCID: PMC8340844 DOI: 10.1002/npr2.12164] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 01/25/2021] [Accepted: 02/01/2021] [Indexed: 12/28/2022] Open
Abstract
Aim This study aimed to assess the response of endogenous beta‐hydroxybutyrate to psychological stress, and its association with nucleotide‐binding domain, leucine‐rich repeat, pyrin domain‐containing 3 (NLRP3) inflammasome, and stress‐induced behavior. Methods Male C57BL/6J mice were subjected to 1‐hour restraint stress to examine changes in the endogenous beta‐hydroxybutyrate and active NLRP3 levels in the prefrontal cortex. Subsequently, we created a depression model applying 10‐day social defeat stress to the male C57BL/6J mice. Results One‐hour restraint stress rapidly increased beta‐hydroxybutyrate levels in the blood. The active NLRP3 levels in the prefrontal cortex also increased significantly. A correlation was found between the increased beta‐hydroxybutyrate levels in the blood and the active NLRP3 levels in the prefrontal cortex. The mice exposed to social defeat stress exhibited depression‐ and anxiety‐like behavioral changes in the open field, social interaction, and forced swim tests. There was a correlation between these behavioral changes and endogenous beta‐hydroxybutyrate levels. Among the social defeat model mice, those with high beta‐hydroxybutyrate levels tended to have more depression‐ and anxiety‐like behavior. Conclusions The increased blood beta‐hydroxybutyrate levels due to psychological stress correlate with the active NLRP3 levels in the prefrontal cortex, suggesting that the increased beta‐hydroxybutyrate levels due to stress may reflect a reaction to brain inflammation. In addition, mice with higher blood beta‐hydroxybutyrate levels tend to exhibit increased depression‐ and anxiety‐like behaviors; thus, an increase in blood beta‐hydroxybutyrate levels due to stress may indicate stress vulnerability. Psychological stress increased blood beta‐hydroxybutyrate (BHB) levels. Mice with higher blood BHB levels tend to exhibit increased depression‐ and anxiety‐like behaviors. The increased blood BHB levels correlate with the active NLRP3 levels in the prefrontal cortex, suggesting that increased blood BHB levels may represent stress vulnerability.![]()
Collapse
Affiliation(s)
- Tsuyoshi Nishiguchi
- Faculty of Medicine, Department of Neuropsychiatry, Tottori University, Yonago, Japan
| | - Masaaki Iwata
- Faculty of Medicine, Department of Neuropsychiatry, Tottori University, Yonago, Japan
| | - Naofumi Kajitani
- Faculty of Medicine, Department of Neuropsychiatry, Tottori University, Yonago, Japan
| | - Akihiko Miura
- Faculty of Medicine, Department of Neuropsychiatry, Tottori University, Yonago, Japan
| | - Ryoichi Matsuo
- Faculty of Medicine, Department of Neuropsychiatry, Tottori University, Yonago, Japan
| | - Shumei Murakami
- Faculty of Medicine, Department of Neuropsychiatry, Tottori University, Yonago, Japan
| | - Yumeto Nakada
- Division of Clinical Laboratory, Tottori University Hospital, Tottori, Japan
| | - Shenghong Pu
- Faculty of Medicine, Department of Neuropsychiatry, Tottori University, Yonago, Japan
| | - Yuki Shimizu
- Faculty of Medicine, Department of Neuropsychiatry, Tottori University, Yonago, Japan
| | - Tatsuya Tsubakino
- Faculty of Medicine, Department of Neuropsychiatry, Tottori University, Yonago, Japan
| | - Takehiko Yamanashi
- Faculty of Medicine, Department of Neuropsychiatry, Tottori University, Yonago, Japan.,Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Gen Shinozaki
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Jun Tsubota
- Energy Technology Laboratories, Osaka Gas Co., Ltd., Osaka, Japan
| | - Yukihiko Shirayama
- Department of Psychiatry, Teikyo University Chiba Medical Center, Ichihara, Japan
| | | | - Koichi Kaneko
- Faculty of Medicine, Department of Neuropsychiatry, Tottori University, Yonago, Japan
| |
Collapse
|
159
|
Gipson CD, Rawls S, Scofield MD, Siemsen BM, Bondy EO, Maher EE. Interactions of neuroimmune signaling and glutamate plasticity in addiction. J Neuroinflammation 2021; 18:56. [PMID: 33612110 PMCID: PMC7897396 DOI: 10.1186/s12974-021-02072-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 01/05/2021] [Indexed: 02/28/2023] Open
Abstract
Chronic use of drugs of abuse affects neuroimmune signaling; however, there are still many open questions regarding the interactions between neuroimmune mechanisms and substance use disorders (SUDs). Further, chronic use of drugs of abuse can induce glutamatergic changes in the brain, but the relationship between the glutamate system and neuroimmune signaling in addiction is not well understood. Therefore, the purpose of this review is to bring into focus the role of neuroimmune signaling and its interactions with the glutamate system following chronic drug use, and how this may guide pharmacotherapeutic treatment strategies for SUDs. In this review, we first describe neuroimmune mechanisms that may be linked to aberrant glutamate signaling in addiction. We focus specifically on the nuclear factor-kappa B (NF-κB) pathway, a potentially important neuroimmune mechanism that may be a key player in driving drug-seeking behavior. We highlight the importance of astroglial-microglial crosstalk, and how this interacts with known glutamatergic dysregulations in addiction. Then, we describe the importance of studying non-neuronal cells with unprecedented precision because understanding structure-function relationships in these cells is critical in understanding their role in addiction neurobiology. Here we propose a working model of neuroimmune-glutamate interactions that underlie drug use motivation, which we argue may aid strategies for small molecule drug development to treat substance use disorders. Together, the synthesis of this review shows that interactions between glutamate and neuroimmune signaling may play an important and understudied role in addiction processes and may be critical in developing more efficacious pharmacotherapies to treat SUDs.
Collapse
Affiliation(s)
- Cassandra D Gipson
- Department of Family and Community Medicine, University of Kentucky, 741 S. Limestone, BBSRB, Room 363, Lexington, KY, 40536-0509, USA.
| | - Scott Rawls
- Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, USA
| | - Michael D Scofield
- Department of Anesthesiology, Medical University of South Carolina, Charleston, USA
- Department of Neuroscience, Medical University of South Carolina, Charleston, USA
| | - Benjamin M Siemsen
- Department of Anesthesiology, Medical University of South Carolina, Charleston, USA
| | - Emma O Bondy
- Department of Family and Community Medicine, University of Kentucky, 741 S. Limestone, BBSRB, Room 363, Lexington, KY, 40536-0509, USA
| | - Erin E Maher
- Department of Family and Community Medicine, University of Kentucky, 741 S. Limestone, BBSRB, Room 363, Lexington, KY, 40536-0509, USA
| |
Collapse
|
160
|
He JH, Liu RP, Peng YM, Guo Q, Zhu LB, Lian YZ, Hu BL, Fan HH, Zhang X, Zhu JH. Differential and paradoxical roles of new-generation antidepressants in primary astrocytic inflammation. J Neuroinflammation 2021; 18:47. [PMID: 33602262 PMCID: PMC7890881 DOI: 10.1186/s12974-021-02097-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 02/02/2021] [Indexed: 11/18/2022] Open
Abstract
Background Selective serotonin reuptake inhibitors (SSRIs) and serotonin-norepinephrine reuptake inhibitors (SNRIs) are commonly used new-generation drugs for depression. Depressive symptoms are thought to be closely related to neuroinflammation. In this study, we used up-to-date protocols of culture and stimulation and aimed to understand how astrocytes respond to the antidepressants. Methods Primary astrocytes were isolated and cultured using neurobasal-based serum-free medium. The cells were treated with a cytokine mixture comprising complement component 1q, tumor necrosis factor α, and interleukin 1α with or without pretreatments of antidepressants. Cell viability, phenotypes, inflammatory responses, and the underlying mechanisms were analyzed. Results All the SSRIs, including paroxetine, fluoxetine, sertraline, citalopram, and fluvoxamine, show a visible cytotoxicity within the range of applied doses, and a paradoxical effect on astrocytic inflammatory responses as manifested by the promotion of inducible nitric oxide synthase (iNOS) and/or nitric oxide (NO) and the inhibition of interleukin 6 (IL-6) and/or interleukin 1β (IL-1β). The SNRI venlafaxine was the least toxic to astrocytes and inhibited the production of IL-6 and IL-1β but with no impact on iNOS and NO. All the drugs had no regulation on the polarization of astrocytic A1 and A2 types. Mechanisms associated with the antidepressants in astrocytic inflammation route via inhibition of JNK1 activation and STAT3 basal activity. Conclusions The study demonstrated that the antidepressants possess differential cytotoxicity to astrocytes and function differently, also paradoxically for the SSRIs, to astrocytic inflammation. Our results provide novel pieces into understanding the differential efficacy and tolerability of the antidepressants in treating patients in the context of astrocytes. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02097-z.
Collapse
Affiliation(s)
- Jia-Hui He
- Department of Geriatrics & Neurology, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China.,Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.,Department of Neurology, The Second Affiliated Hospital, Zhejiang University Medical College, Hangzhou, 310009, Zhejiang, China
| | - Rong-Pei Liu
- Department of Geriatrics & Neurology, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Yi-Man Peng
- Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Qing Guo
- Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Lan-Bing Zhu
- Department of Geriatrics & Neurology, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Yi-Zhi Lian
- Department of Geriatrics & Neurology, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Bei-Lei Hu
- Department of Geriatrics & Neurology, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Hui-Hui Fan
- Department of Geriatrics & Neurology, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China.,Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Xiong Zhang
- Department of Geriatrics & Neurology, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China.
| | - Jian-Hong Zhu
- Department of Geriatrics & Neurology, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China. .,Department of Preventive Medicine, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| |
Collapse
|
161
|
Gaspar R, Soares-Cunha C, Domingues AV, Coimbra B, Baptista FI, Pinto L, Ambrósio AF, Rodrigues AJ, Gomes CA. Resilience to stress and sex-specific remodeling of microglia and neuronal morphology in a rat model of anxiety and anhedonia. Neurobiol Stress 2021; 14:100302. [PMID: 33614864 PMCID: PMC7879043 DOI: 10.1016/j.ynstr.2021.100302] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 01/24/2021] [Accepted: 01/26/2021] [Indexed: 02/08/2023] Open
Abstract
Prenatal exposure to stress or glucocorticoids (GC) is associated with the appearance of psychiatric diseases later in life. Microglia, the immune cells of the brain, are altered in stress-related disorders. Synthetic GC such as dexamethasone (DEX) are commonly prescribed in case of preterm risk labour in order to promote fetal lung maturation. Recently, we reported long-lasting differences in microglia morphology in a model of in utero exposure to DEX (iuDEX), that presents an anxious phenotype. However, it is still unclear if stress differentially affects iuDEX males and females. In this work, we evaluated how iuDEX animals of both sexes cope with chronic mild stress for 2 weeks. We evaluated emotional behavior and microglia and neuronal morphology in the dorsal hippocampus (dHIP) and nucleus accumbens (NAc), two brain regions involved in emotion-related disorders. We report that males and females prenatally exposed to DEX have better performance in anxiety- and depression-related behavioral tests after chronic stress exposure in adulthood than non-exposed animals. Interestingly, iuDEX animals present sex-dependent changes in microglia morphology in the dHIP (hypertrophy in females) and in the NAc (atrophy in females and hypertrophy in males). After chronic stress, these cells undergo sex-specific morphological remodeling. Paralleled to these alterations in cytoarchitecture of microglia, we report inter-regional differences in dendritic morphology in a sex-specific manner. iuDEX females present fewer complex neurons in the NAc, whereas iuDEX males presented less complex neuronal morphology in the dHIP. Interestingly, these alterations were modified by stress exposure. Our work shows that stressful events during pregnancy can exert a preserved sex-specific effect in adulthood. Although the role of the observed cellular remodeling is still unknown, sex-specific differences in microglia plasticity induced by long-term stress exposure may anticipate differences in drug efficacy in the context of stress-induced anxiety- or depression-related behaviors. iuDEX induces anxiety- and depression-related behavioral in both sexes. iuDEX induces sex dependent fine structural alterations in neurons and microglia morphology in the dHIP and in the NAc. uCMS in combination to iuDEX normalize the behavior as well the morphology of neurons in the NAc. Stressful events during pregnancy can exert a preserved sex-specific effect in adulthood.
Collapse
Affiliation(s)
- Rita Gaspar
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal.,University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal.,Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Carina Soares-Cunha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, Braga, Portugal.,ICVS/3B's -PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Ana Verónica Domingues
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, Braga, Portugal.,ICVS/3B's -PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Bárbara Coimbra
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, Braga, Portugal.,ICVS/3B's -PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Filipa I Baptista
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal.,University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal.,Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Luísa Pinto
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, Braga, Portugal.,ICVS/3B's -PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - António F Ambrósio
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal.,University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal.,Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Ana João Rodrigues
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, Braga, Portugal.,ICVS/3B's -PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Catarina A Gomes
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal.,University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal.,Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal.,University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal
| |
Collapse
|
162
|
Zefferino R, Di Gioia S, Conese M. Molecular links between endocrine, nervous and immune system during chronic stress. Brain Behav 2021; 11:e01960. [PMID: 33295155 PMCID: PMC7882157 DOI: 10.1002/brb3.1960] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 08/17/2020] [Accepted: 10/30/2020] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION The stress response is different in various individuals, however, the mechanisms that could explain these distinct effects are not well known and the molecular correlates have been considered one at the time. Particular harmful conditions occur if the subject, instead to cope the stressful events, succumb to them, in this case, a cascade reaction happens that through different signaling causes a specific reaction named "sickness behaviour." The aim of this article is to review the complex relations among important molecules belonging to Central nervous system (CNS), immune system (IS), and endocrine system (ES) during the chronic stress response. METHODS After having verified the state of art concerning the function of cortisol, norepinephrine (NE), interleukin (IL)-1β and melatonin, we describe as they work together. RESULTS We propose a speculative hypothesis concerning the complex interplay of these signaling molecules during chronic stress, highlighting the role of IL-1β as main biomarker of this effects, indeed, during chronic stress its increment transforms this inflammatory signal into a nervous signal (NE), in turn, this uses the ES (melatonin and cortisol) to counterbalance again IL-1β. During cortisol resistance, a vicious loop occurs that increments all mediators, unbalancing IS, ES, and CNS networks. This IL-1β increase would occur above all when the individual succumbs to stressful events, showing the Sickness Behaviour Symptoms. IL-1β might, through melatonin and vice versa, determine sleep disorders too. CONCLUSION The molecular links here outlined could explain how stress plays a role in etiopathogenesis of several diseases through this complex interplay.
Collapse
Affiliation(s)
- Roberto Zefferino
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Sante Di Gioia
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Massimo Conese
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| |
Collapse
|
163
|
Tran H, Sagi V, Jarrett S, Palzer EF, Badgaiyan RD, Gupta K. Diet and companionship modulate pain via a serotonergic mechanism. Sci Rep 2021; 11:2330. [PMID: 33526805 PMCID: PMC7851147 DOI: 10.1038/s41598-021-81654-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 01/08/2021] [Indexed: 01/30/2023] Open
Abstract
Treatment of severe chronic and acute pain in sickle cell disease (SCD) remains challenging due to the interdependence of pain and psychosocial modulation. We examined whether modulation of the descending pain pathway through an enriched diet and companionship could alleviate pain in transgenic sickle mice. Mechanical and thermal hyperalgesia were reduced significantly with enriched diet and/or companionship. Upon withdrawal of both conditions, analgesic effects observed prior to withdrawal were diminished. Serotonin (5-hydroxytryptamine, 5-HT) was found to be increased in the spinal cords of mice provided both treatments. Additionally, 5-HT production improved at the rostral ventromedial medulla and 5-HT accumulated at the dorsal horn of the spinal cord of sickle mice, suggesting the involvement of the descending pain pathway in the analgesic response. Modulation of 5-HT and its effect on hyperalgesia was also investigated through pharmaceutical approaches. Duloxetine, a serotonin-norepinephrine reuptake inhibitor, showed a similar anti-nociceptive effect as the combination of diet and companionship. Depletion of 5-HT through p-chlorophenylalanine attenuated the anti-hyperalgesic effect of enriched diet and companionship. More significantly, improved diet and companionship enhanced the efficacy of a sub-optimal dose of morphine for analgesia in sickle mice. These findings offer the potential to reduce opioid use without pharmacological interventions to develop effective pain management strategies.
Collapse
Affiliation(s)
- Huy Tran
- grid.17635.360000000419368657 Division of Hematology, Oncology and Transplantation, Department of Medicine, Vascular Biology Center, University of Minnesota, Minneapolis, MN USA
| | - Varun Sagi
- grid.17635.360000000419368657 Division of Hematology, Oncology and Transplantation, Department of Medicine, Vascular Biology Center, University of Minnesota, Minneapolis, MN USA
| | - Sarita Jarrett
- grid.16753.360000 0001 2299 3507Northwestern University, Evanston, IL USA
| | - Elise F. Palzer
- grid.17635.360000000419368657Biostatistical Design and Analysis Center, Clinical and Translational Sciences Institute, University of Minnesota, Minneapolis, MN USA
| | - Rajendra D. Badgaiyan
- grid.267309.90000 0001 0629 5880Department of Psychiatry, Long School of Medicine, University of Texas Health Science Center, San Antonio, Texas USA
| | - Kalpna Gupta
- grid.17635.360000000419368657 Division of Hematology, Oncology and Transplantation, Department of Medicine, Vascular Biology Center, University of Minnesota, Minneapolis, MN USA ,Hematology/Oncology, Department of Medicine, University of California, Irvine and Southern California Institute for Research and Education, VA Medical Center, 5901 East 7th St, Long Beach, CA 90822 USA
| |
Collapse
|
164
|
Li J, Tian S, Wang H, Wang Y, Du C, Fang J, Wang X, Wang Y, Gong Z, Yan B, Wang M. Protection of hUC-MSCs against neuronal complement C3a receptor-mediated NLRP3 activation in CUMS-induced mice. Neurosci Lett 2021; 741:135485. [PMID: 33161108 DOI: 10.1016/j.neulet.2020.135485] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 09/21/2020] [Accepted: 11/01/2020] [Indexed: 01/28/2023]
Abstract
BACKGROUND Hyperactivation of complement C3 and inflammation-related activation of NLR family pyrin domain containing 3 (NLRP3) inflammasome are implicated in the etiology of stress-related disorders. Studies have shown that human umbilical cord mesenchymal stromal cells (hUC-MSCs) have immunomodulatory and anti-inflammatory effects; however, the mechanism remains unclear. METHODS hUC-MSCs were administered to chronic unpredictable mild stress (CUMS) model mice once a week for four weeks. After the administration of hUC-MSCs, several parameters were assessed, including behavioral performance, synapse-related proteins, complement C3 receptors (C3aR) in neurons, and the NLRP3 inflammasome. Then, CUMS mice were injected with SB290157, a complement C3aR antagonist, and the behavioral index and NLRP3 inflammasome activation were tested. RESULTS The open-field and forced swimming behavioral tests showed an improvement in depression-like behaviors in the CUMS-exposed mice after the administration of hUC-MSCs. Treatment with hUC-MSCs significantly decreased the neuronal C3aR levels and alleviated synaptic damage. Furthermore, the levels of the NLRP3 inflammasome and inflammatory factors were reduced after hUC-MSC administration. In particular, treatment with a C3aR antagonist also decreased NLRP3 inflammasome expression and inflammation, which was consistent with the observed improvements after hUC-MSC treatment. CONCLUSION hUC-MSCs can attenuate NLRP3 activation in CUMS-induced mice, which may be correlated with C3aR in neurons.
Collapse
Affiliation(s)
- Jing Li
- Department of Neurology, the First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Brain Aging and Cognitive Neuroscience Key Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Shujuan Tian
- Department of Neurology, the First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Brain Aging and Cognitive Neuroscience Key Laboratory of Hebei Province, Shijiazhuang, Hebei, China.
| | - Hualong Wang
- Department of Neurology, the First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Brain Aging and Cognitive Neuroscience Key Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Yanyong Wang
- Department of Neurology, the First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Brain Aging and Cognitive Neuroscience Key Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Chongbo Du
- Department of Neurology, the First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Brain Aging and Cognitive Neuroscience Key Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Jiyu Fang
- Department of Neurology, the First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Brain Aging and Cognitive Neuroscience Key Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Xiaoxiao Wang
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yufeng Wang
- Basic Medical College, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zhexuan Gong
- Basic Medical College, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Baoyong Yan
- Cell Therapy Laboratory, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
| | - Mingwei Wang
- Department of Neurology, the First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; Brain Aging and Cognitive Neuroscience Key Laboratory of Hebei Province, Shijiazhuang, Hebei, China.
| |
Collapse
|
165
|
Grolli RE, Mingoti MED, Bertollo AG, Luzardo AR, Quevedo J, Réus GZ, Ignácio ZM. Impact of COVID-19 in the Mental Health in Elderly: Psychological and Biological Updates. Mol Neurobiol 2021; 58:1905-1916. [PMID: 33404981 PMCID: PMC7786865 DOI: 10.1007/s12035-020-02249-x] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 12/04/2020] [Indexed: 12/13/2022]
Abstract
Since December 2019, the world has been experiencing the challenge of facing coronavirus disease-19 (COVID-19), a severe infectious disease caused by the new coronavirus, SARS-CoV-2. The individuals with the most severe symptoms and the highest risk of death are the elderly and those with chronic illness. Among chronic conditions, those with a certain degree of chronic inflammation may predispose to a more severe evolution of COVID-19. Elderly with psychiatric disorders can present a persistent inflammatory state, a characteristic of the age’s immunological senescence, but the disorder can accentuate that. Social isolation is still the safest way to avoid contamination. However, isolated older people may have or worsen mental health conditions due to isolation and health concerns. In this scenario, a SARS-CoV-2 infection may progress to more severe disease. Conversely, COVID-19 can predispose or aggravate psychiatric disorders, as it induces a cytokine storm, causing systemic hyper inflammation. It may damage the blood-brain barrier, resulting in inflammation in the central nervous system. Besides, SARS-CoV-2 is likely to reach and trigger an inflammatory process directly in the nervous system. This review makes an update about research on the mental health of the elderly during the pandemic. Also, it discusses the vulnerability of these individuals in the face of stress and in the case of contracting COVID-19, considering mainly the stress’s hormonal and inflammatory mechanisms. Finally, the review points out possible care and attention strategies and entertainment and activities that can reduce the damage to mental and physical health and improve the elderly’s quality of life. Isolation and concerns about COVID-19 may harm elderly mental health. Immunosenescence and pandemic stress increase the risk of psychiatric disorders. Stress and disorders may potentiate the elderly’s inflammation and COVID-19 symptoms. SARS-CoV-2 hyperinflammation is a risk factor for elderly psychiatric disorders ![]()
Collapse
Affiliation(s)
- Roberta Eduarda Grolli
- Undergraduate Nursing Course, Federal University of the Southern Frontier, Chapeco, SC, Brazil.,Laboratory of Physiology, Pharmacology, Psychopathology, Postgraduate Program in Biomedical Sciences, Federal University of Southern Frontier, Chapeco, SC, Brazil
| | - Maiqueli Eduarda Dama Mingoti
- Undergraduate Nursing Course, Federal University of the Southern Frontier, Chapeco, SC, Brazil.,Laboratory of Physiology, Pharmacology, Psychopathology, Postgraduate Program in Biomedical Sciences, Federal University of Southern Frontier, Chapeco, SC, Brazil
| | - Amanda Gollo Bertollo
- Undergraduate Nursing Course, Federal University of the Southern Frontier, Chapeco, SC, Brazil.,Laboratory of Physiology, Pharmacology, Psychopathology, Postgraduate Program in Biomedical Sciences, Federal University of Southern Frontier, Chapeco, SC, Brazil
| | - Adriana Remião Luzardo
- Undergraduate Nursing Course, Federal University of the Southern Frontier, Chapeco, SC, Brazil.,Laboratory of Innovation and Health Technologies, Federal University of the Southern Frontier, Chapeco, SC, Brazil
| | - João Quevedo
- Laboratory of Translational Psychiatry, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, SC, Brazil.,Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.,Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.,Neuroscience Graduate Program, Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Gislaine Zilli Réus
- Laboratory of Translational Psychiatry, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Zuleide Maria Ignácio
- Undergraduate Nursing Course, Federal University of the Southern Frontier, Chapeco, SC, Brazil. .,Laboratory of Physiology, Pharmacology, Psychopathology, Postgraduate Program in Biomedical Sciences, Federal University of Southern Frontier, Chapeco, SC, Brazil. .,Laboratory of Translational Psychiatry, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciuma, SC, Brazil.
| |
Collapse
|
166
|
Dudek KA, Dion‐Albert L, Kaufmann FN, Tuck E, Lebel M, Menard C. Neurobiology of resilience in depression: immune and vascular insights from human and animal studies. Eur J Neurosci 2021; 53:183-221. [PMID: 31421056 PMCID: PMC7891571 DOI: 10.1111/ejn.14547] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 07/22/2019] [Accepted: 08/12/2019] [Indexed: 12/12/2022]
Abstract
Major depressive disorder (MDD) is a chronic and recurrent psychiatric condition characterized by depressed mood, social isolation and anhedonia. It will affect 20% of individuals with considerable economic impacts. Unfortunately, 30-50% of depressed individuals are resistant to current antidepressant treatments. MDD is twice as prevalent in women and associated symptoms are different. Depression's main environmental risk factor is chronic stress, and women report higher levels of stress in daily life. However, not every stressed individual becomes depressed, highlighting the need to identify biological determinants of stress vulnerability but also resilience. Based on a reverse translational approach, rodent models of depression were developed to study the mechanisms underlying susceptibility vs resilience. Indeed, a subpopulation of animals can display coping mechanisms and a set of biological alterations leading to stress resilience. The aetiology of MDD is multifactorial and involves several physiological systems. Exacerbation of endocrine and immune responses from both innate and adaptive systems are observed in depressed individuals and mice exhibiting depression-like behaviours. Increasing attention has been given to neurovascular health since higher prevalence of cardiovascular diseases is found in MDD patients and inflammatory conditions are associated with depression, treatment resistance and relapse. Here, we provide an overview of endocrine, immune and vascular changes associated with stress vulnerability vs. resilience in rodents and when available, in humans. Lack of treatment efficacy suggests that neuron-centric treatments do not address important causal biological factors and better understanding of stress-induced adaptations, including sex differences, could contribute to develop novel therapeutic strategies including personalized medicine approaches.
Collapse
Affiliation(s)
- Katarzyna A. Dudek
- Department of Psychiatry and NeuroscienceFaculty of Medicine and CERVO Brain Research CenterUniversité LavalQuebec CityQCCanada
| | - Laurence Dion‐Albert
- Department of Psychiatry and NeuroscienceFaculty of Medicine and CERVO Brain Research CenterUniversité LavalQuebec CityQCCanada
| | - Fernanda Neutzling Kaufmann
- Department of Psychiatry and NeuroscienceFaculty of Medicine and CERVO Brain Research CenterUniversité LavalQuebec CityQCCanada
| | - Ellen Tuck
- Smurfit Institute of GeneticsTrinity CollegeDublinIreland
| | - Manon Lebel
- Department of Psychiatry and NeuroscienceFaculty of Medicine and CERVO Brain Research CenterUniversité LavalQuebec CityQCCanada
| | - Caroline Menard
- Department of Psychiatry and NeuroscienceFaculty of Medicine and CERVO Brain Research CenterUniversité LavalQuebec CityQCCanada
| |
Collapse
|
167
|
Turkheimer FE, Althubaity N, Schubert J, Nettis MA, Cousins O, Dima D, Mondelli V, Bullmore ET, Pariante C, Veronese M. Increased serum peripheral C-reactive protein is associated with reduced brain barriers permeability of TSPO radioligands in healthy volunteers and depressed patients: implications for inflammation and depression. Brain Behav Immun 2021; 91:487-497. [PMID: 33160089 DOI: 10.1016/j.bbi.2020.10.025] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 10/30/2020] [Accepted: 10/31/2020] [Indexed: 01/08/2023] Open
Abstract
The relationship between peripheral and central immunity and how these ultimately may cause depressed behaviour has been the focus of a number of imaging studies conducted with Positron Emission Tomography (PET). These studies aimed at testing the immune-mediated model of depression that proposes a direct effect of peripheral cytokines and immune cells on the brain to elicit a neuroinflammatory response via a leaky blood-brain barrier and ultimately depressive behaviour. However, studies conducted so far using PET radioligands targeting the neuroinflammatory marker 18 kDa translocator protein (TSPO) in patient cohorts with depression have demonstrated mild inflammatory brain status but no correlation between central and peripheral immunity. To gain a better insight into the relationship between heightened peripheral immunity and neuroinflammation, we estimated blood-to-brain and blood-to-CSF perfusion rates for two TSPO radiotracers collected in two separate studies, one large cross-sectional study of neuroinflammation in normal and depressed cohorts (N = 51 patients and N = 25 controls) and a second study where peripheral inflammation in N = 7 healthy controls was induced via subcutaneous injection of interferon (IFN)-α. In both studies we observed a consistent negative association between peripheral inflammation, measured with c-reactive protein P (CRP), and radiotracer perfusion into and from the brain parenchyma and CSF. Importantly, there was no association of this effect with the marker of BBB leakage S100β, that was unchanged. These results suggest a different model of peripheral-to-central immunity interaction whereas peripheral inflammation may cause a reduction in BBB permeability. This effect, on the long term, is likely to disrupt brain homeostasis and induce depressive behavioural symptoms.
Collapse
Affiliation(s)
- Federico E Turkheimer
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| | - Noha Althubaity
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Julia Schubert
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Maria A Nettis
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Oliver Cousins
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Danai Dima
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Department of Psychology, School of Arts and Social Sciences, City, University of London, London, UK
| | - Valeria Mondelli
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Edward T Bullmore
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, UK; Cambridgeshire and Peterborough NHS Foundation Trust, Cambridge, UK
| | - Carmine Pariante
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Mattia Veronese
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
168
|
Benedetti F, Poletti S, Vai B, Mazza MG, Lorenzi C, Brioschi S, Aggio V, Branchi I, Colombo C, Furlan R, Zanardi R. Higher baseline interleukin-1β and TNF-α hamper antidepressant response in major depressive disorder. Eur Neuropsychopharmacol 2021; 42:35-44. [PMID: 33191075 DOI: 10.1016/j.euroneuro.2020.11.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 10/18/2020] [Accepted: 11/06/2020] [Indexed: 01/06/2023]
Abstract
Raised pro-inflammatory immune/inflammatory setpoints, leading to an increased production of peripheral cytokines, have been associated with Major Depressive Disorder (MDD) and with failure to respond to first-line antidepressant drugs. However, the usefulness of these biomarkers in clinical psychopharmacology has been questioned because single findings did not translate into the clinical practice, where patients are prescribed treatments upon clinical need. We studied a panel of 27 inflammatory biomarkers in a sample of 108 inpatients with MDD, treated with antidepressant monotherapy for 4 weeks upon clinical need in a specialized hospital setting, and assessed the predictive effect of baseline peripheral measures of inflammation on antidepressing efficacy (response rates and time-lagged pattern of decrease of depression severity) using a machine-learning approach with elastic net penalized regression, and multivariate analyses in the context of the general linear model. When considering both categorical and continuous measures of response, baseline levels of IL-1β predicted non-response to antidepressants, with the predicted probability to respond being highly dispersed at low levels of IL-1β, and stratifying toward non-response when IL-1β is high. Significant negative effects were also detected for TNF-α, while IL-12 weakly predicted response. These findings support the usefulness of inflammatory biomarkers in the clinical psychopharmacology of depression, and add to ongoing research efforts aiming at defining reliable cutoff values to identify depressed patients in clinical settings with high inflammation, and low probability to respond.
Collapse
Affiliation(s)
- Francesco Benedetti
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute Ospedale San Raffaele, Milano, Italy; Vita-Salute San Raffaele University, Milano, Italy.
| | - Sara Poletti
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute Ospedale San Raffaele, Milano, Italy; Vita-Salute San Raffaele University, Milano, Italy
| | - Benedetta Vai
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute Ospedale San Raffaele, Milano, Italy; Vita-Salute San Raffaele University, Milano, Italy; Fondazione Centro San Raffaele, Milano, Italy
| | - Mario Gennaro Mazza
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute Ospedale San Raffaele, Milano, Italy; Vita-Salute San Raffaele University, Milano, Italy
| | - Cristina Lorenzi
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Silvia Brioschi
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Veronica Aggio
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute Ospedale San Raffaele, Milano, Italy; Vita-Salute San Raffaele University, Milano, Italy
| | - Igor Branchi
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| | - Cristina Colombo
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute Ospedale San Raffaele, Milano, Italy; Vita-Salute San Raffaele University, Milano, Italy
| | - Roberto Furlan
- Vita-Salute San Raffaele University, Milano, Italy; Clinical Neuroimmunology, Division of Neuroscience, IRCCS Scientific Institute Ospedale San Raffaele, Milano, Italy
| | - Raffaella Zanardi
- Psychiatry and Clinical Psychobiology, Division of Neuroscience, IRCCS Scientific Institute Ospedale San Raffaele, Milano, Italy; Vita-Salute San Raffaele University, Milano, Italy
| |
Collapse
|
169
|
Demin KA, Taranov AS, Ilyin NP, Lakstygal AM, Volgin AD, de Abreu MS, Strekalova T, Kalueff AV. Understanding neurobehavioral effects of acute and chronic stress in zebrafish. Stress 2021; 24:1-18. [PMID: 32036720 DOI: 10.1080/10253890.2020.1724948] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Stress is a common cause of neuropsychiatric disorders, evoking multiple behavioral, endocrine and neuro-immune deficits. Animal models have been extensively used to understand the mechanisms of stress-related disorders and to develop novel strategies for their treatment. Complementing rodent and clinical studies, the zebrafish (Danio rerio) is one of the most important model organisms in biomedicine. Rapidly becoming a popular model species in stress neuroscience research, zebrafish are highly sensitive to both acute and chronic stress, and show robust, well-defined behavioral and physiological stress responses. Here, we critically evaluate the utility of zebrafish-based models for studying acute and chronic stress-related CNS pathogenesis, assess the advantages and limitations of these aquatic models, and emphasize their relevance for the development of novel anti-stress therapies. Overall, the zebrafish emerges as a powerful and sensitive model organism for stress research. Although these fish generally display evolutionarily conserved behavioral and physiological responses to stress, zebrafish-specific aspects of neurogenesis, neuroprotection and neuro-immune responses may be particularly interesting to explore further, as they may offer additional insights into stress pathogenesis that complement (rather than merely replicate) rodent findings. Compared to mammals, zebrafish models are also characterized by increased availability of gene-editing tools and higher throughput of drug screening, thus being able to uniquely empower translational research of genetic determinants of stress and resilience, as well as to foster innovative CNS drug discovery and the development of novel anti-stress therapies.
Collapse
Affiliation(s)
- Konstantin A Demin
- Institute of Experimental Biomedicine, Almazov National Medical Research Center, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
- Laboratory of Biological Psychiatry, Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Alexander S Taranov
- Laboratory of Biological Psychiatry, Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
- Laboratory of Preclinical Bioscreening, Granov Russian Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, Pesochny, Russia
| | - Nikita P Ilyin
- Laboratory of Biological Psychiatry, Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
- Laboratory of Preclinical Bioscreening, Granov Russian Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, Pesochny, Russia
| | - Anton M Lakstygal
- Laboratory of Biological Psychiatry, Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
- Laboratory of Preclinical Bioscreening, Granov Russian Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, Pesochny, Russia
| | - Andrey D Volgin
- Laboratory of Preclinical Bioscreening, Granov Russian Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, Pesochny, Russia
| | - Murilo S de Abreu
- Bioscience Institute, University of Passo Fundo, Passo Fundo, Brazil
- The International Zebrafish Neuroscience Research Consortium (ZNRC), Slidell, LA, USA
| | - Tatyana Strekalova
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia
- Maastricht University, Maastricht, The Netherlands
- Research Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Allan V Kalueff
- School of Pharmacy, Southwest University, Chongqing, China
- Ural Federal University, Ekaterinburg, Russia
| |
Collapse
|
170
|
Cruz SA, Qin Z, Ricke KM, Stewart AFR, Chen HH. Neuronal protein-tyrosine phosphatase 1B hinders sensory-motor functional recovery and causes affective disorders in two different focal ischemic stroke models. Neural Regen Res 2021; 16:129-136. [PMID: 32788467 PMCID: PMC7818877 DOI: 10.4103/1673-5374.286970] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Ischemic brain injury causes neuronal death and inflammation. Inflammation activates protein-tyrosine phosphatase 1B (PTP1B). Here, we tested the significance of PTP1B activation in glutamatergic projection neurons on functional recovery in two models of stroke: by photothrombosis, focal ischemic lesions were induced in the sensorimotor cortex (SM stroke) or in the peri-prefrontal cortex (peri-PFC stroke). Elevated PTP1B expression was detected at 4 days and up to 6 weeks after stroke. While ablation of PTP1B in neurons of neuronal knockout (NKO) mice had no effect on the volume or resorption of ischemic lesions, markedly different effects on functional recovery were observed. SM stroke caused severe sensory and motor deficits (adhesive removal test) in wild type and NKO mice at 4 days, but NKO mice showed drastically improved sensory and motor functional recovery at 8 days. In addition, peri-PFC stroke caused anxiety-like behaviors (elevated plus maze and open field tests), and depression-like behaviors (forced swimming and tail suspension tests) in wild type mice 9 and 28 days after stroke, respectively, with minimal effect on sensory and motor function. Peri-PFC stroke-induced affective disorders were associated with fewer active (FosB+) neurons in the PFC and nucleus accumbens but more FosB+ neurons in the basolateral amygdala, compared to sham-operated mice. In contrast, mice with neuronal ablation of PTP1B were protected from anxiety-like and depression-like behaviors and showed no change in FosB+ neurons after peri-PFC stroke. Taken together, our study identifies neuronal PTP1B as a key component that hinders sensory and motor functional recovery and also contributes to the development of anxiety-like and depression-like behaviors after stroke. Thus, PTP1B may represent a novel therapeutic target to improve stroke recovery. All procedures for animal use were approved by the Animal Care and Use Committee of the University of Ottawa Animal Care and Veterinary Service (protocol 1806) on July 27, 2018.
Collapse
Affiliation(s)
- Shelly A Cruz
- Ottawa Hospital Research Institute, Neuroscience Program; Brain and Mind Institute, University of Ottawa, Ottawa, ON, Canada
| | - Zhaohong Qin
- Ottawa Hospital Research Institute, Neuroscience Program; Brain and Mind Institute, University of Ottawa, Ottawa, ON, Canada
| | - Konrad M Ricke
- Brain and Mind Institute; Department of Biochemistry, Microbiology and Immunology, University of Ottawa; University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Alexandre F R Stewart
- Department of Biochemistry, Microbiology and Immunology; Centre for Infection, Immunity and Inflammation, University of Ottawa; University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Hsiao-Huei Chen
- Ottawa Hospital Research Institute, Neuroscience Program; Brain and Mind Institute; Cellular and Molecular Medicine; Department of Medicine; Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
171
|
Bliss-Moreau E, Rudebeck PH. Animal models of human mood. Neurosci Biobehav Rev 2021; 120:574-582. [PMID: 33007355 PMCID: PMC10474843 DOI: 10.1016/j.neubiorev.2020.06.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/16/2020] [Accepted: 06/17/2020] [Indexed: 11/28/2022]
Abstract
Humans' everyday experience of the world is influenced by our moods. Moods are consciously accessible affective states that extend over time that are characterized by their valence and arousal. They also likely have a long evolutionary heritage and serve as an important adaptive affective mechanism. When they become maladaptive or overly biased, pathological affective states such as depression can emerge. Despite the importance of moods for human experience, little is known about their causal neurobiological mechanisms. In humans, limitations related to methods and interpretations of the data prevent causal investigations into the origins of mood, highlighting the importance of animal models. Nonhuman primates that share key neuroanatomical, affective, and social features with humans will be essential to uncovering their foundation. Identifying and validating mood-like states in animals is, however, challenging not least because mood is a human construct requiring verbal communication. Here we outline a theoretical framework for animal models of human mood, drawing upon established psychological literature where it exists before reviewing the extant studies of non-human primate models of mood-like states.
Collapse
Affiliation(s)
- Eliza Bliss-Moreau
- Department of Psychology, California National Primate Research Center, University of California, Davis, CA, 95616, USA.
| | - Peter H Rudebeck
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA.
| |
Collapse
|
172
|
Dudek KA, Kaufmann FN, Lavoie O, Menard C. Central and peripheral stress-induced epigenetic mechanisms of resilience. Curr Opin Psychiatry 2021; 34:1-9. [PMID: 33141775 DOI: 10.1097/yco.0000000000000664] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW Resilience is an adaptation process presented by an individual despite facing adversities. Epigenetic changes, such as histone acetylation/methylation and DNA methylation, have been demonstrated to mediate stress response. In this review, we summarize recent findings on epigenetic mechanisms contributing to stress resilience. RECENT FINDINGS Epigenetic modifications of genes involved in synaptic plasticity, endocrine, immune, and vascular systems are linked to resilience. For instance, increased DNA methylation of the nonneuronal growth factor Gdnf in specific brain regions promotes stress resilience. Additionally, high DNA methylation at the glucocorticoid receptor gene was associated with resilience in both rodents and humans. At the immune level, chronic stress induces increased DNA methylation at IL6 gene, a mediator of stress vulnerability. Moreover, epigenetic adaptations of the blood--brain barrier have been recently associated with stress resilience, which could lead to innovative therapeutic approaches to treat depression. SUMMARY Identification of both central and peripheral epigenetic changes promoting stress resilience represent promising novel targets in the development of preventive and personalized medicine. Nevertheless, more research is needed to establish sex specific differences and to identify novel epigenetic mechanisms, such as serotonylation and dopaminylation, that hold great promises for the field of psychiatry.
Collapse
Affiliation(s)
- Katarzyna Anna Dudek
- Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Center, Université Laval, Quebec City, Canada
| | | | | | | |
Collapse
|
173
|
Zhou JH, Zhang DL, Ning BL, Xue XJ, Zhao L, Wu Q, Yan LD, Liu M, Fu WB. The Role of Acupuncture in Hormonal Shock-Induced Cognitive-Related Symptoms in Perimenopausal Depression: A Randomized Clinical Controlled Trial. Front Psychiatry 2021; 12:772523. [PMID: 35095593 PMCID: PMC8793332 DOI: 10.3389/fpsyt.2021.772523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 12/20/2021] [Indexed: 12/04/2022] Open
Abstract
Introduction: Perimenopausal depression is predominantly caused by hormone shock, but the underlying physical and psychological factors are still unclear. Objectives: To explore the constituent components of perimenopausal depression by dynamically depicting its influencing factors and interactive pathways from the perspective of embodied cognition. Methods: This is a randomized clinical controlled trial. In this study, electroacupuncture was compared with escitalopram. A total of 242 participants with mild to moderate perimenopausal depression were enrolled from 6 hospitals in China. Each participant had a 12-week intervention and a 12-week follow-up period. The primary outcome of this study is the Hamilton Depression Rating Scale (HAMD-17), and the secondary outcome is the menopause-specific quality of life scale (MENQOL), serum Follicle-stimulating hormone (FSH), luteinizing hormone (LH), and estrogen (E2) levels. Results: The structural equation model suggested that hormone levels were not directly associated with HAMD-17 (P = 0.852), while MENQOL was statistically correlated with HAMD-17 as an intermediary variable (P < 0.001). Electroacupuncture gradually showed positive impacts on MENQOL and HAMD-17 during the follow-up period (P < 0.05). Cognitive impairment is the dominant dimension of perimenopausal depression. Conclusions: Hormonal shock may affect clinical symptoms and poor quality of life to induce cognitive impairment leading perimenopausal depression, and this impact on cognition is embodied. Electroacupuncture has positive effect on perimenopausal depression and quality of life.
Collapse
Affiliation(s)
- Jun-He Zhou
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, Guangzhou, China; School of Psychology Center for Studies of Psychological Application, and Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, Guangzhou, China.,The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - De-Long Zhang
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, Guangzhou, China; School of Psychology Center for Studies of Psychological Application, and Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, Guangzhou, China
| | - Bai-Le Ning
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, Guangzhou, China; School of Psychology Center for Studies of Psychological Application, and Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, Guangzhou, China
| | - Xiao-Juan Xue
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, Guangzhou, China; School of Psychology Center for Studies of Psychological Application, and Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, Guangzhou, China
| | - Lin Zhao
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qian Wu
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lu-Da Yan
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ming Liu
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, Guangzhou, China; School of Psychology Center for Studies of Psychological Application, and Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, Guangzhou, China
| | - Wen-Bin Fu
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Innovative Research Team of Acupuncture for Depression and Related Disorders, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| |
Collapse
|
174
|
Hou J, Wang C, Ma D, Chen Y, Jin H, An Y, Jia J, Huang L, Zhao H. The cardioprotective and anxiolytic effects of Chaihujialonggumuli granule on rats with anxiety after acute myocardial infarction is partly mediated by suppression of CXCR4/NF-κB/GSDMD pathway. Biomed Pharmacother 2021; 133:111015. [PMID: 33232924 DOI: 10.1016/j.biopha.2020.111015] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/04/2020] [Accepted: 11/11/2020] [Indexed: 12/17/2022] Open
Abstract
AIMS Over-expression of CXCR4 activates nuclear translocation of NF-κB, induces high expression of NLRP3, GSDMD, IL-1β and IL-18, which promotes severe inflammatory response following myocardial infarction. Previous studies revealed inflammation induces anxiety after myocardial infarction. The Chaihujialonggumuli granule has anti-inflammatory properties and could tranquillize mind. But the mechanism of its efficacy remains unknown. This study was to investigate the possible mechanism of BFG on cardioprotective and anxiolytic. METHODS The expression of CXCR4, NF-κB, NLRP3and GSDMD was measured with western-blot, QRT-PCR. The expression location of CXCR4, NLRP3, GSDMD were determined by immunohistochemistry. IL-1β、IL-18 in the peripheral blood were measured by ELISA. HE staining, Masson staining and transmission electron microscopy were used to observe morphological changes of cardiomyocytes. Echocardiography was used to assess cardiac function after cardiac surgery. Elevated cross maze test and open field test were used to evaluate behaviours. Western blot was used to detect the protein expressions of 5-HT, DA, IL-1β, IL-18 and neuron damage was investigated by Nissl staining in the hippocampus. RESULTS The up-regulation of CXCR4, NF-κB, NLRP3 and GSDMD were found in the infarcted area after left coronary artery ligation. Pathological staining and analysis showed that more severe inflammatory cytokines infiltration, myocardial fibrosis, were found in myocardial tissue of the complex group rats. And when compared to the sham group, the levels of IL-1β, IL-18 was increased of the complex group in both peripheral blood and brain. Behavioural test and echocardiography indicated that the rats in complex group exploration behaviours was significantly reduced, and with poor cardiac functional recovery. The AMD3100 had an inhibitory impact of CXCR4 on the activition of its downstream effectors, alleviating inflammatory reaction. Furthermore, the BFG decreased the expression level of CXCR4, NF-κB, GSDMD, NLRP3 in the infarcted area after myocardial infarction, when compared to the complex group. The assays in the brain indicated the BFG suppressed expression and activity of IL-1β, IL-18, and improved 5-HT and DA synthesis. CONCLUSIONS In sum, our study indicated that BFG may reduce inflammation, treat co-existing anxiety after myocardial infarction through inhibition of CXCR4/NF-κB/GSDMD signalling.
Collapse
Affiliation(s)
- Jiqiu Hou
- The Third Affiliate Hospital of Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Chao Wang
- The DongFang Hospital of Beijing University of Chinese Medicine, Beijing, 100078, China
| | - Di Ma
- The Third Affiliate Hospital of Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yali Chen
- The Third Affiliate Hospital of Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Huihui Jin
- The Third Affiliate Hospital of Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Ying An
- The Third Affiliate Hospital of Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Jingyun Jia
- The Third Affiliate Hospital of Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Lexi Huang
- The Third Affiliate Hospital of Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Haibin Zhao
- The DongFang Hospital of Beijing University of Chinese Medicine, Beijing, 100078, China.
| |
Collapse
|
175
|
Granulocyte-Colony Stimulating Factor Reduces Cocaine-Seeking and Downregulates Glutamatergic Synaptic Proteins in Medial Prefrontal Cortex. J Neurosci 2020; 41:1553-1565. [PMID: 33361463 DOI: 10.1523/jneurosci.1452-20.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 12/08/2020] [Accepted: 12/14/2020] [Indexed: 12/12/2022] Open
Abstract
Psychostimulant use disorder is a major public health issue, and despite the scope of the problem there are currently no Food and Drug Administration (FDA)-approved treatments. There would be tremendous utility in development of a treatment that could help patients both achieve and maintain abstinence. Previous work from our group has identified granulocyte-colony stimulating factor (G-CSF) as a neuroactive cytokine that alters behavioral response to cocaine, increases synaptic dopamine release, and enhances cognitive flexibility. Here, we investigate the role of G-CSF in affecting extinction and reinstatement of cocaine-seeking and perform detailed characterization of its proteomic effects in multiple limbic substructures. Male Sprague Dawley rats were injected with PBS or G-CSF during (1) extinction or (2) abstinence from cocaine self-administration, and drug seeking behavior was measured. Quantitative assessment of changes in the proteomic landscape in the nucleus accumbens (NAc) and medial prefrontal cortex (mPFC) were performed via data-independent acquisition (DIA) mass spectrometry analysis. Administration of G-CSF during extinction accelerated the rate of extinction, and administration during abstinence attenuated cue-induced cocaine-seeking. Analysis of global protein expression demonstrated that G-CSF regulated proteins primarily in mPFC that are critical to glutamate signaling and synapse maintenance. Taken together, these findings support G-CSF as a viable translational research target with the potential to reduce drug craving or seeking behaviors. Importantly, recombinant G-CSF exists as an FDA-approved medication which may facilitate rapid clinical translation. Additionally, using cutting-edge multiregion discovery proteomics analyses, these studies identify a novel mechanism underlying G-CSF effects on behavioral plasticity.SIGNIFICANCE STATEMENT Pharmacological treatments for psychostimulant use disorder are desperately needed, especially given the disease's chronic, relapsing nature. However, there are currently no Food and Drug Administration (FDA)-approved pharmacotherapies. Emerging evidence suggests that targeting the immune system may be a viable translational research strategy; preclinical studies have found that the neuroactive cytokine granulocyte-colony stimulating factor (G-CSF) alters cocaine reward and reinforcement and can enhance cognitive flexibility. Given this basis of evidence we studied the effects of G-CSF treatment on extinction and reinstatement of cocaine seeking. We find that administration of G-CSF accelerates extinction and reduces cue-induced drug seeking after cocaine self-administration. In addition, G-CSF leads to downregulation of synaptic glutamatergic proteins in medial prefrontal cortex (mPFC), suggesting that G-CSF influences drug seeking via glutamatergic mechanisms.
Collapse
|
176
|
Shi Z, Qin M, Huang L, Xu T, Chen Y, Hu Q, Peng S, Peng Z, Qu LN, Chen SG, Tuo QH, Liao DF, Wang XP, Wu RR, Yuan TF, Li YH, Liu XM. Human torpor: translating insights from nature into manned deep space expedition. Biol Rev Camb Philos Soc 2020; 96:642-672. [PMID: 33314677 DOI: 10.1111/brv.12671] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 11/09/2020] [Accepted: 11/17/2020] [Indexed: 12/12/2022]
Abstract
During a long-duration manned spaceflight mission, such as flying to Mars and beyond, all crew members will spend a long period in an independent spacecraft with closed-loop bioregenerative life-support systems. Saving resources and reducing medical risks, particularly in mental heath, are key technology gaps hampering human expedition into deep space. In the 1960s, several scientists proposed that an induced state of suppressed metabolism in humans, which mimics 'hibernation', could be an ideal solution to cope with many issues during spaceflight. In recent years, with the introduction of specific methods, it is becoming more feasible to induce an artificial hibernation-like state (synthetic torpor) in non-hibernating species. Natural torpor is a fascinating, yet enigmatic, physiological process in which metabolic rate (MR), body core temperature (Tb ) and behavioural activity are reduced to save energy during harsh seasonal conditions. It employs a complex central neural network to orchestrate a homeostatic state of hypometabolism, hypothermia and hypoactivity in response to environmental challenges. The anatomical and functional connections within the central nervous system (CNS) lie at the heart of controlling synthetic torpor. Although progress has been made, the precise mechanisms underlying the active regulation of the torpor-arousal transition, and their profound influence on neural function and behaviour, which are critical concerns for safe and reversible human torpor, remain poorly understood. In this review, we place particular emphasis on elaborating the central nervous mechanism orchestrating the torpor-arousal transition in both non-flying hibernating mammals and non-hibernating species, and aim to provide translational insights into long-duration manned spaceflight. In addition, identifying difficulties and challenges ahead will underscore important concerns in engineering synthetic torpor in humans. We believe that synthetic torpor may not be the only option for manned long-duration spaceflight, but it is the most achievable solution in the foreseeable future. Translating the available knowledge from natural torpor research will not only benefit manned spaceflight, but also many clinical settings attempting to manipulate energy metabolism and neurobehavioural functions.
Collapse
Affiliation(s)
- Zhe Shi
- National Clinical Research Center for Mental Disorders, and Department of Psychaitry, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.,Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.,State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China.,Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai, 200030, China
| | - Meng Qin
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Lu Huang
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, 510632, China
| | - Tao Xu
- Department of Anesthesiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Ying Chen
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Qin Hu
- College of Life Sciences and Bio-Engineering, Beijing University of Technology, Beijing, 100024, China
| | - Sha Peng
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Zhuang Peng
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Li-Na Qu
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Shan-Guang Chen
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Qin-Hui Tuo
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Duan-Fang Liao
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Xiao-Ping Wang
- National Clinical Research Center for Mental Disorders, and Department of Psychaitry, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Ren-Rong Wu
- National Clinical Research Center for Mental Disorders, and Department of Psychaitry, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Ti-Fei Yuan
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai, 200030, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226000, China
| | - Ying-Hui Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Xin-Min Liu
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.,State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China.,Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China
| |
Collapse
|
177
|
Effect of gut microbiota on depressive-like behaviors in mice is mediated by the endocannabinoid system. Nat Commun 2020; 11:6363. [PMID: 33311466 PMCID: PMC7732982 DOI: 10.1038/s41467-020-19931-2] [Citation(s) in RCA: 186] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 11/06/2020] [Indexed: 12/15/2022] Open
Abstract
Depression is the leading cause of disability worldwide. Recent observations have revealed an association between mood disorders and alterations of the intestinal microbiota. Here, using unpredictable chronic mild stress (UCMS) as a mouse model of depression, we show that UCMS mice display phenotypic alterations, which could be transferred from UCMS donors to naïve recipient mice by fecal microbiota transplantation. The cellular and behavioral alterations observed in recipient mice were accompanied by a decrease in the endocannabinoid (eCB) signaling due to lower peripheral levels of fatty acid precursors of eCB ligands. The adverse effects of UCMS-transferred microbiota were alleviated by selectively enhancing the central eCB or by complementation with a strain of the Lactobacilli genus. Our findings provide a mechanistic scenario for how chronic stress, diet and gut microbiota generate a pathological feed-forward loop that contributes to despair behavior via the central eCB system.
Collapse
|
178
|
Daria S, Proma MA, Shahriar M, Islam SMA, Bhuiyan MA, Islam MR. Serum interferon-gamma level is associated with drug-naïve major depressive disorder. SAGE Open Med 2020; 8:2050312120974169. [PMID: 33282305 PMCID: PMC7682211 DOI: 10.1177/2050312120974169] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 10/26/2020] [Indexed: 01/30/2023] Open
Abstract
Objectives: Major depressive disorder is a leading heterogeneous psychiatric illness manifested by persistent low mood, a feeling of sadness, and diminished interest in daily activities. Many biological, genetic, and social factors are thought to be linked with depression. But any suitable early risk assessment markers are absent for this illness. Therefore, we aimed to investigate the serum levels of IFN-γ in major depressive disorder patients to further investigate the association between serum levels of this cytokine and major depression. Methods: This prospective case-control study enrolled 120 major depressive disorder patients and 100 healthy controls matched by age, sex, and body mass index. A qualified psychiatrist diagnosed the major depressive disorder patients and evaluated healthy controls according to the Diagnostic and Statistical Manual of Mental Health Disorders (5th ed.; DSM-5). The Hamilton depression rating scale was applied for all the study participants to measure the severity of depression. Serum IFN-γ levels were measured by a commercially available enzyme-linked immunosorbent assay kit (Boster Biological Technology, Pleasanton, CA, USA). Results: This study observed that serum IFN-γ levels were significantly decreased in major depressive disorder patients compared to healthy controls. A significant negative correlation (r = −0.375; p < 0.001) was obtained between serum IFN-γ levels and Hamilton depression scores. Receiver operating characteristic analysis showed good diagnostic performance of lowered serum IFN-γ levels in depression with an area under the curve at 0.790. Conclusion: We suggest the altered serum IFN-γ levels are associated with the pathophysiology of depression. The reduced levels of serum IFN-γ might be used as an early risk assessment tool for major depression.
Collapse
Affiliation(s)
- Sohel Daria
- Department of Pharmacy, University of Asia Pacific, Dhaka, Bangladesh
| | | | - Mohammad Shahriar
- Department of Pharmacy, University of Asia Pacific, Dhaka, Bangladesh
| | | | | | - Md Rabiul Islam
- Department of Pharmacy, University of Asia Pacific, Dhaka, Bangladesh
| |
Collapse
|
179
|
Bertollo AG, Grolli RE, Plissari ME, Gasparin VA, Quevedo J, Réus GZ, Bagatini MD, Ignácio ZM. Stress and serum cortisol levels in major depressive disorder: a cross-sectional study. AIMS Neurosci 2020; 7:459-469. [PMID: 33263081 PMCID: PMC7701370 DOI: 10.3934/neuroscience.2020028] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 11/10/2020] [Indexed: 12/15/2022] Open
Abstract
Major depressive disorder (MDD) is one of the disorders that most causes disability and affects about 265 million people worldwide, according to the World Health Organization (WHO). Chronic stress is one of the most prevalent factors that trigger MDD. Among the most relevant biological mechanisms that mediate stress and MDD are changes in the hypothalamic-pituitary-adrenal (HPA) axis function. Hypercortisolism is one of the relevant mechanisms involved in response to stress and is present in many people with MDD and in animals subjected to stress in the laboratory. This study aimed to investigate the levels of stress and cortisol in individuals diagnosed with MDD from the Basic Health Unit (BHU) in a small city in the western region of Santa Catarina, Brazil. Depression scores were assessed using Beck's inventory. For the investigation of stress, an adaptation with twenty-four questions of the Checklist-90-R manual was performed. The analysis of the cortisol levels in the individuals' serum was by the chemiluminescence method. Depression and stress scores were significantly higher in individuals with MDD than in control subjects (p < 0.001). Cortisol levels were also significantly higher in individuals with MDD (p < 0.05). Besides, depression scores were positively correlated with stress scores in individuals with MDD (Pearson's "r" = 0.70). Conclusion: Individuals with MDD had higher stress levels and cortisol than control subjects. The positive correlation between the levels of stress and depression in MDD individuals suggests that these conditions are related to a dysregulation of the HPA axis function.
Collapse
Affiliation(s)
- Amanda G Bertollo
- Laboratory of Physiology Pharmacology and Psychopathology, Graduate Program in Biomedical Sciences, Federal University of the Southern Frontier, Chapecó, SC, Brazil
| | - Roberta E Grolli
- Laboratory of Physiology Pharmacology and Psychopathology, Graduate Program in Biomedical Sciences, Federal University of the Southern Frontier, Chapecó, SC, Brazil
| | - Marcos E Plissari
- Laboratory of Physiology Pharmacology and Psychopathology, Graduate Program in Biomedical Sciences, Federal University of the Southern Frontier, Chapecó, SC, Brazil
| | - Vanessa A Gasparin
- Santa Catarina State University, West Higher Education Center, Chapecó, SC, Brazil
| | - João Quevedo
- Laboratory of Translational Psychiatry, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciúma, SC, Brazil
- Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Neuroscience Graduate Program, Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Gislaine Z Réus
- Laboratory of Translational Psychiatry, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - Margarete D Bagatini
- Laboratory of Cell Culture, Graduate Program in Biomedical Sciences, Federal University of the Southern Frontier, Chapecó, SC, Brazil
| | - Zuleide M Ignácio
- Laboratory of Physiology Pharmacology and Psychopathology, Graduate Program in Biomedical Sciences, Federal University of the Southern Frontier, Chapecó, SC, Brazil
- Laboratory of Translational Psychiatry, Graduate Program in Health Sciences, University of Southern Santa Catarina, Criciúma, SC, Brazil
| |
Collapse
|
180
|
Athira KV, Bandopadhyay S, Samudrala PK, Naidu VGM, Lahkar M, Chakravarty S. An Overview of the Heterogeneity of Major Depressive Disorder: Current Knowledge and Future Prospective. Curr Neuropharmacol 2020; 18:168-187. [PMID: 31573890 PMCID: PMC7327947 DOI: 10.2174/1570159x17666191001142934] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 08/05/2019] [Accepted: 09/27/2019] [Indexed: 02/08/2023] Open
Abstract
Major depressive disorder (MDD) is estimated to impose maximum debilitating effects on the society by 2030, with its critical effects on health, functioning, quality of life and concomitant high levels of morbidity and mortality. Yet, the disease is inadequately understood, diagnosed and treated. Moreover, with the recent drastic rise in the pace of life, stress has materialized as one of the most potent environmental factors for depression. In this scenario, it is important to understand the modern pathogenetic hypotheses and mechanisms, and possibly try to shift from the traditional approaches in depression therapy. These include the elaboration of pathophysiological changes in heterogeneous systems such as genetic, epigenetic, serotonergic, noradrenergic, gamma-aminobutyric acid, glutamatergic and endocannabinoid systems, neurotrophic factors, HPA axis, immune system as well as cellular stress mechanisms. These components interact with each other in a complex matrix and further elucidation of their mechanism and cascade pathways are needed. This might aid in the identification of MDD subtypes as well as the development of sophisticated biomarkers. Further, characterization might also aid in developing multitargeted therapies that hold much promise as compared to the conventional monoamine based treatment. New candidate pharmacons, refined psychotherapeutic modalities, advanced neuro-surgical and imaging techniques as well as the implementation of pharmacokinetic, pharmacogenetic prescribing guidelines constitute the emerging expanses of MDD treatment.
Collapse
Affiliation(s)
- Kaipuzha Venu Athira
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, 781125, Assam, India.,Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Tarnaka, Uppal Road, Hyderabad 500007, India.,Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, 682 041, Kerala, India
| | - Sikta Bandopadhyay
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Tarnaka, Uppal Road, Hyderabad 500007, India
| | - Pavan Kumar Samudrala
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, 781125, Assam, India
| | - V G M Naidu
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, 781125, Assam, India
| | - Mangala Lahkar
- Department of Pharmacology, Gauhati Medical College, Guwahati, 781032, Assam, India
| | - Sumana Chakravarty
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Tarnaka, Uppal Road, Hyderabad 500007, India
| |
Collapse
|
181
|
Cao C, Liu M, Qu S, Huang R, Qi M, Zhu Z, Zheng J, Chen Z, Wang Z, Han Z, Zhu Y, Huang F, Duan JA. Chinese medicine formula Kai-Xin-San ameliorates depression-like behaviours in chronic unpredictable mild stressed mice by regulating gut microbiota-inflammation-stress system. JOURNAL OF ETHNOPHARMACOLOGY 2020; 261:113055. [PMID: 32592887 DOI: 10.1016/j.jep.2020.113055] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 05/11/2020] [Accepted: 05/30/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Kai-Xin-San (KXS) has been prescribed by TCM doctors for treating psychiatric diseases with the core symptoms of anhedonia, amnesia, and dizziness. According to the symptoms of patients, KXS series formulae are created by varying the compatible ratio of herbs. Today, these formulae are still used in the clinic to treat major depressive disorders. AIM OF THE STUDY We hoped to evaluate the antidepressant-like effect of Kai-Xin-San via regulation of the gut-brain axis. MATERIALS AND METHODS Standardized extracts of three representative compatible ratios of KXS had been prepared, and quality control of the extracts was performed by HPLC-MS/MS. Chronic unpredictable mild stress (CUMS)-induced depression-like mice were used as the depression animal model. After KXS treatment, the antidepressant-like effects of KXS were assessed by behavioural tests. The gut microbiota compositions in the faeces were determined by 16S rRNA sequencing technology. The levels of LPS, pro-inflammatory cytokines and HPA-axis-related hormones were measured by ELISA kits, and the expression of barrier proteins in the small intestines and prefrontal cortex were determined by Western blot analysis. Furthermore, antibiotics were used to determine the correlation between KXS exerting an antidepressant-like effect and regulating the gut-brain axis. RESULTS KXS alleviated depression-like behaviours in CUMS-exposed mice. Furthermore, these parameters were also found to be changed after KXS treatment. Alteration of the gut microbiota composition were found in the small intestines. A decrease in the LPS and the pro-inflammatory cytokines were found in both the small intestine and brain. An increase in the tight junction proteins was found in the gut epithelium barrier and the blood-brain barrier. A decrease in the stress-related hormones was found in the central nervous system. Furthermore, antibiotic treatment attenuated the antidepressant-like effect of KXS in CUMS-exposed mice. CONCLUSIONS KXS exerted an antidepressant-like effect regulating the gut-brain axis, which included gut micro-environment modification, suppression of neuronal inflammation in the brain and inhibition of HPA axis activation in CUMS-induced depression-like mice.
Collapse
MESH Headings
- Animals
- Antidepressive Agents/pharmacology
- Behavior, Animal/drug effects
- Brain/drug effects
- Brain/metabolism
- Chronic Disease
- Cytokines/metabolism
- Depression/drug therapy
- Depression/metabolism
- Depression/microbiology
- Depression/psychology
- Disease Models, Animal
- Drugs, Chinese Herbal/pharmacology
- Dysbiosis
- Fluoxetine/pharmacology
- Gastrointestinal Microbiome/drug effects
- Host-Pathogen Interactions
- Inflammation Mediators/metabolism
- Intestine, Small/metabolism
- Intestine, Small/microbiology
- Male
- Mice, Inbred ICR
- Stress, Psychological/drug therapy
- Stress, Psychological/metabolism
- Stress, Psychological/microbiology
- Stress, Psychological/psychology
Collapse
Affiliation(s)
- Cheng Cao
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine,Nanjing University of Chinese Medicine, Nan Jing, Jiangsu Province, China.
| | - Mengqiu Liu
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine,Nanjing University of Chinese Medicine, Nan Jing, Jiangsu Province, China.
| | - Suchen Qu
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine,Nanjing University of Chinese Medicine, Nan Jing, Jiangsu Province, China.
| | - Renjie Huang
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine,Nanjing University of Chinese Medicine, Nan Jing, Jiangsu Province, China.
| | - Mingzhu Qi
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine,Nanjing University of Chinese Medicine, Nan Jing, Jiangsu Province, China.
| | - Ziqiang Zhu
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine,Nanjing University of Chinese Medicine, Nan Jing, Jiangsu Province, China.
| | - Jiani Zheng
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine,Nanjing University of Chinese Medicine, Nan Jing, Jiangsu Province, China.
| | - Zhichun Chen
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine,Nanjing University of Chinese Medicine, Nan Jing, Jiangsu Province, China.
| | - Zhikang Wang
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine,Nanjing University of Chinese Medicine, Nan Jing, Jiangsu Province, China.
| | - Zhengxiang Han
- Department of Neurology and Rehabilitation, Shanghai Seventh People's Hospital, Shanghai University of TCM, Shanghai, China.
| | - Yue Zhu
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine,Nanjing University of Chinese Medicine, Nan Jing, Jiangsu Province, China.
| | - Fei Huang
- Department of Endocrinology, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Su Zhou, Jiangsu Province, China.
| | - Jin-Ao Duan
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine,Nanjing University of Chinese Medicine, Nan Jing, Jiangsu Province, China.
| |
Collapse
|
182
|
Lucerne KE, Kiraly DD. The role of gut-immune-brain signaling in substance use disorders. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 157:311-370. [PMID: 33648673 DOI: 10.1016/bs.irn.2020.09.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Substance use disorders (SUDs) are debilitating neuropsychiatric conditions that exact enormous costs in terms of loss of life and individual suffering. While much progress has been made defining the neurocircuitry and intracellular signaling cascades that contribute to SUDs, these studies have yielded limited effective treatment options. This has prompted greater exploration of non-traditional targets in addiction. Emerging data suggest inputs from peripheral systems, such as the immune system and the gut microbiome, impact multiple neuropsychiatric diseases, including SUDs. Until recently the gut microbiome, peripheral immune system, and the CNS have been studied independently; however, current work shows the gut microbiome and immune system critically interact to modulate brain function. Additionally, the gut microbiome and immune system intimately regulate one another via extensive bidirectional communication. Accumulating evidence suggests an important role for gut-immune-brain communication in the pathogenesis of substance use disorders. Thus, a better understanding of gut-immune-brain signaling could yield important insight to addiction pathology and potential treatment options.
Collapse
Affiliation(s)
- Kelsey E Lucerne
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Drew D Kiraly
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| |
Collapse
|
183
|
Jia X, Gao Z, Hu H. Microglia in depression: current perspectives. SCIENCE CHINA-LIFE SCIENCES 2020; 64:911-925. [PMID: 33068286 DOI: 10.1007/s11427-020-1815-6] [Citation(s) in RCA: 131] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/09/2020] [Indexed: 12/11/2022]
Abstract
Major depressive disorder (MDD) is a prevalent psychiatric disease that involves malfunctions of different cell types in the brain. Accumulating studies started to reveal that microglia, the primary resident immune cells, play an important role in the development and progression of depression. Microglia respond to stress-triggered neuroinflammation, and through the release of proinflammatory cytokines and their metabolic products, microglia may modulate the function of neurons and astrocytes to regulate depression. In this review, we focused on the role of microglia in the etiology of depression. We discussed the dynamic states of microglia; the correlative and causal evidence of microglial abnormalities in depression; possible mechanisms of how microglia sense depression-related stress and modulate depression state; and how antidepressive therapies affect microglia. Understanding the role of microglia in depression may shed light on developing new treatment strategies to fight against this devastating mental illness.
Collapse
Affiliation(s)
- Xiaoning Jia
- Department of Psychiatry of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.,The MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Brain Science and Brain Medicine, Hangzhou, 310012, China
| | - Zhihua Gao
- The MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Brain Science and Brain Medicine, Hangzhou, 310012, China. .,NHC and CAMS Key Laboratory of Medical Neurobiology, Mental Health Center, Zhejiang University, Hangzhou, 310058, China. .,Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.
| | - Hailan Hu
- Department of Psychiatry of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China. .,The MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Brain Science and Brain Medicine, Hangzhou, 310012, China. .,NHC and CAMS Key Laboratory of Medical Neurobiology, Mental Health Center, Zhejiang University, Hangzhou, 310058, China. .,Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou, 510515, China. .,Fountain-Valley Institute for Life Sciences, Guangzhou, 510530, China. .,Research Units of Brain Mechanisms Underlying Emotion and Emotion disorders, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| |
Collapse
|
184
|
Alshammari TK, Alghamdi HM, Alduhailan HE, Saja MF, Alrasheed NM, Alshammari MA. Examining the central effects of chronic stressful social isolation on rats. Biomed Rep 2020; 13:56. [PMID: 33123370 PMCID: PMC7583698 DOI: 10.3892/br.2020.1363] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 09/16/2020] [Indexed: 11/23/2022] Open
Abstract
Stress-related disorders are extremely complex and current treatment strategies have limitations. The present study investigated alternative pathological mechanisms using a combination of multiple environmental approaches with biochemical and molecular tools. The aim of the present study was to evaluate blood-brain-barrier (BBB) integrity in socially manipulated animal housing conditions. Multiple environmentally-related models were employed in the current study. The main model proposed (chronically isolated rats) was biochemically validated using the level of peripheral corticosterone. The current study examined and compared the mRNA levels of certain inflammatory and BBB markers in the hippocampal tissue of chronically isolated rats, including claudin-5 (cldn5) and tight junction protein (tjp). Animals were divided into four groups: i) Standard housed rats (controls); ii) chronically isolated rats; iii) control rats treated with fluoxetine, which is a standard selective serotonin reuptake inhibitor; and iv) isolated rats treated with fluoxetine. To further examine the effect of environmental conditions on BBB markers, the current study assessed BBB markers in enriched environmental (EE) housing and short-term isolation conditions. The results demonstrated a significant increase in cldn5 and tjp levels in the chronically isolated group. Despite some anomalous results, alterations in mRNA levels were further confirmed in EE housing conditions compared with chronically isolated rats. This trend was also observed in rats subjected to short-term isolation compared with paired controls. Additionally, levels of IL-6, an inflammatory marker associated with neuroinflammation, were markedly increased in the isolated group. However, treatment with fluoxetine treatment reversed these effects. The results indicated that BBB integrity may be compromised in stress-related disorders, highlighting a need for further functional studies on the kinetics of BBB in stress-related models.
Collapse
Affiliation(s)
- Tahani K Alshammari
- Department of Pharmacology and Toxicology, College of Medicine, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia
| | - Hajar M Alghamdi
- Pharmacology and Toxicology Graduate Program, College of Pharmacy, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia
| | - Hessa E Alduhailan
- Department of Pharmacology and Toxicology, College of Medicine, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia
| | - Maha F Saja
- Department of Physiology, College of Medicine, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia
| | - Nouf M Alrasheed
- Department of Pharmacology and Toxicology, College of Medicine, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia
| | - Musaad A Alshammari
- Department of Pharmacology and Toxicology, College of Medicine, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia
| |
Collapse
|
185
|
Resilience to Stress and Resilience to Pain: Lessons from Molecular Neurobiology and Genetics. Trends Mol Med 2020; 26:924-935. [PMID: 32976800 DOI: 10.1016/j.molmed.2020.03.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/18/2020] [Accepted: 03/25/2020] [Indexed: 12/12/2022]
Abstract
What biological factors account for resilience to pain or to behavioral stress? Here, we discuss examples of cellular and molecular mechanisms within disparate parts of the nervous system as contributors to such resilience. In some especially well-studied humans, it is possible to identify particular neuronal cell types in the peripheral nervous system (PNS) and pinpoint specific genes that are major contributors to pain resilience. We also discuss more complex factors that operate within the central nervous system (CNS) to confer resilience to behavioral stress. We propose that genetic and neurobiological substrates for resilience are discoverable and suggest more generally that neurology and psychiatry hold lessons for each other as investigators search for actionable, biological underpinnings of disease.
Collapse
|
186
|
Carpita B, Marazziti D, Palego L, Giannaccini G, Betti L, Dell'Osso L. Microbiota, Immune System and Autism Spectrum Disorders: An Integrative Model towards Novel Treatment Options. Curr Med Chem 2020; 27:5119-5136. [PMID: 31448708 DOI: 10.2174/0929867326666190328151539] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 03/05/2019] [Accepted: 03/06/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Autism Spectrum Disorder (ASD) is a condition strongly associated with genetic predisposition and familial aggregation. Among ASD patients, different levels of symptoms severity are detectable, while the presence of intermediate autism phenotypes in close relatives of ASD probands is also known in literature. Recently, increasing attention has been paid to environmental factors that might play a role in modulating the relationship between genomic risk and development and severity of ASD. Within this framework, an increasing body of evidence has stressed a possible role of both gut microbiota and inflammation in the pathophysiology of neurodevelopment. The aim of this paper is to review findings about the link between microbiota dysbiosis, inflammation and ASD. METHODS Articles ranging from 1990 to 2018 were identified on PUBMED and Google Scholar databases, with keyword combinations as: microbiota, immune system, inflammation, ASD, autism, broad autism phenotype, adult. RESULTS Recent evidence suggests that microbiota alterations, immune system and neurodevelopment may be deeply intertwined, shaping each other during early life. However, results from both animal models and human samples are still heterogeneous, while few studies focused on adult patients and ASD intermediate phenotypes. CONCLUSION A better understanding of these pathways, within an integrative framework between central and peripheral systems, might not only shed more light on neural basis of ASD symptoms, clarifying brain pathophysiology, but it may also allow to develop new therapeutic strategies for these disorders, still poorly responsive to available treatments.
Collapse
Affiliation(s)
- Barbara Carpita
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, Via Roma, 6756100 Pisa, Italy
| | - Donatella Marazziti
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, Via Roma, 6756100 Pisa, Italy
| | - Lionella Palego
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, Via Roma, 6756100 Pisa, Italy
| | - Gino Giannaccini
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, Via Roma, 6756100 Pisa, Italy
| | - Laura Betti
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, Via Roma, 6756100 Pisa, Italy
| | - Liliana Dell'Osso
- Department of Clinical and Experimental Medicine, Section of Psychiatry, University of Pisa, Via Roma, 6756100 Pisa, Italy
| |
Collapse
|
187
|
A green and efficient pseudotargeted lipidomics method for the study of depression based on ultra-high performance supercritical fluid chromatography-tandem mass spectrometry. J Pharm Biomed Anal 2020; 192:113646. [PMID: 33017797 DOI: 10.1016/j.jpba.2020.113646] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 09/04/2020] [Accepted: 09/15/2020] [Indexed: 12/23/2022]
Abstract
The pseudotargeted lipidomics method integrates the advantages of untargeted andtargeted lipidomics methods as a novel emerging approach. In this study, a green andefficient pseudotargeted lipidomics method based on ultra-high performancesupercritical fluid chromatography-tandem mass spectrometry (UHPSFC-MS/MS) wasdeveloped. The tandem mass spectra of the analytes were obtained by using UHPSFCwith quadrupole-time of flight MS (Q-TOF MS) in MS E mode and the multiplereaction monitoring (MRM) transitions of the lipidome were defined. Then, thecandidate MRM transitions were verified by UHPSFC with triple quadrupole massspectrometry (QqQ MS) in the scheduled MRM mode. In total, 758 potential lipidscorresponding to 509 and 249 MRM transitions were detected within 8 min in positiveand negative modes, respectively. The established pseudotargeted lipidomics methodwas validated to have excellent analytical characteristics. Compared with thepseudotargeted method based on ultra-high performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS), the UHPSFC-MS/MS-basedpseudotargeted method not only reduced the analytical time by half but also improvedthe sensitivity and resolution for most analytes, especially had better separation forlipid isomers. Besides, the UHPSFC-MS/MS-based pseudotargeted method showedhigher sensitivity and better repeatability for most analytes than the UHPSFC-MS/MS-based untargeted method. The established method was finally applied to investigatingthe lipid profiles of the plasma from the depressed rats and 33 differential variableswere screened, which related to three metabolic pathways. The results indicated thatthe UHPSFC-MS/MS-based pseudotargeted method is reliable and efficient and couldbe used in the lipidomics studies.
Collapse
|
188
|
Deonaraine KK, Wang Q, Cheng H, Chan KL, Lin HY, Liu K, Parise LF, Cathomas F, Leclair KB, Flanigan ME, Li L, Aleyasin H, Guevara C, Hao K, Zhang B, Russo SJ, Wang J. Sex-specific peripheral and central responses to stress-induced depression and treatment in a mouse model. J Neurosci Res 2020; 98:2541-2553. [PMID: 32918293 DOI: 10.1002/jnr.24724] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 06/17/2020] [Accepted: 08/15/2020] [Indexed: 11/11/2022]
Abstract
Major depressive disorder affects ~20% of the world population and is characterized by strong sexual dimorphism with females being two to three times more likely to develop this disorder. Previously, we demonstrated that a combination therapy with dihydrocaffeic acid and malvidin-glucoside to synergistically target peripheral inflammation and stress-induced synaptic maladaptation in the brain was effective in alleviating chronic social defeat stress (CSDS)-induced depression-like phenotype in male mice. Here, we test the combination therapy in a female CSDS model for depression and compared sex-specific responses to stress in the periphery and the central nervous system. Similar to male mice, the combination treatment is also effective in promoting resilience against the CSDS-induced depression-like behavior in female mice. However, there are sex-specific differences in peripheral immune responses and differential gene regulation in the prefrontal cortex to chronic stress and to the treatment. These data indicate that while therapeutic approaches to combat stress-related disorders may be effective in both sexes, the mechanisms underlying these effects differ, emphasizing the need for inclusion of both sexes in preclinical studies using animal models.
Collapse
Affiliation(s)
- Kristina K Deonaraine
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Qian Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Icahn Institute of Genomics and Multi-Scale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Haoxiang Cheng
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kenny L Chan
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hsiao-Yun Lin
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kalena Liu
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lyonna F Parise
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Flurin Cathomas
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Katherine B Leclair
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Meghan E Flanigan
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Long Li
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hossein Aleyasin
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christopher Guevara
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ke Hao
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Icahn Institute of Genomics and Multi-Scale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Scott J Russo
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jun Wang
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Geriatric Research, Education and Clinical Center, James J. Peters Veterans Affairs Medical Center, Bronx, NY, USA
| |
Collapse
|
189
|
Circulating T helper 17 and IFN-γ positive Th17 cells in Major Depressive Disorder. Behav Brain Res 2020; 394:112811. [DOI: 10.1016/j.bbr.2020.112811] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 07/14/2020] [Accepted: 07/15/2020] [Indexed: 01/20/2023]
|
190
|
Childhood trauma, suicide risk and inflammatory phenotypes of depression: insights from monocyte gene expression. Transl Psychiatry 2020; 10:296. [PMID: 32839428 PMCID: PMC7445278 DOI: 10.1038/s41398-020-00979-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 07/14/2020] [Accepted: 07/22/2020] [Indexed: 12/20/2022] Open
Abstract
Circulating monocytes contribute to inflammatory processes. We here validate abnormal expression of inflammation-related genes in monocytes of a large and well-characterised group of MDD patients, and relate the outcomes to pertinent clinical characteristics. Thirty-two genes of a previously established inflammation-related gene signature were assessed in 197 patients with MDD, and 151 controls collected during the EU-MOODINFLAME project. Monocyte gene- expression data were related to age, sex, BMI, depression severity, childhood adversity (CA) and suicide risk (SR). Three distinct gene profiles were identified within the MDD group (downregulated, mixed upregulated and strongly upregulated genes). Patients in the merged upregulated groups had a significantly higher prevalence of CA and high SR. Using hierarchical clustering of the genes, we found a cluster of mainly cytokine (production)-related genes; patients with SR had a significantly higher expression of this cluster than patients without SR (particularly for IL-6, IL1A and IL1B). Such difference did not emerge for patients with and without CA. A downregulated gene profile was found for patients not exposed to CA and without SR (particularly for glucocorticoid-signalling genes NR3C1a and HSPA1/B). No inflammatory changes were observed for healthy controls exposed to CA. Our data show that inflammatory activation in MDD is not uniform, and that immunologically discernible phenotypes of depression can be linked to CA and high SR. The absence of monocyte inflammatory activation in healthy controls exposed to CA suggests an inflammatory involvement in MDD-prone individuals exposed to early stressors, but not healthy controls.
Collapse
|
191
|
Abstract
Depression is one of the most common comorbidities of many chronic medical diseases including cancer and cardiovascular, metabolic, inflammatory and neurological disorders. Indeed, the prevalence of depression in these patient groups is often substantially higher than in the general population, and depression accounts for a substantial part of the psychosocial burden of these disorders. Many factors can contribute to the occurrence of comorbid depression, such as shared genetic factors, converging biological pathways, social factors, health behaviours and psychological factors. Diagnosis of depression in patients with a medical disorder can be particularly challenging owing to symptomatic overlap. Although pharmacological and psychological treatments can be effective, adjustments may need to be made for patients with a comorbid medical disorder. In addition, symptoms or treatments of medical disorders may interfere with the treatment of depression. Conversely, symptoms of depression may decrease adherence to treatment of both disorders. Thus, comprehensive treatment plans are necessary to optimize care.
Collapse
|
192
|
Dattilo V, Amato R, Perrotti N, Gennarelli M. The Emerging Role of SGK1 (Serum- and Glucocorticoid-Regulated Kinase 1) in Major Depressive Disorder: Hypothesis and Mechanisms. Front Genet 2020; 11:826. [PMID: 32849818 PMCID: PMC7419621 DOI: 10.3389/fgene.2020.00826] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 07/09/2020] [Indexed: 12/28/2022] Open
Abstract
Major depressive disorder (MDD) is a heterogeneous psychiatric disease characterized by persistent low mood, diminished interests, and impaired cognitive and social functions. The multifactorial etiology of MDD is still largely unknown because of the complex genetic and environmental interactions involved. Therefore, no established mechanism can explain all the aspects of the disease. In this light, an extensive research about the pathophysiology of MDD has been carried out. Several pathogenic hypotheses, such as monoamines deficiency and neurobiological alterations in the stress-responsive system, including the hypothalamic-pituitary-adrenal (HPA) axis and the immune system, have been proposed for MDD. Over time, remarkable studies, mainly on preclinical rodent models, linked the serum- and glucocorticoid-regulated kinase 1 (SGK1) to the main features of MDD. SGK1 is a serine/threonine kinase belonging to the AGK Kinase family. SGK1 is ubiquitously expressed, which plays a pivotal role in the hormonal regulation of several ion channels, carriers, pumps, and transcription factors or regulators. SGK1 expression is modulated by cell stress and hormones, including gluco- and mineralocorticoids. Compelling evidence suggests that increased SGK1 expression or function is related to the pathogenic stress hypothesis of major depression. Therefore, the first part of the present review highlights the putative role of SGK1 as a critical mediator in the dysregulation of the HPA axis, observed under chronic stress conditions, and its controversial role in the neuroinflammation as well. The second part depicts the negative regulation exerted by SGK1 in the expression of both the brain-derived neurotrophic factor (BDNF) and the vascular endothelial growth factor (VEGF), resulting in an anti-neurogenic activity. Finally, the review focuses on the antidepressant-like effects of anti-oxidative nutraceuticals in several preclinical model of depression, resulting from the restoration of the physiological expression and/or activity of SGK1, which leads to an increase in neurogenesis. In summary, the purpose of this review is a systematic analysis of literature depicting SGK1 as molecular junction of the complex mechanisms underlying the MDD in an effort to suggest the kinase as a potential biomarker and strategic target in modern molecular antidepressant therapy.
Collapse
Affiliation(s)
- Vincenzo Dattilo
- Genetic Unit, IRCCS Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Rosario Amato
- Department of Health Sciences, Magna Graecia University of Catanzaro, Catanzaro, Italy.,Medical Genetics Unit, Mater Domini University Hospital, Catanzaro, Italy
| | - Nicola Perrotti
- Department of Health Sciences, Magna Graecia University of Catanzaro, Catanzaro, Italy.,Medical Genetics Unit, Mater Domini University Hospital, Catanzaro, Italy
| | - Massimo Gennarelli
- Genetic Unit, IRCCS Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.,Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
193
|
Zhang Q, Sun Y, He Z, Xu Y, Li X, Ding J, Lu M, Hu G. Kynurenine regulates NLRP2 inflammasome in astrocytes and its implications in depression. Brain Behav Immun 2020; 88:471-481. [PMID: 32283293 DOI: 10.1016/j.bbi.2020.04.016] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/27/2020] [Accepted: 04/07/2020] [Indexed: 12/21/2022] Open
Abstract
Increased kynurenine (Kyn) metabolized from tryptophan (Try) is a biomarker in the immune dysfunction of depression. However, the mechanism by which Kyn change promotes depression is poorly defined. Astrocytes are involved in the neuroinflammation of depression. Among the numerous inflammatory cytokines, interleukin-1β (IL-1β) produced by astrocytic Nod-like receptor protein (NLRP) inflammasome is crucial in the pathogenesis of depression. In the present study, Kyn was shown to be a proinflammatory metabolite in the neuroimmune signaling network mediating depressive-like behavior. First, in chronic mild stress (CMS)-induced depressive mice, the level of Kyn notably increased in the hippocampus, accompanied by the activation of astrocytic NLRP2 inflammasome. Kyn treatment specifically upregulated Nod-like receptor protein 2 (NLPR2) expression in primary mouse astrocytes. Kyn + ATP activated NLRP2 inflammasome, evidenced by increased caspase-1 expression and IL-1β release. After Kyn treatment, nuclear factor kappa-B (NF-κB) could translocate to the nucleus and bind the promoter of NLRP2, subsequently increased NLRP2 transcription in cultured astrocytes in vitro. Intraperitoneal injection of Kyn activated NLRP2 inflammasome in astrocytes of hippocampus in mice, while NLRP2 knockdown in astrocytes abolished depressive-like behaviors in mice induced by Kyn, suggesting the critical role of NLRP2 in Kyn-induced depression. These findings demonstrate a novel mechanism that Kyn upregulates NLRP2 in an NF-κB-dependent pathway and provide a new strategy for treatment of depression.
Collapse
Affiliation(s)
- Qingyu Zhang
- Department of Pharmacology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Yiming Sun
- Department of Pharmacology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Zhang He
- Department of Pharmacology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Ying Xu
- Department of Pharmacology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Xueting Li
- Department of Pharmacology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Jianhua Ding
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu 211166, China
| | - Ming Lu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu 211166, China
| | - Gang Hu
- Department of Pharmacology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China; Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, Jiangsu 211166, China.
| |
Collapse
|
194
|
Shurin MR, Shurin GV, Zlotnikov SB, Bunimovich YL. The Neuroimmune Axis in the Tumor Microenvironment. THE JOURNAL OF IMMUNOLOGY 2020; 204:280-285. [PMID: 31907270 DOI: 10.4049/jimmunol.1900828] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 08/23/2019] [Indexed: 12/14/2022]
Abstract
Cancer is a complex ecosystem and should be considered in the context of its cellular and molecular microenvironment, which includes the nerves. Peripheral nerves can modulate phenotype and behavior of the malignant cells and thus affect tumor growth and metastasis. Only recently has the role of neuroimmune cross-talk surfaced as a key contributor to cancer progression. However, little is known about the immunomodulatory role of the neuroglial cells in cancer progression and metastasis and the response to therapy. Schwann cells, the principal glial cells of the peripheral nervous system, are now considered to be important players in the tumor microenvironment. They can directly accelerate malignant cell migration and the formation of metastases. Better understanding of the neuroimmune circuits in the tumor milieu will be instrumental in the development of novel therapeutic approaches for the malignancies known to be associated with inflammation and dysregulated immune responses.
Collapse
Affiliation(s)
- Michael R Shurin
- Department of Pathology, University of Pittsburgh Medical Center and University of Pittsburgh Cancer Institute, Pittsburgh, PA 15232; .,Department of Immunology, University of Pittsburgh Medical Center and University of Pittsburgh Cancer Institute, Pittsburgh, PA 15232; and
| | - Galina V Shurin
- Department of Pathology, University of Pittsburgh Medical Center and University of Pittsburgh Cancer Institute, Pittsburgh, PA 15232
| | - Samuel B Zlotnikov
- Department of Pathology, University of Pittsburgh Medical Center and University of Pittsburgh Cancer Institute, Pittsburgh, PA 15232
| | - Yuri L Bunimovich
- Department of Dermatology, University of Pittsburgh Medical Center and University of Pittsburgh Cancer Institute, Pittsburgh, PA 15232
| |
Collapse
|
195
|
Kokkosis AG, Tsirka SE. Neuroimmune Mechanisms and Sex/Gender-Dependent Effects in the Pathophysiology of Mental Disorders. J Pharmacol Exp Ther 2020; 375:175-192. [PMID: 32661057 DOI: 10.1124/jpet.120.266163] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 07/09/2020] [Indexed: 12/12/2022] Open
Abstract
Innate and adaptive immune mechanisms have emerged as critical regulators of CNS homeostasis and mental health. A plethora of immunologic factors have been reported to interact with emotion- and behavior-related neuronal circuits, modulating susceptibility and resilience to mental disorders. However, it remains unclear whether immune dysregulation is a cardinal causal factor or an outcome of the pathologies associated with mental disorders. Emerging variations in immune regulatory pathways based on sex differences provide an additional framework for discussion in these psychiatric disorders. In this review, we present the current literature pertaining to the effects that disrupted immune pathways have in mental disorder pathophysiology, including immune dysregulation in CNS and periphery, microglial activation, and disturbances of the blood-brain barrier. In addition, we present the suggested origins of such immune dysregulation and discuss the gender and sex influence of the neuroimmune substrates that contribute to mental disorders. The findings challenge the conventional view of these disorders and open the window to a diverse spectrum of innovative therapeutic targets that focus on the immune-specific pathophenotypes in neuronal circuits and behavior. SIGNIFICANCE STATEMENT: The involvement of gender-dependent inflammatory mechanisms on the development of mental pathologies is gaining momentum. This review addresses these novel factors and presents the accumulating evidence introducing microglia and proinflammatory elements as critical components and potential targets for the treatment of mental disorders.
Collapse
Affiliation(s)
- Alexandros G Kokkosis
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York
| | - Stella E Tsirka
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York
| |
Collapse
|
196
|
Zavvari F, Nahavandi A, Shahbazi A. Neuroprotective effects of cerium oxide nanoparticles on experimental stress-induced depression in male rats. J Chem Neuroanat 2020; 106:101799. [DOI: 10.1016/j.jchemneu.2020.101799] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/19/2020] [Accepted: 04/20/2020] [Indexed: 12/12/2022]
|
197
|
Deng XF, Shi GQ, Guo LL, Zhu CA, Chen YJ. Analysis on Risk Factors of Depressive Symptoms in Occupational Noise-induced Hearing Loss Patients: A Cross-sectional Study. Noise Health 2020; 21:17-24. [PMID: 32098927 PMCID: PMC7050233 DOI: 10.4103/nah.nah_16_18] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Objective The aim of this study was to evaluate the risk factors of depressive symptoms in occupational noise-induced hearing loss (NIHL) patients. Methods A total of 106 patients were divided into depressive symptoms (ONHLPD) and without depressive symptoms (non-ONHLPD) according to the Self-rating Depression Scale. Questionnaires and laboratory data were collected and analyzed. Data were analyzed with independent t-test, Wilcoxon test, Pearson correlation analysis and multiple linear regression models. Results The prevalence of depressive symptoms was 53.8% in occupational NIHL patients. In ONHLPD, duration of the hearing loss, level of serum cortisol, scores of Pittsburgh Sleep Quality Index and Tinnitus Handicap Inventory were all significantly higher than those of non-ONHLPD. Conclusion The prevalence of depressive symptoms was relatively high in occupational NIHL patients. Duration of the hearing loss, sleep quality and tinnitus severity were the risk factors for occupational NHIL patients with depressive symptoms.
Collapse
Affiliation(s)
- Xiao-Feng Deng
- South China Research Center for Acupuncture and Moxibustion, Clinical Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong; Guangdong Provincial Hospital for Occupational Disease Prevention and Treatment, Guangzhou, Guangdong Province, PR China
| | - Guo-Qi Shi
- South China Research Center for Acupuncture and Moxibustion, Clinical Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China
| | - Li-Li Guo
- South China Research Center for Acupuncture and Moxibustion, Clinical Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China
| | - Chuan-An Zhu
- South China Research Center for Acupuncture and Moxibustion, Clinical Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China
| | - Yong-Jun Chen
- South China Research Center for Acupuncture and Moxibustion, Clinical Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong; Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong-Macao Greater Bay Area, Guangzhou, PR China
| |
Collapse
|
198
|
Wang J, Zhao D, Tiano S, Esteban-Fernández A, Yuan B, Smith C, Brathwaite J, Jlayer Z, Wu Q, Simon JE, Trageser KJ, Pasinetti GM. Prophylactic effect of flavanol rich preparation metabolites in promoting resilience to a mouse model of social stress. Transl Psychiatry 2020; 10:183. [PMID: 32518327 PMCID: PMC7283298 DOI: 10.1038/s41398-020-0859-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 05/04/2020] [Accepted: 05/15/2020] [Indexed: 01/16/2023] Open
Abstract
Major depressive disorder (MDD) is a leading cause of disability, and there is an urgent need for new therapeutics. Stress-mediated induction of pro-inflammation in the periphery contributes to depression-like behaviors both in humans and in experimental models. Inflammatory cytokine interleukin-6 (IL-6) has emerged as a potential therapeutic target. Our studies demonstrated that metabolism of flavanol rich cocoa preparation (FRP) led to the accumulation of select phenolic acids that may contribute to its anti-inflammatory activity. Using a repeated social defeat stress (RSDS) model of depression, we showed that oral administration of FRP attenuates susceptibility to RSDS-mediated depression, supporting the further development of FRP as a novel therapeutic for the treatment of stress disorders and anxiety in humans.
Collapse
Affiliation(s)
- Jun Wang
- grid.59734.3c0000 0001 0670 2351Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA ,grid.274295.f0000 0004 0420 1184Geriatric Research, Education and Clinical Center, James J. Peters Veterans Affairs Medical Center, Bronx, New York, NY 10468 USA
| | - Danyue Zhao
- grid.430387.b0000 0004 1936 8796New Use Agriculture & Natural Plant Products Program, Department of Plant Biology, School of Environmental and Biological Sciences, Rutgers University, New Brunswick, NJ 08901 USA
| | - Simoni Tiano
- grid.59734.3c0000 0001 0670 2351Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Adelaida Esteban-Fernández
- grid.59734.3c0000 0001 0670 2351Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Bo Yuan
- grid.430387.b0000 0004 1936 8796New Use Agriculture & Natural Plant Products Program, Department of Plant Biology, School of Environmental and Biological Sciences, Rutgers University, New Brunswick, NJ 08901 USA
| | - Chad Smith
- grid.59734.3c0000 0001 0670 2351Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Justin Brathwaite
- grid.59734.3c0000 0001 0670 2351Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Zahra Jlayer
- grid.59734.3c0000 0001 0670 2351Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Qingli Wu
- grid.430387.b0000 0004 1936 8796New Use Agriculture & Natural Plant Products Program, Department of Plant Biology, School of Environmental and Biological Sciences, Rutgers University, New Brunswick, NJ 08901 USA
| | - James E. Simon
- grid.274295.f0000 0004 0420 1184Geriatric Research, Education and Clinical Center, James J. Peters Veterans Affairs Medical Center, Bronx, New York, NY 10468 USA
| | - Kyle J. Trageser
- grid.59734.3c0000 0001 0670 2351Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Giulio M. Pasinetti
- grid.59734.3c0000 0001 0670 2351Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA ,grid.274295.f0000 0004 0420 1184Geriatric Research, Education and Clinical Center, James J. Peters Veterans Affairs Medical Center, Bronx, New York, NY 10468 USA
| |
Collapse
|
199
|
Systemic immunization with altered myelin basic protein peptide produces sustained antidepressant-like effects. Mol Psychiatry 2020; 25:1260-1274. [PMID: 31375779 DOI: 10.1038/s41380-019-0470-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 06/17/2019] [Accepted: 06/24/2019] [Indexed: 12/21/2022]
Abstract
Immune dysregulation, specifically of inflammatory processes, has been linked to behavioral symptoms of depression in both human and rodent studies. Here, we evaluated the antidepressant effects of immunization with altered peptide ligands of myelin basic protein (MBP)-MBP87-99[A91, A96], MBP87-99[A91], and MBP87-99[R91, A96]-in different models of depression and examined the mechanism by which these peptides protect against stress-induced depression. We found that a single dose of subcutaneously administered MBP87-99[A91, A96] produced antidepressant-like effects by decreasing immobility in the forced swim test and by reducing the escape latency and escape failures in the learned helplessness paradigm. Moreover, immunization with MBP87-99[A91, A96] prevented and reversed depressive-like and anxiety-like behaviors that were induced by chronic unpredictable stress (CUS). However, MBP87-99[R91, A96] tended to aggravate CUS-induced anxiety-like behavior. Chronic stress increased the production of peripheral and central proinflammatory cytokines and induced the activation of microglia in the prelimbic cortex (PrL), which was blocked by MBP87-99[A91, A96]. Immunization with MBP-derived altered peptide ligands also rescued chronic stress-induced deficits in p11, phosphorylated cyclic adenosine monophosphate response element binding protein, and brain-derived neurotrophic factor expression. Moreover, microinjections of recombinant proinflammatory cytokines and the knockdown of p11 in the PrL blunted the antidepressant-like behavioral response to MBP87-99[A91, A96]. Altogether, these findings indicate that immunization with altered MBP peptide produces prolonged antidepressant-like effects in rats, and the behavioral response is mediated by inflammatory factors (particularly interleukin-6), and p11 signaling in the PrL. Immune-neural interactions may impact central nervous system function and alter an individual's response to stress.
Collapse
|
200
|
Mason BL. Mapping Immune System Dysfunction to Provide Clinically Actionable Biomarkers and to Understand Psychiatric Pathology. Psychiatr Ann 2020. [DOI: 10.3928/00485713-20200506-01] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|