51
|
Melanson B, Leri F. Effect of ketamine on the physiological responses to combined hypoglycemic and psychophysical stress. IBRO Neurosci Rep 2021; 11:81-87. [PMID: 34485972 PMCID: PMC8406162 DOI: 10.1016/j.ibneur.2021.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/20/2021] [Indexed: 12/01/2022] Open
Abstract
There is evidence that hypoglycemic stress can interact with other stressors, and that ketamine can mitigate the impact of these stressors on behavior and physiology. The current study in male Sprague-Dawley rats investigated whether pre-treatment with 0, 10, or 20 mg/kg ketamine could modulate the interaction between hypoglycemia induced by 0 or 300 mg/kg 2-deoxy-D-glucose (2-DG) and the psychophysical stress of forced swimming (FSS; 6 sessions, 10 min/session) on serum concentrations of corticosterone (CORT) and the pro-inflammatory cytokine, tumor necrosis factor (TNF)-α. It was found that 2-DG enhanced the CORT response to an initial session of FSS, and this effect dissipated after multiple sessions. More importantly, animals displayed significantly higher levels of CORT and lower levels of TNF-α in response to a drug-free test swim conducted 1 week after exposure to the combined stressors, and these responses were not observed in rats that were pre-treated with ketamine. Overall, these findings indicate that ketamine has the potential to reduce the negative impact of interacting stressors on the biological reactivity of the hypothalamic-pituitary-adrenal axis and the immune system.
Collapse
Affiliation(s)
- Brett Melanson
- Department of Psychology and Collaborative Neuroscience, Program University of Guelph, Guelph, ON, Canada
| | - Francesco Leri
- Department of Psychology and Collaborative Neuroscience, Program University of Guelph, Guelph, ON, Canada
| |
Collapse
|
52
|
Nasca C, Dobbin J, Bigio B, Watson K, de Angelis P, Kautz M, Cochran A, Mathé AA, Kocsis JH, Lee FS, Murrough JW, McEwen BS, Rasgon N. Insulin receptor substrate in brain-enriched exosomes in subjects with major depression: on the path of creation of biosignatures of central insulin resistance. Mol Psychiatry 2021; 26:5140-5149. [PMID: 32536688 PMCID: PMC7787430 DOI: 10.1038/s41380-020-0804-7] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 05/17/2020] [Accepted: 05/28/2020] [Indexed: 01/08/2023]
Abstract
Insulin signaling is critical for neuroplasticity, cerebral metabolism as well as for systemic energy metabolism. In rodent studies, impaired brain insulin signaling with resultant insulin resistance (IR) modulates synaptic plasticity and the corresponding behavioral functions. Despite discoveries of central actions of insulin, in vivo molecular mechanisms of brain IR until recently have proven difficult to study in the human brain. In the current study, we leveraged recent technological advances in molecular biology and herein report an increased number of exosomes enriched for L1CAM, a marker predominantly expressed in the brain, in subjects with major depressive disorder (MDD) as compared with age- and sex-matched healthy controls (HC). We also report increased concentration of the insulin receptor substrate-1 (IRS-1) in L1CAM+ exosomes in subjects with MDD as compared with age- and sex-matched HC. We found a relationship between expression of IRS-1 in L1CAM+ exosomes and systemic IR as assessed by homeostatic model assessment of IR in HC, but not in subjects with MDD. The increased IRS-1 levels in L1CAM+ exosomes were greater in subjects with MDD and were associated with suicidality and anhedonia. Finally, our data suggested sex differences in serine-312 phosphorylation of IRS-1 in L1CAM+ exosomes in subjects with MDD. These findings provide a starting point for creating mechanistic framework of brain IR in further development of personalized medicine strategies to effectively treat MDD.
Collapse
Affiliation(s)
- Carla Nasca
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY, USA.
| | - Josh Dobbin
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, USA
| | - Benedetta Bigio
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, USA
| | - Kathleen Watson
- Center for Neuroscience in Women’s Health, Stanford University, Palo Alto, CA 91304, USA
| | - Paolo de Angelis
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, USA
| | - Marin Kautz
- Mood and Anxiety Disorders Program, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY, USA
| | - Ashly Cochran
- Department of Psychiatry, Weill Cornell Medical College, New York, NY, USA
| | - Aleksander A Mathé
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - James H Kocsis
- Department of Psychiatry, Weill Cornell Medical College, New York, NY, USA
| | - Francis S Lee
- Department of Psychiatry, Weill Cornell Medical College, New York, NY, USA.,Sackler Institute for Developmental Psychobiology, Weill Cornell Medical College, New York, NY, USA
| | - James W Murrough
- Mood and Anxiety Disorders Program, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY, USA
| | - Bruce S McEwen
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, USA
| | - Natalie Rasgon
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY, USA. .,Center for Neuroscience in Women's Health, Stanford University, Palo Alto, CA, 91304, USA.
| |
Collapse
|
53
|
The kynurenine pathway and bipolar disorder: intersection of the monoaminergic and glutamatergic systems and immune response. Mol Psychiatry 2021; 26:4085-4095. [PMID: 31732715 PMCID: PMC7225078 DOI: 10.1038/s41380-019-0589-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 10/21/2019] [Accepted: 10/30/2019] [Indexed: 12/19/2022]
Abstract
Dysfunction in a wide array of systems-including the immune, monoaminergic, and glutamatergic systems-is implicated in the pathophysiology of depression. One potential intersection point for these three systems is the kynurenine (KYN) pathway. This study explored the impact of the prototypic glutamatergic modulator ketamine on the endogenous KYN pathway in individuals with bipolar depression (BD), as well as the relationship between response to ketamine and depression-related behavioral and peripheral inflammatory markers. Thirty-nine participants with treatment-resistant BD (23 F, ages 18-65) received a single ketamine infusion (0.5 mg/kg) over 40 min. KYN pathway analytes-including plasma concentrations of indoleamine 2,3-dioxygenase (IDO), KYN, kynurenic acid (KynA), and quinolinic acid (QA)-were assessed at baseline (pre-infusion), 230 min, day 1, and day 3 post-ketamine. General linear models with restricted maximum likelihood estimation and robust sandwich variance estimators were implemented. A repeated effect of time was used to model the covariance of the residuals with an unstructured matrix. After controlling for age, sex, and body mass index (BMI), post-ketamine IDO levels were significantly lower than baseline at all three time points. Conversely, ketamine treatment significantly increased KYN and KynA levels at days 1 and 3 versus baseline. No change in QA levels was observed post-ketamine. A lower post-ketamine ratio of QA/KYN was observed at day 1. In addition, baseline levels of proinflammatory cytokines and behavioral measures predicted KYN pathway changes post ketamine. The results suggest that, in addition to having rapid and sustained antidepressant effects in BD participants, ketamine also impacts key components of the KYN pathway.
Collapse
|
54
|
Kopra E, Mondelli V, Pariante C, Nikkheslat N. Ketamine's effect on inflammation and kynurenine pathway in depression: A systematic review. J Psychopharmacol 2021; 35:934-945. [PMID: 34180293 PMCID: PMC8358579 DOI: 10.1177/02698811211026426] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Ketamine is a novel rapid-acting antidepressant with high efficacy in treatment-resistant patients. Its exact therapeutic mechanisms of action are unclear; however, in recent years its anti-inflammatory properties and subsequent downstream effects on tryptophan (TRP) metabolism have sparked research interest. AIM This systematic review examined the effect of ketamine on inflammatory markers and TRP-kynurenine (KYN) pathway metabolites in patients with unipolar and bipolar depression and in animal models of depression. METHODS MEDLINE, Embase, and PsycINFO databases were searched on October 2020 (1806 to 2020). RESULTS Out of 807 initial results, nine human studies and 22 animal studies on rodents met the inclusion criteria. Rodent studies provided strong support for ketamine-induced decreases in pro-inflammatory cytokines, namely in interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α and indicated anti-inflammatory effects on TRP metabolism, including decreases in the enzyme indoleamine 2,3-dioxygenase (IDO). Clinical evidence was less robust with high heterogeneity between sample characteristics, but most experiments demonstrated decreases in peripheral inflammation including in IL-1β, IL-6, and TNF-α. Preliminary support was also found for reduced activation of the neurotoxic arm of the KYN pathway. CONCLUSION Ketamine appears to induce anti-inflammatory effects in at least a proportion of depressed patients. Suggestions for future research include investigation of markers in the central nervous system and examination of clinical relevance of inflammatory changes.
Collapse
Affiliation(s)
- Emma Kopra
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| | - Valeria Mondelli
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
- National Institute for Health Research (NIHR) Mental Health Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London, UK
| | - Carmine Pariante
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
- National Institute for Health Research (NIHR) Mental Health Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London, UK
| | - Naghmeh Nikkheslat
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| |
Collapse
|
55
|
Lucerne KE, Osman A, Meckel KR, Kiraly DD. Contributions of neuroimmune and gut-brain signaling to vulnerability of developing substance use disorders. Neuropharmacology 2021; 192:108598. [PMID: 33965398 PMCID: PMC8220934 DOI: 10.1016/j.neuropharm.2021.108598] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 03/19/2021] [Accepted: 05/03/2021] [Indexed: 02/06/2023]
Abstract
Epidemiology and clinical research indicate that only a subset of people who are exposed to drugs of abuse will go on to develop a substance use disorder. Numerous factors impact individual susceptibility to developing a substance use disorder, including intrinsic biological factors, environmental factors, and interpersonal/social factors. Given the extensive morbidity and mortality that is wrought as a consequence of substance use disorders, a substantial body of research has focused on understanding the risk factors that mediate the shift from initial drug use to pathological drug use. Understanding these risk factors provides a clear path for the development of risk mitigation strategies to help reduce the burden of substance use disorders in the population. Here we will review the rapidly growing body of literature that examines the importance of interactions between the peripheral immune system, the gut microbiome, and the central nervous system (CNS) in mediating the transition to pathological drug use. While these systems had long been viewed as distinct, there is growing evidence that there is bidirectional communication between both the immune system and the gut microbiome that drive changes in neural and behavioral plasticity relevant to substance use disorders. Further, both of these systems are highly sensitive to environmental perturbations and are implicated in numerous neuropsychiatric conditions. While the field of study examining these interactions in substance use disorders is in its relative infancy, clarifying the relationship between gut-immune-brain signaling and substance use disorders has potential to improve our understanding of individual propensity to developing addiction and yield important insight into potential treatment options.
Collapse
Affiliation(s)
- Kelsey E Lucerne
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Aya Osman
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Katherine R Meckel
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Drew D Kiraly
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
56
|
Wilkowska A, Szałach ŁP, Cubała WJ. Gut Microbiota in Depression: A Focus on Ketamine. Front Behav Neurosci 2021; 15:693362. [PMID: 34248517 PMCID: PMC8261217 DOI: 10.3389/fnbeh.2021.693362] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 05/27/2021] [Indexed: 01/09/2023] Open
Abstract
According to the WHO, major depressive disorder is the leading cause of disability worldwide, and it is a major contributor to the overall global burden of disease. The pathophysiology of this common and chronic disease is still not completely understood. The gut microbiome is an increasingly recognized environmental factor that can have a role in depression, acting through the gut-microbiota-brain axis. The available treatment for depression is still insufficient since 30% of patients are treatment-resistant. There is an unquestionable need for novel strategies. Ketamine is an effective antidepressant in treatment-resistant patients. It is suggested that the antidepressant effect of ketamine may be partially mediated by the modification of gut microbiota. In this study, we presented a review of data on gut microbiota in depression with special attention to the effect of ketamine on the microbiome in animal models of depression. Earlier reports are preliminary and are still insufficient to draw firm conclusion, but further studies in this field might help to understand the role of the gut-brain axis in the treatment of depression and might be the ground for developing new effective treatment strategies.
Collapse
|
57
|
Jones GH, Vecera CM, Pinjari OF, Machado-Vieira R. Inflammatory signaling mechanisms in bipolar disorder. J Biomed Sci 2021; 28:45. [PMID: 34112182 PMCID: PMC8194019 DOI: 10.1186/s12929-021-00742-6] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/08/2021] [Indexed: 12/12/2022] Open
Abstract
Bipolar disorder is a decidedly heterogeneous and multifactorial disease, with a high individual and societal burden. While not all patients display overt markers of elevated inflammation, significant evidence suggests that aberrant immune signaling contributes to all stages of the disease, and likely explains the elevated rates of comorbid inflammatory illnesses seen in this population. While individual systems have been intensely studied and targeted, a relative paucity of attention has been given to the interconnecting role of inflammatory signals therein. This review presents an updated overview of some of the most prominent pathophysiologic mechanisms in bipolar disorder, from mitochondrial, endoplasmic reticular, and calcium homeostasis, to purinergic, kynurenic, and hormonal/neurotransmitter signaling, showing inflammation to act as a powerful nexus between these systems. Several areas with a high degree of mechanistic convergence within this paradigm are highlighted to present promising future targets for therapeutic development and screening.
Collapse
Affiliation(s)
- Gregory H Jones
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at Houston (UTHealth), 1941 East Road, Houston, TX, 77054, USA.
| | - Courtney M Vecera
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at Houston (UTHealth), 1941 East Road, Houston, TX, 77054, USA
| | - Omar F Pinjari
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at Houston (UTHealth), 1941 East Road, Houston, TX, 77054, USA
| | - Rodrigo Machado-Vieira
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at Houston (UTHealth), 1941 East Road, Houston, TX, 77054, USA
| |
Collapse
|
58
|
Zhang N, Yao L, Wang P, Liu Z. Immunoregulation and antidepressant effect of ketamine. Transl Neurosci 2021; 12:218-236. [PMID: 34079622 PMCID: PMC8155793 DOI: 10.1515/tnsci-2020-0167] [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: 11/01/2020] [Revised: 04/04/2021] [Accepted: 04/06/2021] [Indexed: 12/15/2022] Open
Abstract
Major depressive disorder (MDD) is a common mental health disorder that brings severe disease burden worldwide. Traditional antidepressants are mainly targeted at monoamine neurotransmitters, with low remission rates and high recurrence rates. Ketamine is a noncompetitive glutamate N-methyl-d-aspartate receptor (NMDAR) antagonist, and its rapid and powerful antidepressant effects have come to light. Its antidepressant mechanism is still unclarified. Research found that ketamine had not only antagonistic effect on NMDAR but also strong immunomodulatory effect, both of which were closely related to the pathophysiology of MDD. Although there are many related studies, they are relatively heterogeneous. Therefore, this review mainly describes the immune mechanisms involved in MDD and how ketamine plays an antidepressant role by regulating peripheral and central immune system, including peripheral inflammatory cytokines, central microglia, and astrocytes. This review summarizes the related research, finds out the deficiencies of current research, and provides ideas for future research and the development of novel antidepressants.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Rd. 238, 430060, Wuhan, China
| | - Lihua Yao
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Rd. 238, 430060, Wuhan, China
| | - Peilin Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Rd. 238, 430060, Wuhan, China
| | - Zhongchun Liu
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Rd. 238, 430060, Wuhan, China
| |
Collapse
|
59
|
Lan X, Zhou Y, Wu F, Wu K, Zhan Y, Wang C, Zheng W, Yu M, Deng X, Ning Y. The relationship between plasma cytokine levels and antidepressant response in patients with first-episode major depressive disorder. J Affect Disord 2021; 287:327-333. [PMID: 33813252 DOI: 10.1016/j.jad.2021.03.036] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/12/2021] [Accepted: 03/15/2021] [Indexed: 12/28/2022]
Abstract
BACKGROUND Cytokines are involved in the pathophysiology of major depressive disorder (MDD) and treatment response. Efforts have been made to identify inflammatory markers but results are often contradictory. The present study explored the plasma levels of multiple cytokines in first-episode MDD using a longitudinal design, with the aim to determine the involvement of cytokines in depression and identify the inflammatory markers. METHOD Fifty-four first-episode drug naïve MDD patients and 60 healthy controls (HCs) were enrolled in this study. The 17-item Hamilton Depression Rating Scale (HAMD-17) was administered and blood samples were collected at baseline and four-week posttreatment in MDD group, while blood samples were only collected once in HC group. Plasma levels of nineteen cytokines were examined by a multiplexed flow cytometric assay. RESULTS Sixteen out of 19 cytokines levels in MDD group were significantly higher than those in HC group (all P < 0.05). After 4-week of antidepressant treatment, levels of the 14 cytokines elevated at baseline decreased to normal levels (all P < 0.05). Partial correlation showed that baseline level of interferon-inducible T cell alpha chemoattractant (ITAC) was negatively correlated with reduction in HAMD-17 score (r=-0.319, p=0.020), and multiple regression showed lower baseline ITAC level was associated with better treatment response (p = 0.020). LIMITATION The sample size was relatively small. CONCLUSION A range of cytokines were abnormal in patients with first-episode drug naïve MDD and most of the cytokines could be normalized after antidepressant treatment. Furthermore, baseline ITAC level could be a predictive factor of antidepressant response.
Collapse
Affiliation(s)
- Xiaofeng Lan
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Yanling Zhou
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Fengchun Wu
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Kai Wu
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China; Department of Biomedical Engineering, School of Material Science and Engineering, South China University of Technology, Guangzhou, China
| | - Yanni Zhan
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Chengyu Wang
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Wei Zheng
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Min Yu
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Xiurong Deng
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Yuping Ning
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China; The First School of Clinical Medicine, Southern Medical University, Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China.
| |
Collapse
|
60
|
Methiwala HN, Vaidya B, Addanki VK, Bishnoi M, Sharma SS, Kondepudi KK. Gut microbiota in mental health and depression: role of pre/pro/synbiotics in their modulation. Food Funct 2021; 12:4284-4314. [PMID: 33955443 DOI: 10.1039/d0fo02855j] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The microbiome residing in the human gut performs a wide range of biological functions. Recently, it has been elucidated that a change in dietary habits is associated with alteration in the gut microflora which results in increased health risks and vulnerability towards various diseases. Falling in line with the same concept, depression has also been shown to increase its prevalence around the globe, especially in the western world. Various research studies have suggested that changes in the gut microbiome profile further result in decreased tolerance of stress. Although currently available medications help in relieving the symptoms of depressive disorders briefly, these drugs are not able to completely reverse the multifactorial pathology of depression. The discovery of the communication pathway between gut microbes and the brain, i.e. the Gut-Brain Axis, has led to new areas of research to find more effective and safer alternatives to current antidepressants. The use of probiotics and prebiotics has been suggested as being effective in various preclinical studies and clinical trials for depression. Therefore, in the present review, we address the new antidepressant mechanisms via gut microbe alterations and provide insight into how these can provide an alternative to antidepressant therapy without the side effects and risk of adverse drug reactions.
Collapse
Affiliation(s)
- Hasnain N Methiwala
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab, India.
| | | | | | | | | | | |
Collapse
|
61
|
M1/M2 polarization in major depressive disorder: Disentangling state from trait effects in an individualized cell-culture-based approach. Brain Behav Immun 2021; 94:185-195. [PMID: 33607231 DOI: 10.1016/j.bbi.2021.02.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 02/06/2021] [Accepted: 02/09/2021] [Indexed: 12/17/2022] Open
Abstract
Accumulating evidence indicates the specific involvement of inflammatory processes in major depressive disorder (MDD), particularly affecting innate immunity. Most immune alterations have so far been determined based on plasma or cerebrospinal fluid cytokine levels. To precisely characterize putative innate immune-mediated mechanisms in MDD pathogenesis, we sought to disentangle "state" from "trait" effects in a patient-specific cell model by quantifying the impact of patient-derived autologous sera (AS) on patient-specific monocyte-derived macrophages (Mo-MФs) polarization in vitro. Mo-MФs were generated from 28 patients with moderate to severe MDD and 28 age-, sex-, smoking status- and BMI-matched healthy controls (HC). Cells were treated either with AS or fetal calf serum (FCS) and polarized into M1 (LPS), M2 (IL-10, IL-4, TGF-β) or M0 (unstimulated) macrophages. Polarization capacity was quantified by means of specific M1 (CCR7, CD86, CXCL10, IL-12p70, TNF-α, IL-6, IL-1β, IL-12p40, IL-23, IP-10) and M2 (CD206, IL-10, TARC, IL-1RA) markers. Compared to HC, significantly increased M1-polarization was observed for MDD patients in the presence of FCS, however, polarization in AS enriched media determined an increased M2-polarization in patients. Moreover, female MDD patients exhibited increased M1- and decreased M2-polarization in both conditions compared to male MDD patients. Our data suggests that Mo-MФs derived from patients with MDD exhibit facilitated M1-polarization under traditional cell culture conditions and an increased potential for M2-polarization when cultured in AS. Striking inter-individual variation and pronounced gender effects highlight the potential utility of our personalized cell model-based approach to aid diagnostic and therapeutic decisions.
Collapse
|
62
|
Carboni E, Carta AR, Carboni E, Novelli A. Repurposing Ketamine in Depression and Related Disorders: Can This Enigmatic Drug Achieve Success? Front Neurosci 2021; 15:657714. [PMID: 33994933 PMCID: PMC8120160 DOI: 10.3389/fnins.2021.657714] [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] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 03/31/2021] [Indexed: 12/27/2022] Open
Abstract
Repurposing ketamine in the therapy of depression could well represent a breakthrough in understanding the etiology of depression. Ketamine was originally used as an anesthetic drug and later its use was extended to other therapeutic applications such as analgesia and the treatment of addiction. At the same time, the abuse of ketamine as a recreational drug has generated a concern for its psychotropic and potential long-term effects; nevertheless, its use as a fast acting antidepressant in treatment-resistant patients has boosted the interest in the mechanism of action both in psychiatry and in the wider area of neuroscience. This article provides a comprehensive overview of the actions of ketamine and intends to cover: (i) the evaluation of its clinical use in the treatment of depression and suicidal behavior; (ii) the potential use of ketamine in pediatrics; (iii) a description of its mechanism of action; (iv) the involvement of specific brain areas in producing antidepressant effects; (v) the potential interaction of ketamine with the hypothalamic-pituitary-adrenal axis; (vi) the effect of ketamine on neuronal transmission in the bed nucleus of stria terminalis and on its output; (vii) the evaluation of any gender-dependent effects of ketamine; (viii) the interaction of ketamine with the inflammatory processes involved in depression; (ix) the evaluation of the effects observed with single or repeated administration; (x) a description of any adverse or cognitive effects and its abuse potential. Finally, this review attempts to assess whether ketamine's use in depression can improve our knowledge of the etiopathology of depression and whether its therapeutic effect can be considered an actual cure for depression rather than a therapy merely aimed to control the symptoms of depression.
Collapse
Affiliation(s)
- Ezio Carboni
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Anna R. Carta
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Elena Carboni
- Unit of Paediatrics, ASST Cremona Maggiore Hospital, Cremona, Italy
| | - Antonello Novelli
- Department of Psychology and University Institute of Biotechnology of Asturias, University of Oviedo, Oviedo, Spain
- Sanitary Institute of the Princedom of Asturias, Oviedo, Spain
| |
Collapse
|
63
|
Valiuliene G, Valiulis V, Dapsys K, Vitkeviciene A, Gerulskis G, Navakauskiene R, Germanavicius A. Brain stimulation effects on serum BDNF, VEGF, and TNFα in treatment-resistant psychiatric disorders. Eur J Neurosci 2021; 53:3791-3802. [PMID: 33861484 DOI: 10.1111/ejn.15232] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 04/01/2021] [Accepted: 04/08/2021] [Indexed: 02/06/2023]
Abstract
Resistance to pharmacological treatment poses a notable challenge for psychiatry. Such cases are usually treated with brain stimulation techniques, including repetitive transcranial magnetic stimulation (rTMS) and electroconvulsive therapy (ECT). Empirical evidence links treatment resistance to insufficient brain plasticity and chronic inflammation. Therefore, this study encompasses analysis of neurotrophic and inflammatory factors in psychiatric patients undergoing rTMS and ECT in order to refine the selection of patients and predict clinical outcomes. This study enrolled 25 drug-resistant depressive patients undergoing rTMS and 31 drug-resistant schizophrenia patients undergoing ECT. Clinical efficacy of brain stimulation therapies was gauged using MADRS and HAM-D scales in the depression group and PANSS scale in the schizophrenia group. Blood-derived BDNF, VEGF, and TNFα were analysed during the treatment course. For reference, 19 healthy control subjects were also enrolled. After statistical analysis, no significant differences were detected in BDNF, VEGF, and TNFα concentrations among healthy, depressive, and schizophrenic subject groups before the treatment. However, depressive patient treatment with rTMS has increased BDNF concentration, while schizophrenic patient treatment with ECT has lowered the concentration of TNFα. Our findings suggest that a lower initial TNFα concentration could be a marker for treatment success in depressed patients undergoing rTMS, whereas in schizophrenic patient group treated with ECT, a higher concentration of VEGF correlates to milder symptoms post-treatment, especially in the negative scale.
Collapse
Affiliation(s)
- Giedre Valiuliene
- Life Sciences Center, Institute of Biochemistry, Vilnius University, Vilnius, Lithuania
| | - Vladas Valiulis
- Life Sciences Center, Institute of Biochemistry, Vilnius University, Vilnius, Lithuania.,Republican Vilnius Psychiatric Hospital, Vilnius, Lithuania
| | - Kastytis Dapsys
- Life Sciences Center, Institute of Biochemistry, Vilnius University, Vilnius, Lithuania.,Republican Vilnius Psychiatric Hospital, Vilnius, Lithuania.,Life Sciences Center, Institute of Biosciences, Vilnius University, Vilnius, Lithuania
| | - Aida Vitkeviciene
- Life Sciences Center, Institute of Biochemistry, Vilnius University, Vilnius, Lithuania
| | - Giedrius Gerulskis
- Life Sciences Center, Institute of Biochemistry, Vilnius University, Vilnius, Lithuania.,Republican Vilnius Psychiatric Hospital, Vilnius, Lithuania
| | - Ruta Navakauskiene
- Life Sciences Center, Institute of Biochemistry, Vilnius University, Vilnius, Lithuania
| | - Arunas Germanavicius
- Life Sciences Center, Institute of Biochemistry, Vilnius University, Vilnius, Lithuania.,Republican Vilnius Psychiatric Hospital, Vilnius, Lithuania.,Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
64
|
Nettis MA, Lombardo G, Hastings C, Zajkowska Z, Mariani N, Nikkheslat N, Worrell C, Enache D, McLaughlin A, Kose M, Sforzini L, Bogdanova A, Cleare A, Young AH, Pariante CM, Mondelli V. Augmentation therapy with minocycline in treatment-resistant depression patients with low-grade peripheral inflammation: results from a double-blind randomised clinical trial. Neuropsychopharmacology 2021; 46:939-948. [PMID: 33504955 PMCID: PMC8096832 DOI: 10.1038/s41386-020-00948-6] [Citation(s) in RCA: 122] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 11/27/2020] [Accepted: 12/14/2020] [Indexed: 01/30/2023]
Abstract
This study aimed to investigate the role of baseline levels of peripheral inflammation when testing the efficacy of antidepressant augmentation with minocycline in patients with treatment-resistant depression. We conducted a 4-week, placebo-controlled, randomised clinical trial of minocycline (200 mg/day) added to antidepressant treatment in 39 patients selected for elevated levels of serum C-reactive protein (CRP ≥ 1 mg/L), n = 18 randomised to minocycline (M) and n = 21 to placebo (P). The main outcome was the change in Hamilton Depression Rating Scale (HAM-D-17) score from baseline to week 4, expressed both as mean and as full or partial response, in the overall sample and after further stratification for baseline CRP≥3 mg/L. Secondary outcomes included changes in other clinical and inflammatory measures. Changes in HAM-D-17 scores and the proportion of partial responders did not differ between study arms. After stratification for CRP levels <3 mg/L (CRP-) or ≥3 mg/L (CRP+), CRP+/M patients showed the largest changes in HAM-D-17 scores (mean ± SD = 12.00 ± 6.45) compared with CRP-/M (2.42 ± 3.20, p < 0.001), CRP+/P (3.50 ± 4.34, p = 0.003) and CRP-/P (2.11 ± 3.26, p = 0.006) patients, and the largest proportion (83.3%, p = 0.04) of partial treatment response at week 4. The threshold point for baseline CRP to distinguish responders from non-responders to minocycline was 2.8 mg/L. Responders to minocycline had higher baseline IL-6 concentrations than non-responders (p = 0.03); IFNγ was significantly reduced after treatment with minocycline compared with placebo (p = 0.03). Our data show some evidence of efficacy of add-on treatment with minocycline in MDD patients but only in those with low-grade inflammation defined as CRP ≥3 mg/L.
Collapse
Affiliation(s)
- Maria Antonietta Nettis
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Psychological Medicine, London, UK
- National Institute for Health Research Mental Health Biomedical Research Centre, South London and Maudsley NHS Foundation Trust and King's College London, London, UK
| | - Giulia Lombardo
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Psychological Medicine, London, UK
| | - Caitlin Hastings
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Psychological Medicine, London, UK
| | - Zuzanna Zajkowska
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Psychological Medicine, London, UK
| | - Nicole Mariani
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Psychological Medicine, London, UK
| | - Naghmeh Nikkheslat
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Psychological Medicine, London, UK
| | - Courtney Worrell
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Psychological Medicine, London, UK
| | - Daniela Enache
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Psychological Medicine, London, UK
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden
| | - Anna McLaughlin
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Psychological Medicine, London, UK
| | - Melisa Kose
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Psychological Medicine, London, UK
| | - Luca Sforzini
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Psychological Medicine, London, UK
| | - Anna Bogdanova
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Psychological Medicine, London, UK
| | - Anthony Cleare
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Psychological Medicine, London, UK
- National Institute for Health Research Mental Health Biomedical Research Centre, South London and Maudsley NHS Foundation Trust and King's College London, London, UK
| | - Allan H Young
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Psychological Medicine, London, UK
- National Institute for Health Research Mental Health Biomedical Research Centre, South London and Maudsley NHS Foundation Trust and King's College London, London, UK
| | - Carmine M Pariante
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Psychological Medicine, London, UK
- National Institute for Health Research Mental Health Biomedical Research Centre, South London and Maudsley NHS Foundation Trust and King's College London, London, UK
| | - Valeria Mondelli
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Psychological Medicine, London, UK.
- National Institute for Health Research Mental Health Biomedical Research Centre, South London and Maudsley NHS Foundation Trust and King's College London, London, UK.
| |
Collapse
|
65
|
Kruse JL, Vasavada MM, Olmstead R, Hellemann G, Wade B, Breen EC, Brooks JO, Congdon E, Espinoza R, Narr KL, Irwin MR. Depression treatment response to ketamine: sex-specific role of interleukin-8, but not other inflammatory markers. Transl Psychiatry 2021; 11:167. [PMID: 33723220 PMCID: PMC7960960 DOI: 10.1038/s41398-021-01268-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 01/08/2021] [Accepted: 01/20/2021] [Indexed: 12/11/2022] Open
Abstract
Inflammation plays a role in depression pathophysiology and treatment response, with effects varying by sex and therapeutic modality. Lower levels of interleukin(IL)-8 predict depression response to antidepressant medication and to electroconvulsive therapy (ECT), although ECT effects are specific to females. Whether IL-8 predicts depression response to ketamine and in a sex-specific manner is not known. Here, depressed patients (n = 46; female, n = 17) received open label infusion of ketamine (0.5 mg/kg over 40 min; NCT02165449). Plasma levels of IL-8 were evaluated at baseline and post-treatment. Baseline levels of IL-8 had a trending association with response to ketamine, depending upon sex (responder status × sex interaction: p = 0.096), in which lower baseline levels of IL-8 in females (p = 0.095) but not males (p = 0.96) trended with treatment response. Change in levels of IL-8 from baseline to post-treatment differed significantly by responder status (defined as ≥50% reduction in Hamilton Depression Rating Scale [HAM-D] Score), depending upon sex (responder status × sex × time interaction: F(1,42)=6.68, p = 0.01). In addition, change in IL-8 interacted with sex to predict change in HAM-D score (β = -0.63, p = 0.003); increasing IL-8 was associated with decreasing HAM-D score in females (p = 0.08) whereas the inverse was found in males (p = 0.02). Other inflammatory markers (IL-6, IL-10, tumor necrosis factor-α, C-reactive protein) were explored with no significant relationships identified. Given these preliminary findings, further evaluation of sex differences in the relationship between IL-8 and treatment response is warranted to elucidate mechanisms of response and aid in the development of personalized approaches to depression treatment.
Collapse
Affiliation(s)
- Jennifer L. Kruse
- grid.19006.3e0000 0000 9632 6718Cousins Center for Psychoneuroimmunology, University of California at Los Angeles, Los Angeles, CA USA ,grid.19006.3e0000 0000 9632 6718Jane and Terry Semel Institute for Neuroscience and Human Behavior at UCLA, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, Los Angeles, CA USA
| | - Megha M. Vasavada
- grid.19006.3e0000 0000 9632 6718Department of Neurology, University of California at Los Angeles, Los Angeles, CA USA
| | - Richard Olmstead
- grid.19006.3e0000 0000 9632 6718Cousins Center for Psychoneuroimmunology, University of California at Los Angeles, Los Angeles, CA USA ,grid.19006.3e0000 0000 9632 6718Jane and Terry Semel Institute for Neuroscience and Human Behavior at UCLA, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, Los Angeles, CA USA
| | - Gerhard Hellemann
- grid.19006.3e0000 0000 9632 6718Jane and Terry Semel Institute for Neuroscience and Human Behavior at UCLA, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, Los Angeles, CA USA
| | - Benjamin Wade
- grid.19006.3e0000 0000 9632 6718Department of Neurology, University of California at Los Angeles, Los Angeles, CA USA
| | - Elizabeth C. Breen
- grid.19006.3e0000 0000 9632 6718Cousins Center for Psychoneuroimmunology, University of California at Los Angeles, Los Angeles, CA USA ,grid.19006.3e0000 0000 9632 6718Jane and Terry Semel Institute for Neuroscience and Human Behavior at UCLA, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, Los Angeles, CA USA
| | - John O. Brooks
- grid.19006.3e0000 0000 9632 6718Jane and Terry Semel Institute for Neuroscience and Human Behavior at UCLA, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, Los Angeles, CA USA
| | - Eliza Congdon
- grid.19006.3e0000 0000 9632 6718Jane and Terry Semel Institute for Neuroscience and Human Behavior at UCLA, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, Los Angeles, CA USA
| | - Randall Espinoza
- grid.19006.3e0000 0000 9632 6718Jane and Terry Semel Institute for Neuroscience and Human Behavior at UCLA, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, Los Angeles, CA USA
| | - Katherine L. Narr
- grid.19006.3e0000 0000 9632 6718Jane and Terry Semel Institute for Neuroscience and Human Behavior at UCLA, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, Los Angeles, CA USA ,grid.19006.3e0000 0000 9632 6718Department of Neurology, University of California at Los Angeles, Los Angeles, CA USA
| | - Michael R. Irwin
- grid.19006.3e0000 0000 9632 6718Cousins Center for Psychoneuroimmunology, University of California at Los Angeles, Los Angeles, CA USA ,grid.19006.3e0000 0000 9632 6718Jane and Terry Semel Institute for Neuroscience and Human Behavior at UCLA, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, Los Angeles, CA USA
| |
Collapse
|
66
|
Lebowitz ER, Orbach M, Marin CE, Salmaso N, Vaccarino FM, Silverman WK. Fibroblast Growth Factor 2 Implicated in Childhood Anxiety and Depression Symptoms. J Affect Disord 2021; 282:611-616. [PMID: 33445083 PMCID: PMC7897422 DOI: 10.1016/j.jad.2020.12.055] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/14/2020] [Accepted: 12/17/2020] [Indexed: 01/18/2023]
Abstract
BACKGROUND Research links fibroblast growth factor 2 (FGF2) to anxiety and depression in rodents and human adults. Our study is the first to examine FGF2 levels in a pediatric population. METHODS We assayed serum FGF2 in 163 children with a broad range of anxiety and depressive symptoms; 111 were clinic-referred anxious and depressed children; 52 were non-referred children. We examined associations between FGF2 and anxiety and depression symptoms, and between each of the three facets of behavioral activation (Reward-Responsiveness, Drive, Fun-Seeking) and behavioral avoidance. We used confirmatory factor analysis (CFA) to determine the relative contribution of anxiety and depression indicators and of FGF2 to a latent variable of Anxiety/Depression. We also examined stability of FGF2 levels. RESULTS FGF2 levels in clinic-referred children were significantly lower compared with non-referred children. Bivariate correlations and CFA showed negative associations between FGF2 and anxiety, depression and behavioral avoidance. FGF2 levels were positively correlated with the Reward-Responsiveness facet of behavioral activation, implicated in depression. FGF2 levels were stable over six months. LIMITATIONS We did not have data on behavioral avoidance and stability of FGF2 in the entire sample. CONCLUSIONS Our results implicate FGF2 in anxiety and depression in children, providing an important first step in showing FGF2 may serve as a stable biomarker for these prevalent and impairing problems.
Collapse
Affiliation(s)
- Eli R. Lebowitz
- Child Study Center, Yale School of Medicine, New Haven, CT, 06510, USA,Anxiety and Mood Disorders Program, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Meital Orbach
- Child Study Center, Yale School of Medicine, New Haven, CT, 06510, USA,Anxiety and Mood Disorders Program, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Carla E. Marin
- Child Study Center, Yale School of Medicine, New Haven, CT, 06510, USA,Anxiety and Mood Disorders Program, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Natalina Salmaso
- Department of Neuroscience, Carleton University, Ottawa, Ontario, K1S 5B6, Canada
| | - Flora M. Vaccarino
- Child Study Center, Yale School of Medicine, New Haven, CT, 06510, USA,Department of Neuroscience, Yale School of Medicine, New Haven, CT, 06510, USA,Program in Neurodevelopment and Regeneration, Yale University, New Haven, CT, 06510, USA
| | - Wendy K. Silverman
- Child Study Center, Yale School of Medicine, New Haven, CT, 06510, USA,Anxiety and Mood Disorders Program, Yale School of Medicine, New Haven, CT, 06510, USA,Department of Psychology, Yale University, New Haven, CT, 06520, USA
| |
Collapse
|
67
|
Dong Z, Kuang W, Shen X, Tian L. Plasma levels of interleukin-6 and antidepressant response to Paroxetine in Chinese depressive patients. Psychiatry Res 2021; 297:113723. [PMID: 33545432 DOI: 10.1016/j.psychres.2021.113723] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 01/11/2021] [Indexed: 02/08/2023]
Abstract
This study aimed to investigate the correlation between interleukin-6 (IL-6) plasma levels and treatment outcomes of selective serotonin reuptake inhibitors in patients with major depressive disorder (MDD). A total of 104 patients (33 males and 71 females), aged 18 to 72 years, were enrolled. Peripheral blood samples were collected before treatment initiation (baseline) and eight weeks after oral paroxetine treatment. The Hamilton Depression Rating Scale (HAMD)-17 was used to evaluate the efficacy of paroxetine. Baseline plasma IL-6 levels were found to be significantly lower in patients who responded to treatment than in non-responders. A negative correlation was found between the HAMD-17 reduction rate and baseline IL-6 levels. Furthermore, associations were examined between HAMD-17 reduction rate in patients and other factors, such as IL-6 levels, sex, age, and body mass index. Baseline IL-6 was the only factor showing a significant impact on the reduction rate of HAMD-17 at week 8. These results suggest that plasma IL-6 level may be a promising biological marker for predicting the likely treatment response to paroxetine in individual patients with MDD.
Collapse
Affiliation(s)
- Zaiquan Dong
- Department of Mental Health Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Weihong Kuang
- Department of Mental Health Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaoling Shen
- Department of Mental Health Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Liantian Tian
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
68
|
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
|
69
|
Guo ZP, Sörös P, Zhang ZQ, Yang MH, Liao D, Liu CH. Use of Transcutaneous Auricular Vagus Nerve Stimulation as an Adjuvant Therapy for the Depressive Symptoms of COVID-19: A Literature Review. Front Psychiatry 2021; 12:765106. [PMID: 34975571 PMCID: PMC8714783 DOI: 10.3389/fpsyt.2021.765106] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 11/12/2021] [Indexed: 12/17/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) comprises more than just severe acute respiratory syndrome. It also interacts with the cardiovascular, nervous, renal, and immune systems at multiple levels, increasing morbidity in patients with underlying cardiometabolic conditions and inducing myocardial injury or dysfunction. Transcutaneous auricular vagus nerve stimulation (taVNS), which is derived from auricular acupuncture, has become a popular therapy that is increasingly accessible to the general public in modern China. Here, we begin by outlining the historical background of taVNS, and then describe important links between dysfunction in proinflammatory cytokine release and related multiorgan damage in COVID-19. Furthermore, we emphasize the important relationships between proinflammatory cytokines and depressive symptoms. Finally, we discuss how taVNS improves immune function via the cholinergic anti-inflammatory pathway and modulates brain circuits via the hypothalamic-pituitary-adrenal axis, making taVNS an important treatment for depressive symptoms on post-COVID-19 sequelae. Our review suggests that the link between anti-inflammatory processes and brain circuits could be a potential target for treating COVID-19-related multiorgan damage, as well as depressive symptoms using taVNS.
Collapse
Affiliation(s)
- Zhi-Peng Guo
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Peter Sörös
- Research Center Neurosensory Science, Carl von Ossietzky University of Oldenburg, Oldenburg, Germany
| | - Zhu-Qing Zhang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Ming-Hao Yang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Dan Liao
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Chun-Hong Liu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Acupuncture Neuromodulation, Beijing Institute of Traditional Chinese Medicine, Beijing, China
| |
Collapse
|
70
|
Lasselin J, Lekander M, Benson S, Schedlowski M, Engler H. Sick for science: experimental endotoxemia as a translational tool to develop and test new therapies for inflammation-associated depression. Mol Psychiatry 2021; 26:3672-3683. [PMID: 32873895 PMCID: PMC8550942 DOI: 10.1038/s41380-020-00869-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 07/25/2020] [Accepted: 08/06/2020] [Indexed: 12/16/2022]
Abstract
Depression is one of the global leading causes of disability, but treatments remain limited and classical antidepressants were found to be ineffective in a substantial proportion of patients. Thus, novel effective therapies for the treatment of depression are urgently needed. Given the emerging role of inflammation in the etiology and pathophysiology of affective disorders, we herein illustrate how experimental endotoxemia, a translational model of systemic inflammation, could be used as a tool to develop and test new therapeutic options against depression. Our concept is based on the striking overlap of inflammatory, neural, and affective characteristics in patients with inflammation-associated depression and in endotoxin-challenged healthy subjects. Experimental administration of endotoxin in healthy volunteers is safe, well-tolerated, and without known long-term health risks. It offers a highly standardized translational approach to characterize potential targets of therapies against inflammation-associated depression, as well as to identify characteristics of patients that would benefit from these interventions, and, therefore, could contribute to improve personalization of treatment and to increase the overall rate of responders.
Collapse
Affiliation(s)
- Julie Lasselin
- Institute of Medical Psychology and Behavioral Immunobiology, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122, Essen, Germany. .,Stress Research Institute, Stockholm University, 10691, Stockholm, Sweden. .,Department of Clinical Neuroscience, Division for Psychology, Karolinska Institutet, Nobels väg 9, 17177, Stockholm, Sweden. .,Osher Center for Integrative Medicine, ME Neuroradiologi, Karolinska Universitetssjukhuset, Stockholm, Sweden.
| | - Mats Lekander
- grid.10548.380000 0004 1936 9377Stress Research Institute, Stockholm University, 10691 Stockholm, Sweden ,grid.4714.60000 0004 1937 0626Department of Clinical Neuroscience, Division for Psychology, Karolinska Institutet, Nobels väg 9, 17177 Stockholm, Sweden ,grid.24381.3c0000 0000 9241 5705Osher Center for Integrative Medicine, ME Neuroradiologi, Karolinska Universitetssjukhuset, Stockholm, Sweden
| | - Sven Benson
- grid.5718.b0000 0001 2187 5445Institute of Medical Psychology and Behavioral Immunobiology, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122 Essen, Germany
| | - Manfred Schedlowski
- grid.5718.b0000 0001 2187 5445Institute of Medical Psychology and Behavioral Immunobiology, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122 Essen, Germany ,grid.4714.60000 0004 1937 0626Department of Clinical Neuroscience, Division for Psychology, Karolinska Institutet, Nobels väg 9, 17177 Stockholm, Sweden
| | - Harald Engler
- grid.5718.b0000 0001 2187 5445Institute of Medical Psychology and Behavioral Immunobiology, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45122 Essen, Germany
| |
Collapse
|
71
|
Fox HC, Milivojevic V, MacDougall A, LaVallee H, Simpson C, Angarita GA, Sinha R. Stress-related suppression of peripheral cytokines predicts future relapse in alcohol-dependent individuals with and without subclinical depression. Addict Biol 2020; 25:e12832. [PMID: 31736187 DOI: 10.1111/adb.12832] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 07/22/2019] [Accepted: 08/28/2019] [Indexed: 12/01/2022]
Abstract
Chronic alcohol abuse and depressive symptoms are both associated with peripheral cytokine changes. Despite this, cytokine adaptations have not been assessed in co-morbid populations or prospectively as predictors of relapse. We examine cytokine responses to stress in alcohol-dependent individuals and social drinkers, both with and without subclinical depression. We also examine the potential link between cytokine adaptations in response to stress and prospective alcohol relapse risk. Thirty-three, alcohol-dependent individuals (21 with and 12 without high depressive symptoms) and 37 controls (16 with and 21 without high depressive symptoms) were exposed to two 5-minute personalized guided imagery conditions (stress and neutral) across consecutive days in a randomized and counterbalanced order. Alcohol craving and serum measures of tumor necrosis factor alpha (TNFα), tumor necrosis factor receptor 1 (TNFR1), interleukin-6 (IL-6), and interleukin-1 receptor antagonist (IL-1ra) were collected prior to and following imagery exposure. Following treatment discharge, follow-up interviews were conducted over 90 days to assess relapse. Dampened IL-1ra and IL-6 in response to stress was observed as a function of alcohol dependence and not moderated by depressive symptoms. Lower levels of IL-6 following stress also predicted greater drinking days following treatment. Conversely, high depressive symptomatology was associated solely with pro-inflammatory adaptations. Stress-related suppression of TNFα predicted drinking severity only in alcohol-dependent individuals with subclinical depression, and suppressed TNFR1 following stress was only seen in individuals with subclinical depression. Stress-induced suppression of pro-inflammatory TNF markers may indicate a risk factor for alcohol-dependent individuals with co-occurring depressive symptoms.
Collapse
Affiliation(s)
- Helen C. Fox
- Department of Psychiatry, School of Medicine Stony Brook University Stony Brook NY USA
| | - Verica Milivojevic
- The Yale Stress Center, Department of Psychiatry Yale University School of Medicine New Haven CT USA
| | - Alicia MacDougall
- The Yale Stress Center, Department of Psychiatry Yale University School of Medicine New Haven CT USA
| | - Heather LaVallee
- The Yale Stress Center, Department of Psychiatry Yale University School of Medicine New Haven CT USA
| | - Christine Simpson
- Department of Internal Medicine Yale University School of Medicine New Haven CT USA
| | - Gustavo A. Angarita
- Clinical Neuroscience Research Unit, The Connecticut Mental Health Center, Department of Psychiatry Yale University School of Medicine New Haven CT USA
| | - Rajita Sinha
- The Yale Stress Center, Department of Psychiatry Yale University School of Medicine New Haven CT USA
| |
Collapse
|
72
|
Zhou YL, Wu FC, Wang CY, Zheng W, Lan XF, Deng XR, Ning YP. Relationship between hippocampal volume and inflammatory markers following six infusions of ketamine in major depressive disorder. J Affect Disord 2020; 276:608-615. [PMID: 32871692 DOI: 10.1016/j.jad.2020.06.068] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 05/22/2020] [Accepted: 06/23/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Evidences suggest that inflammatory marker-mediated neuroplasticity contributes critically to brain changes following antidepressant treatment. To date, no study has examined the relationship between changes in hippocampal volume, depressive symptoms, and inflammatory markers following repeated ketamine treatment. METHODS Forty-four patients with major depressive disorder received six intravenous ketamine (0.5 mg/kg) infusions over 12 days. The Montgomery-Asberg Depression Rating Scale (MADRS) was used to assess depressive symptoms, and peripheral blood was collected to test multiple cytokines and tryptophan (TRP) metabolites at baseline, 24 h and 14 days after the sixth infusion (day 13 and day 26). Magnetic resonance imaging (MRI) scans were carried out at baseline and day13, and FreeSurfer software was used to process the T1 images and analyze hippocampal volume. RESULTS Following ketamine, a significant improvement in depressive symptoms, a small increase in right hippocampal volume and alterations in inflammatory markers was found. No significant association was found between changes in inflammatory markers and changes in hippocampal volume from baseline to day 13 (P>0.05), while a weak association was found between TRP metabolite changes and other cytokine changes from baseline to day 26 (beta=-0.357, t=-2.600, P = 0.013). LIMITATIONS The patients continued receiving previous medications during ketamine treatment, which may have impacted hippocampal volume and inflammatory markers. CONCLUSIONS Hippocampal volume increase following ketamine was an independent neurobiological effect that was not associated with changes in peripheral inflammatory markers, suggesting a likely complex neurobiological mechanism of the antidepressant effect of ketamine.
Collapse
Affiliation(s)
- Yan-Ling Zhou
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Feng-Chun Wu
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Cheng-Yu Wang
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Wei Zheng
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Xiao-Feng Lan
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Xiu-Rong Deng
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Yu-Ping Ning
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China; The First School of Clinical Medicine, Southern Medical University, Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China.
| |
Collapse
|
73
|
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
|
74
|
Fritz M, Klawonn AM, Zhao Q, Sullivan EV, Zahr NM, Pfefferbaum A. Structural and biochemical imaging reveals systemic LPS-induced changes in the rat brain. J Neuroimmunol 2020; 348:577367. [PMID: 32866714 DOI: 10.1016/j.jneuroim.2020.577367] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/18/2020] [Accepted: 08/19/2020] [Indexed: 12/11/2022]
Abstract
Despite mounting evidence for the role of inflammation in Major Depressive Disorder (MDD), in vivo preclinical investigations of inflammation-induced negative affect using whole brain imaging modalities are scarce, precluding a valid model within which to evaluate pharmacological interventions. Here we used an E. coli lipopolysaccharide (LPS)-based model of inflammation-induced depressive signs in rats to explore brain changes using multimodal neuroimaging methods. During the acute phase of the LPS response (2 h post injection), prior to the emergence of a task-quantifiable depressive phenotype, striatal glutamine levels and splenial, retrosplenial, and peri-callosal hippocampal cortex volumes were greater than at baseline. LPS-induced depressive behaviors observed at 24 h, however, occurred concurrently with lower than control levels of striatal glutamine and a reversibility of volume expansion (i.e., shrinkage of splenial, retrosplenial, and peri-callosal hippocampal cortex to baseline volumes). In both striatum and hippocampus at 24 h, mRNA expression in LPS relative to control animals demonstrated alterations in enzymes and transporters regulating glutamine homeostasis. Collectively, the observed behavioral, in vivo structural and metabolic, and mRNA expression alterations suggest a critical role for astrocytic regulation of inflammation-induced depressive behaviors.
Collapse
Affiliation(s)
- Michael Fritz
- Department of Psychiatry and Behavioral Sciences, Stanford School of Medicine, Stanford University, Stanford, CA 94304, United States of America
| | - Anna M Klawonn
- Department of Psychiatry and Behavioral Sciences, Stanford School of Medicine, Stanford University, Stanford, CA 94304, United States of America
| | - Qingyu Zhao
- Department of Psychiatry and Behavioral Sciences, Stanford School of Medicine, Stanford University, Stanford, CA 94304, United States of America
| | - Edith V Sullivan
- Department of Psychiatry and Behavioral Sciences, Stanford School of Medicine, Stanford University, Stanford, CA 94304, United States of America; Neuroscience Program, SRI International, Menlo Park, CA 94025, United States of America
| | - Natalie M Zahr
- Department of Psychiatry and Behavioral Sciences, Stanford School of Medicine, Stanford University, Stanford, CA 94304, United States of America; Neuroscience Program, SRI International, Menlo Park, CA 94025, United States of America.
| | - Adolf Pfefferbaum
- Department of Psychiatry and Behavioral Sciences, Stanford School of Medicine, Stanford University, Stanford, CA 94304, United States of America; Neuroscience Program, SRI International, Menlo Park, CA 94025, United States of America
| |
Collapse
|
75
|
Martinez-Muniz GA, Wood SK. Sex Differences in the Inflammatory Consequences of Stress: Implications for Pharmacotherapy. J Pharmacol Exp Ther 2020; 375:161-174. [PMID: 32759370 DOI: 10.1124/jpet.120.266205] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 07/28/2020] [Indexed: 12/21/2022] Open
Abstract
Women are at significantly greater risk of developing stress-related disorders such as depression. The increased risk begins during puberty and continues throughout life until menopause, suggesting a role for ovarian hormones in this increased susceptibility. Importantly, inflammation has been gaining momentum in its role in the pathogenesis of depression. Herein, clinical and preclinical studies have been reviewed to better understand how sex differences within the immune system may contribute to exaggerated risk of depression in females. First, studies that investigate the ability of psychologic stress episodes to engage the inflammatory systems both in the brain and periphery are reviewed with a special focus on sex-specific effects. Moreover, studies are discussed that identify whether imbalanced inflammatory milieu contributes to the development of depression in males versus females and whether these effects are regulated by estradiol. Importantly, we propose a locus coeruleus-norepinephrine-cytokine circuit as a conduit through which stress could increase stress susceptibly in females. Finally, the anti-inflammatory capacity of traditional and nontraditional antidepressants is investigated, with the goal of providing a better understanding of pharmacotherapeutics to enhance strategies to personalize antidepressant treatments between the sexes. The studies reviewed herein strongly support the need for further studies to elucidate whether females are especially sensitive to anti-inflammatory compounds as adjuvants to traditional therapies. SIGNIFICANCE STATEMENT: Women have hve an increased risk of developing stress-related disorders such as depression. In this review, literature from clinical and preclinical studies are integrated to define sex differences in stress-induced inflammatory responses as a potential source for the etiology of sex differences in depressive disorders. Moreover, the anti-inflammatory capacity of traditional and nontraditional antidepressants is reviewed to inform on potential pharmacotherapeutic strategies to personalize antidepressant therapy in a sex-dependent manner.
Collapse
Affiliation(s)
- Gustavo A Martinez-Muniz
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, South Carolina and Dorn Veterans Administration Medical Center, Columbia, South Carolina
| | - Susan K Wood
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, South Carolina and Dorn Veterans Administration Medical Center, Columbia, South Carolina
| |
Collapse
|
76
|
Zhan Y, Zhou Y, Zheng W, Liu W, Wang C, Lan X, Deng X, Xu Y, Zhang B, Ning Y. Alterations of multiple peripheral inflammatory cytokine levels after repeated ketamine infusions in major depressive disorder. Transl Psychiatry 2020; 10:246. [PMID: 32699226 PMCID: PMC7376102 DOI: 10.1038/s41398-020-00933-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 05/06/2020] [Accepted: 05/14/2020] [Indexed: 12/21/2022] Open
Abstract
Increasing evidence has demonstrated that inflammatory cytokines play an important role in major depressive disorder (MDD) and are associated with treatment outcomes. Few studies have explored the trajectories of multiple inflammatory cytokines after repeated ketamine infusions in MDD. In this study, we conducted a secondary analysis to investigate the impact of ketamine on the modulation of the inflammatory pathway in depression and whether this pathway contributes to the antidepressant properties of ketamine. A total of 60 patients with depression received six ketamine infusions (0.5 mg/kg) during a 12-day period. The Montgomery-Asberg Scale (MADRS) was administered, and blood samples were collected at baseline and 24 h and 14 days after the sixth infusion (days 0, 13, and 26). Plasma levels of the 19 cytokines were measured using the Luminex assay. At baseline, inflammatory cytokines were associated with the severity of depression. The concentrations of pro- and anti-inflammatory factors, including granulocyte macrophage colony-stimulating factor (GM-CSF), fractalkine, interferon gamma (IFN-γ), interleukin (IL)-10, IL-12p70, IL-17A, IL-1β, IL-2, IL-4, IL-23, IL-5, IL-6, IL-7, and tumor necrosis factor alpha (TNF-α), were downregulated after repeated ketamine administration (all p < 0.05). In addition, alterations in the levels of IL-17A (r = -0.259, p = 0.046) and IL-6 (r = -0.262, p = 0.043) were correlated with symptom improvement. A lower level of interferon-inducible T cell alpha chemoattractant (ITAC) at baseline was predictive of ketamine treatment response on day 13 according to a stepwise linear regression analysis (β = -0.296, p = 0.040). Our results suggest that the inflammatory pathway may be involved in the antidepressant effects of ketamine, which may be conducive to future treatment strategy optimization.
Collapse
Affiliation(s)
- Yanni Zhan
- grid.410737.60000 0000 8653 1072The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China ,grid.284723.80000 0000 8877 7471The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong China ,Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Yanling Zhou
- grid.410737.60000 0000 8653 1072The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China ,Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Wei Zheng
- grid.410737.60000 0000 8653 1072The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China ,Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Weijian Liu
- grid.410737.60000 0000 8653 1072The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China ,grid.284723.80000 0000 8877 7471The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong China ,Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Chengyu Wang
- grid.410737.60000 0000 8653 1072The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China ,Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Xiaofeng Lan
- grid.410737.60000 0000 8653 1072The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China ,Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Xiurong Deng
- grid.410737.60000 0000 8653 1072The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China ,Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Yan Xu
- grid.410737.60000 0000 8653 1072The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China ,Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Bin Zhang
- grid.410737.60000 0000 8653 1072The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China ,Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Yuping Ning
- The Affiliated Brain Hospital of Guangzhou Medical University (Guangzhou Huiai Hospital), Guangzhou, China. .,The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China. .,Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China.
| |
Collapse
|
77
|
Zhang Y, Li L, Zhang J. Curcumin in antidepressant treatments: An overview of potential mechanisms, pre‐clinical/clinical trials and ongoing challenges. Basic Clin Pharmacol Toxicol 2020; 127:243-253. [DOI: 10.1111/bcpt.13455] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 06/08/2020] [Accepted: 06/10/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Yinfeng Zhang
- International Medical Center Beijing Friendship HospitalCapital Medical University Beijing China
| | - Li Li
- International Medical Center Beijing Friendship HospitalCapital Medical University Beijing China
| | - Jinfeng Zhang
- Key Laboratory of Molecular Medicine and Biotherapy School of Life Sciences Beijing Institute of Technology Beijing China
| |
Collapse
|
78
|
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]
|
79
|
Mattos Dos Santos R. Isolation, social stress, low socioeconomic status and its relationship to immune response in Covid-19 pandemic context. Brain Behav Immun Health 2020; 7:100103. [PMID: 32835298 PMCID: PMC7326413 DOI: 10.1016/j.bbih.2020.100103] [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: 06/01/2020] [Revised: 06/18/2020] [Accepted: 06/25/2020] [Indexed: 01/09/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) outbreak was first reported December 2019, in Wuhan, China, and has since spread worldwide. Social distancing or isolation measures were taken to mitigate the pandemic. Furthermore, stress and low socioeconomic status in humans confer increased vulnerability to morbidity and mortality, what can be biologically observed. This condition tends to remain during the Covid-19 pandemic. Social disruption stress (SDR) raises important questions regarding the functioning of the immune system, and the release of several stress hormones. A molecular pattern, conserved transcriptional response to adversity (CTRA), is thought to have evolved to defend against physical injury during periods of heightened risk. Chronic CTRA activation could leave an organism vulnerable to viral infections, leading to increased pro-inflammatory gene expression and a suppression of anti-viral gene expression. The activation of such transcriptional status is related to conditions of social stress through either hostile human contact, or increased predatory vulnerability due to separation from the social group and also low socioeconomic status. This review aims to point out questions for government officials, researchers and health professionals to better target their actions during a pandemic and encourage studies for a better understanding of these characteristics. The coronavirus disease 2019 (COVID-19) outbreak has spread worldwide. Community mitigation guidelines, such as social distancing were taken. Social disruption stress leads to immune response alterations, and stress hormones. CTRA activation may lead to vulnerable to viral infection and systemic inflammation. CTRA may be activated due to social isolation and socioeconomic status.
Collapse
Affiliation(s)
- Rodrigo Mattos Dos Santos
- Department of Tropical Diseases, Botucatu Medical School, São Paulo State University (UNESP - Univ Estadual Paulista), Infectious Diseases Laboratory - UNIPEX - FMB UNESP, Rua Dr. Walter Mauricio Correa s/n, São Paulo, Brazil
| |
Collapse
|
80
|
di Michele F. Why Vitamin D Status Might be Important for Brain Health and Mental Well-Being? Curr Pharm Des 2020; 26:2439-2441. [DOI: 10.2174/138161282621200520085710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Flavia di Michele
- Acute Psychiatric Unit, PTV Foundation, Policlinico Tor Vergata, Rome, Italy
| |
Collapse
|
81
|
di Michele F, Talamo A, Niolu C, Siracusano A. Vitamin D and N-Acetyl Cysteine Supplementation in Treatment-Resistant Depressive Disorder Patients: A General Review. Curr Pharm Des 2020; 26:2442-2459. [DOI: 10.2174/1381612826666200406090051] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 03/31/2020] [Indexed: 12/20/2022]
Abstract
:
Major Depressive Disorder (MDD) is often a lifetime disabling mental illness as individuals with
MDD might not benefit from standard-therapy, including both pharmacological and psychosocial interventions.
Novel therapies are, therefore, required.
:
It was shown by recent preclinical and clinical studies that the dysfunction of glutamatergic neurotransmission
might be involved in the pathophysiology of MDD. Furthermore, neuroimmune alterations could have a significant
role in the pathogenesis of MDD.
:
Vitamin D is a neurosteroid hormone essential for several metabolic processes, immune responses, and for regulating
neurotrophic-neuroprotective processes, neurotransmission and synaptic plasticity. Recent studies have also
shown Vitamin D deficiency in patients with severe psychiatric disorders, including MDD.
:
Lately, clinical studies have shown the neuroprotective action of N-acetyl cysteine (NAC) through the modulation
of inflammatory pathways and via the modulation of synaptic release of glutamate in cortico-subcortical
brain regions; the cysteine-glutamate antiporter.
:
This paper reviews the therapeutic use of Vitamin D and NAC and among individuals with refractory MDD to the
first- line pharmacological interventions, reviewing the clinical studies published in the last decade.
:
A detailed summary of the current evidence in this area aims to better inform psychiatrists and general practitioners
on the potential benefits of Vitamin D and NAC supplementation for this disorder.
:
Nutraceutical supplementation with Vitamin D and NAC in treatment-resistant MDD patients may be important
not only for improving depressive clinical manifestations but also for their safety and tolerability profile. This is
of great interest, especially considering the need for treating special populations affected by MDD, such as
youngsters and elders. Finally, the nutraceutical approach represents a good choice, considering its better compliance
by the patients compared to traditional psychopharmacological treatment.
Collapse
Affiliation(s)
- Flavia di Michele
- Acute Psychiatric Unit, PTV Foundation - Policlinico Tor Vergata, Rome, Italy
| | - Alessandra Talamo
- Acute Psychiatric Unit, PTV Foundation - Policlinico Tor Vergata, Rome, Italy
| | - Cinzia Niolu
- Acute Psychiatric Unit, PTV Foundation - Policlinico Tor Vergata, Rome, Italy
| | - Alberto Siracusano
- Acute Psychiatric Unit, PTV Foundation - Policlinico Tor Vergata, Rome, Italy
| |
Collapse
|
82
|
Neurobiological biomarkers of response to ketamine. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2020; 89:195-235. [PMID: 32616207 DOI: 10.1016/bs.apha.2020.05.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
As a field, psychiatry is undergoing an exciting paradigm shift toward early identification and intervention that will likely minimize both the burden associated with severe mental illnesses as well as their duration. In this context, the rapid-acting antidepressant ketamine has revolutionized our understanding of antidepressant response and greatly expanded the pharmacologic armamentarium for treatment-resistant depression. Efforts to characterize biomarkers of ketamine response support a growing emphasis on early identification, which would allow clinicians to identify biologically enriched subgroups with treatment-resistant depression who are more likely to benefit from ketamine therapy. This chapter presents a broad overview of a range of translational biomarkers, including those drawn from imaging and electrophysiological studies, sleep and circadian rhythms, and HPA axis/endocrine function as well as metabolic, immune, (epi)genetic, and neurotrophic biomarkers related to ketamine response. Ketamine's unique, rapid-acting properties may serve as a model to explore a whole new class of novel rapid-acting treatments with the potential to revolutionize drug development and discovery. However, it should be noted that although several of the biomarkers reviewed here provide promising insights into ketamine's mechanism of action, most studies have focused on acute rather than longer-term antidepressant effects and, at present, none of the biomarkers are ready for clinical use.
Collapse
|
83
|
The Bidirectional Relationship of Depression and Inflammation: Double Trouble. Neuron 2020; 107:234-256. [PMID: 32553197 DOI: 10.1016/j.neuron.2020.06.002] [Citation(s) in RCA: 932] [Impact Index Per Article: 233.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 04/21/2020] [Accepted: 05/29/2020] [Indexed: 12/12/2022]
Abstract
Depression represents the number one cause of disability worldwide and is often fatal. Inflammatory processes have been implicated in the pathophysiology of depression. It is now well established that dysregulation of both the innate and adaptive immune systems occur in depressed patients and hinder favorable prognosis, including antidepressant responses. In this review, we describe how the immune system regulates mood and the potential causes of the dysregulated inflammatory responses in depressed patients. However, the proportion of never-treated major depressive disorder (MDD) patients who exhibit inflammation remains to be clarified, as the heterogeneity in inflammation findings may stem in part from examining MDD patients with varied interventions. Inflammation is likely a critical disease modifier, promoting susceptibility to depression. Controlling inflammation might provide an overall therapeutic benefit, regardless of whether it is secondary to early life trauma, a more acute stress response, microbiome alterations, a genetic diathesis, or a combination of these and other factors.
Collapse
|
84
|
Kim J, Yoon S, Lee S, Hong H, Ha E, Joo Y, Lee EH, Lyoo IK. A double-hit of stress and low-grade inflammation on functional brain network mediates posttraumatic stress symptoms. Nat Commun 2020; 11:1898. [PMID: 32313055 PMCID: PMC7171097 DOI: 10.1038/s41467-020-15655-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 03/18/2020] [Indexed: 12/12/2022] Open
Abstract
Growing evidence indicates a reciprocal relationship between low-grade systemic inflammation and stress exposure towards increased vulnerability to neuropsychiatric disorders, including posttraumatic stress disorder (PTSD). However, the neural correlates of this reciprocity and their influence on the subsequent development of PTSD are largely unknown. Here we investigated alterations in functional connectivity among brain networks related to low-grade inflammation and stress exposure using two large independent data sets. Functional couplings among the higher-order cognitive network system including the salience, default mode, and central executive networks were reduced in association with low-grade inflammation and stress exposure. This reduced functional coupling may also be related to subsequent posttraumatic stress symptom severity. The current findings propose functional couplings among the higher-order cognitive network system as neural correlates of low-grade inflammation and stress exposure, and suggest that low-grade inflammation, alongside with stress, may render individuals more vulnerable to PTSD.
Collapse
Affiliation(s)
- Jungyoon Kim
- Ewha Brain Institute, Ewha Womans University, Seoul, South Korea.,Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul, South Korea
| | - Sujung Yoon
- Ewha Brain Institute, Ewha Womans University, Seoul, South Korea.,Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul, South Korea
| | - Suji Lee
- Ewha Brain Institute, Ewha Womans University, Seoul, South Korea.,Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul, South Korea
| | - Haejin Hong
- Ewha Brain Institute, Ewha Womans University, Seoul, South Korea.,Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul, South Korea
| | - Eunji Ha
- Ewha Brain Institute, Ewha Womans University, Seoul, South Korea.,Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul, South Korea
| | - Yoonji Joo
- Ewha Brain Institute, Ewha Womans University, Seoul, South Korea.,Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, South Korea
| | - Eun Hee Lee
- Green Cross Laboratories, Yongin, South Korea
| | - In Kyoon Lyoo
- Ewha Brain Institute, Ewha Womans University, Seoul, South Korea. .,Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul, South Korea. .,Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, South Korea. .,The Brain Institute and Department of Psychiatry, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
85
|
Ting EYC, Yang AC, Tsai SJ. Role of Interleukin-6 in Depressive Disorder. Int J Mol Sci 2020; 21:ijms21062194. [PMID: 32235786 PMCID: PMC7139933 DOI: 10.3390/ijms21062194] [Citation(s) in RCA: 180] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 03/18/2020] [Accepted: 03/21/2020] [Indexed: 12/20/2022] Open
Abstract
Major depressive disorder (MDD), which is a leading psychiatric illness across the world, severely affects quality of life and causes an increased incidence of suicide. Evidence from animal as well as clinical studies have indicated that increased peripheral or central cytokine interleukin-6 (IL-6) levels play an important role in stress reaction and depressive disorder, especially physical disorders comorbid with depression. Increased release of IL-6 in MDD has been found to be a factor associated with MDD prognosis and therapeutic response, and may affect a wide range of depressive symptomatology. However, study results of the IL6 genetic effects in MDD are controversial. Increased IL-6 activity may cause depression through activation of hypothalamic-pituitary-adrenal axis or influence of the neurotransmitter metabolism. The important role of neuroinflammation in MDD pathogenesis has created a new perspective that the combining of blood IL-6 and other depression-related cytokine levels may help to classify MDD biological subtypes, which may allow physicians to identify the optimal treatment for MDD patients. To modulate the IL-6 activity by IL-6-related agents, current antidepressive agents, herb medication, pre-/probiotics or non-pharmacological interventions may hold great promise for the MDD patients with inflammatory features.
Collapse
Affiliation(s)
- Emily Yi-Chih Ting
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei 11217, Taiwan;
| | - Albert C. Yang
- Brain Research Center, National Yang-Ming University, Taipei 11221, Taiwan;
- Division of Interdisciplinary Medicine and Biotechnology, Beth Israel Deaconess, Medical Center, Boston, MA 02115, USA
| | - Shih-Jen Tsai
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei 11217, Taiwan;
- Brain Research Center, National Yang-Ming University, Taipei 11221, Taiwan;
- School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan
- Correspondence: ; Tel.: +886-2-28757027 (ext. 276); Fax: +886-2-28725643
| |
Collapse
|
86
|
Tsamakis K, Mueller C, Tsirigotis P, Tsiptsios D, Tsamakis C, Charakopoulos E, Charalampous C, Spandidos DA, Douzenis A, Papageorgiou C, Liappas I, Rizos E. Depression following graft-versus-host disease in a patient with acute lymphoblastic leukaemia: A case report. Mol Clin Oncol 2020; 12:208-211. [PMID: 32064096 PMCID: PMC7016518 DOI: 10.3892/mco.2019.1970] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 12/17/2019] [Indexed: 12/27/2022] Open
Abstract
There is increasing evidence to suggest an association between depression and inflammation, with patients suffering from immune mediated-disorders exhibiting higher levels of depression. Inflammation in depression is a potential target for the development of novel treatment strategies. The present study presents a clinical case in which a patient with an underlying inflammatory condition acutely developed a severe depressive episode resulting in a sudden, dramatic change in their clinical picture. This case, with no similar case reports being in the literature thus far, at least to the best of our knowledge, highlights the increasing consideration that there may be a causative role between neuro-inflammation and depression. This study reports the case of a 40-year-old male with acute lymphoblastic leukaemia (ALL) and no previous psychiatric history, who developed an acute onset of a severe depressive episode in the context of the immune-mediated graft-versus-host disease (GVHD). GVHD is a complication of allogeneic hematopoietic cell transplantation, which the patient had undergone, for the treatment of his ALL. The rapid onset of depression could be explained by the neuroinflammatory processes occurring in GVHD. This provides a clinical example for the possible role of the immune system in depression, and clinicians should be aware of this association.
Collapse
Affiliation(s)
- Konstantinos Tsamakis
- Second Department of Psychiatry, School of Medicine, University General Hospital 'Attikon', 12462 Athens, Greece
| | - Christoph Mueller
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, London SE5 8AF, London, UK.,South London and Maudsley NHS Foundation Trust, London, UK
| | - Panagiotis Tsirigotis
- Haematology Clinic, School of Medicine, University General Hospital 'Attikon', 12462 Athens, Greece
| | | | - Charalampos Tsamakis
- Department of Dermatology, School of Medicine, University General Hospital 'Attikon', 12462 Athens
| | - Emmanouil Charakopoulos
- Second Department of Psychiatry, School of Medicine, University General Hospital 'Attikon', 12462 Athens, Greece
| | - Chistophis Charalampous
- Second Department of Psychiatry, School of Medicine, University General Hospital 'Attikon', 12462 Athens, Greece
| | - Demetrios A Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, 71003 Heraklion, Greece
| | - Athanasios Douzenis
- Second Department of Psychiatry, School of Medicine, University General Hospital 'Attikon', 12462 Athens, Greece
| | - Charalabos Papageorgiou
- First Department of Psychiatry, Athens University Medical School, Eginition Hospital, 11528 Athens, Greece
| | - Ioannis Liappas
- First Department of Psychiatry, Athens University Medical School, Eginition Hospital, 11528 Athens, Greece
| | - Emmanouil Rizos
- Second Department of Psychiatry, School of Medicine, University General Hospital 'Attikon', 12462 Athens, Greece
| |
Collapse
|
87
|
Kurt HG, Altinay M, Anand A. Comparative Efficacy of Ketamine in Treatment-Resistant Depression. Psychiatr Ann 2020. [DOI: 10.3928/00485713-20200113-01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
88
|
Wilkowska A, Szałach Ł, Cubała WJ. Ketamine in Bipolar Disorder: A Review. Neuropsychiatr Dis Treat 2020; 16:2707-2717. [PMID: 33209026 PMCID: PMC7670087 DOI: 10.2147/ndt.s282208] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 10/10/2020] [Indexed: 12/25/2022] Open
Abstract
Bipolar disorder (BD) is a psychiatric illness associated with high morbidity, mortality and suicide rate. It has neuroprogressive course and a high rate of treatment resistance. Hence, there is an unquestionable need for new BD treatment strategies. Ketamine appears to have rapid antidepressive and antisuicidal effects. Since most of the available studies concern unipolar depression, here we present a novel insight arguing that ketamine might be a promising treatment for bipolar disorder.
Collapse
Affiliation(s)
- Alina Wilkowska
- Department of Psychiatry, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Łukasz Szałach
- Department of Psychiatry, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Wiesław J Cubała
- Department of Psychiatry, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland
| |
Collapse
|
89
|
Draganov M, Arranz MJ, Salazar J, de Diego-Adeliño J, Gallego-Fabrega C, Jubero M, Carceller-Sindreu M, Portella MJ. Association study of polymorphisms within inflammatory genes and methylation status in treatment response in major depression. Eur Psychiatry 2020; 60:7-13. [DOI: 10.1016/j.eurpsy.2019.05.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 05/03/2019] [Accepted: 05/05/2019] [Indexed: 12/28/2022] Open
Abstract
AbstractBackground:Although pharmacogenetics for major depressive disorder (MDD) is gaining momentum, the role of genetics in differences in response to antidepressant treatment is controversial, as they depend on multifactorial and polygenic phenotypes. Previous studies focused on the genes of the serotonergic system, leaving apart other pathological factors such as the inflammatory pathway. The main objective of the study was to assess whether treatment response might be associated with specific inflammation-related genetic variants or their methylation status.Methods:41 SNPs in 8 inflammatory genes: interleukin (IL) 1-β, IL2, IL6, IL6R, IL10, IL18, tumor necrosis factor (TNF)-α and interferon (IFN)-γ were genotyped in 153 patients with MDD, who were evaluated with the Mausdley Staging Method to determine treatment response profiles. Pyrosequencing reactions and methylation quantification were performed in a PyroMark Q24 in 5 selected CpG islands of IL1- β, IL6 and IL6R. Linear and logistic regression analyses were conducted, including age and gender as covariates using PLINK 1.07.Results:Allelic distribution of IL1- β rs1143643 was significantly associated with MSM scores (FDR corrected p = 0.04). Allelic distribution of IL6R rs57569414 showed a trend towards significance with MSM scores (p = 0.002; FDR corrected p = 0.07). Haplotype analyses showed associations between allelic combinations of IL1-β and IL10 with treatment response (FDR corrected p < 0.01). Methylation percentage of treatment responders was only higher in an IL6R CpG island (p < 0.05).Conclusions:These exploratory findings suggest that IL1-β and, marginally, IL6R polymorphisms may affect treatment response in major depression. If confirmed, these results may account for the heterogeneous phenotypes of major depression that underlie differences in treatment response.
Collapse
|
90
|
Roman M, Irwin MR. Novel neuroimmunologic therapeutics in depression: A clinical perspective on what we know so far. Brain Behav Immun 2020; 83:7-21. [PMID: 31550500 PMCID: PMC6940145 DOI: 10.1016/j.bbi.2019.09.016] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 09/11/2019] [Accepted: 09/20/2019] [Indexed: 12/27/2022] Open
Abstract
Depression, one of the most common mental health disorders, is among the leading causes of health-related disability worldwide. Although antidepressant treatment has been available for decades, depression remains largely refractory to the prevailing limited treatment approach of monoamine transmission modulation. Fortunately, recent evidence points to a link between depression and inflammatory factors within the innate and the adaptive immune system. The purpose of this review is to evaluate current and potential clinical immunotherapies for depression, as contextually focused by an immunologic lens of the pathophysiologic mechanisms of depression. The utility of pro-inflammatory cytokines (primarily interleukin-1β, interleukin -6 and tumor necrosis factor-α) is considered in their role as screening biomarkers in prediction of treatment response or nonresponse. The evidence base of numerous recent clinical studies is discussed as related to their antidepressant efficacy and favorable safety profile, with consideration of multiple agents that target inflammatory mechanisms linked to depression including nonsteroidal anti-inflammatory pathways (i.e., aspirin, celecoxib), cytokine antagonism (i.e., etanercept, infliximab), N-methyl-D-aspartate receptor (NMDA) receptor antagonism (i.e., ketamine), and modulation of kynurenine pathways (i.e., minocycline). Additionally, new and exciting directions in targeting inflammatory mechanisms in the treatment of depression are underway, and future investigation is also warranted to explore the utility of inflammation in diagnosing depression, guiding clinical treatment decision-making, and monitoring disease burden and relapse risk.
Collapse
Affiliation(s)
- Michael Roman
- University of Pennsylvania, Psychiatry Residency Program, Philadelphia, PA, United States
| | - Michael R Irwin
- Cousins Center for Psychoneuroimmunology, Jane and Terry Semel Institute for Neuroscience and Human Behavior at UCLA, and Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, Los Angeles, CA, United States.
| |
Collapse
|
91
|
Rengasamy M, Hsiung K, Price RB. Infusing hope into the treatment of suicidality: A review of ketamine's effects on suicidality. Curr Behav Neurosci Rep 2019; 6:166-176. [PMID: 33457182 PMCID: PMC7809881 DOI: 10.1007/s40473-019-00184-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
PURPOSE OF REVIEW Given recent increases in rates of suicide and lack of rapid treatments for suicidality, ketamine has been identified as a potential fast-acting anti-suicidal treatment. Our review seeks to describe the effects of ketamine on suicidality, given the growing literature on the use of ketamine in reducing suicidality. We examine open-label studies and randomized controlled trials evaluating treatment of suicidality with ketamine. Furthermore, our manuscript identifies potential mechanisms of ketamine's effects on suicidality. RECENT FINDINGS Based on existing RCTs, ketamine appears to have rapid anti-suicidal effects, with most literature studying such effects in timeframes less than one week. Although still in the early stages of research, mechanisms of ketamine include modulation of molecular, inflammatory, neural, cognitive, and behavioral processes. SUMMARY Thus, ketamine appears to be a promising treatment for suicidality, but requires larger scale and more robust RCTs to confirm the potential use of this agent in clinical settings.
Collapse
Affiliation(s)
- Manivel Rengasamy
- Department of Psychiatry, University of Pittsburgh School
of Medicine, Pittsburgh, PA
| | | | - Rebecca B. Price
- Department of Psychiatry, University of Pittsburgh School
of Medicine, Pittsburgh, PA
| |
Collapse
|
92
|
Alterations in the Levels of Growth Factors in Adolescents with Major Depressive Disorder: A Longitudinal Study during the Treatment with Fluoxetine. Mediators Inflamm 2019; 2019:9130868. [PMID: 31827384 PMCID: PMC6885844 DOI: 10.1155/2019/9130868] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 10/14/2019] [Indexed: 12/26/2022] Open
Abstract
Major depressive disorder (MDD) has a prevalence of 5% in adolescents. Several studies have described the association between the inflammatory response and MDD, but little is known about the relationship between MDD and growth factors, such as IL-7, IL-9, IL-17A, VEGF, basic FGF, G-CSF, and GM-CSF. It must be appointed that there are scarce reports on growth factors in adolescents with MDD and even fewer with a clinical follow-up. In this work, we evaluated the levels of growth factors (IL-7, IL-9, IL-17A, VEGF, basic FGF, G-CSF, and GM-CSF) in MDD adolescents and the clinical follow-up during eight weeks of treatment with fluoxetine. Methods. All patients were diagnosed according to the DSM-IV-TR, and the severity of the symptoms was evaluated using the Hamilton Depression Rating Scale (HDRS). Growth factors IL-7, IL-9, IL-17A, VEGF, basic FGF, G-CSF, and GM-CSF were quantified by cytometric bead array using serum samples from 22 adolescents with MDD and 18 healthy volunteers. Results. All patients showed clinical improvement since the fourth week of pharmacological treatment according to the HDRS. Considerably higher levels of IL-7, IL-9, IL-17A, VEGF, basic FGF, G-CSF, and GM-CSF were detected in MDD adolescents as compared to healthy volunteers. A significant but temporal decrease was detected in basic FGF, G-CSF, and GM-CSF at week four of fluoxetine administration. Conclusions. To the best of our knowledge, this is the first report to show alterations in the levels of growth factors, such as IL-7, IL-9, IL-17A, VEGF, basic FGF, G-CSF, and GM-CSF in MDD adolescents during eight weeks of clinical follow-up. These disturbances might be involved in the physiopathology of MDD since such growth factors have been proven to participate in the neural development and correct functioning of the CNS; therefore, subtle alterations in it may contribute to MDD.
Collapse
|
93
|
Ho MF, Zhang C, Zhang L, Li H, Weinshilboum RM. Ketamine and Active Ketamine Metabolites Regulate STAT3 and the Type I Interferon Pathway in Human Microglia: Molecular Mechanisms Linked to the Antidepressant Effects of Ketamine. Front Pharmacol 2019; 10:1302. [PMID: 31827434 PMCID: PMC6848891 DOI: 10.3389/fphar.2019.01302] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 10/11/2019] [Indexed: 11/13/2022] Open
Abstract
Inflammation is an important biological process which contributes to risk for depression, in part as a result of the production of proinflammatory cytokines and of alterations in glutamatergic neurotransmission. Ketamine has anti-inflammatory properties which might contribute to its antidepressant effects. This study was designed to clarify mechanisms of action for ketamine and its active metabolites, (2R,6R;2S,6S)-hydroxynorketamine (HNK), which also appear to play a major role in ketamine's rapid antidepressant effects. An HMC3 human microglial cell line was used as a model system to test a possible role for ketamine in immune response regulation that might contribute to its antidepressant effects. Our results highlight the fact that ketamine and its two active metabolites can regulate the type I interferon pathway mediated, at least partially, through signal transducer and activation of transcription 3 (STAT3) which plays a major role in the immune response. Specifically, STAT3 downstream genes that were modulated by either ketamine or its active metabolites were enriched in the "response to type I interferon" pathway. Our data also suggest that STAT3 might play a role in ketamine's antidepressant effects, mediated, at least in part, through eukaryotic elongation factor 2 (EEF2), resulting in the augmentation of brain-derived neurotropic factor (BDNF) expression and promoting the synthesis of synaptic proteins postsynaptic density protein 95 (PSD95) and synapsin I (SYN1).
Collapse
Affiliation(s)
- Ming-Fen Ho
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
| | - Cheng Zhang
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
| | - Lingxin Zhang
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
| | - Hu Li
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
| | - Richard M Weinshilboum
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
94
|
Mesenchymal Stromal Cells Modulate Peripheral Stress-Induced Innate Immune Activation Indirectly Limiting the Emergence of Neuroinflammation-Driven Depressive and Anxiety-like Behaviors. Biol Psychiatry 2019; 86:712-724. [PMID: 31521333 DOI: 10.1016/j.biopsych.2019.07.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 07/05/2019] [Accepted: 07/15/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND Hyperactivation of innate immunity has been implicated in the etiology of mood disorders, including major depressive disorder (MDD). Mesenchymal stromal cells (MSCs) have demonstrated potent immunomodulatory capabilities in the context of chronic inflammatory disease and injury but have yet to be evaluated in stress-based preclinical models of MDD. We sought to test the ability of intravenous MSCs to modulate innate immune activation and behavioral patterns associated with repeated social defeat (RSD). METHODS Murine RSD-induced innate immune activation as well as depressive and anxiety-like behaviors were assessed in unstressed, RSD, and RSD + human MSC groups. Biodistribution and fate studies were performed to inform potential mechanisms of action. RESULTS MSCs reduced stress-induced circulating proinflammatory cytokines, monocytes, neuroinflammation, and depressive and anxiety-like behaviors. Biodistribution analyses indicated that infused MSCs distributed within peripheral organs without homing to the brain. Murine neutrophils targeted MSCs in the lungs within hours of administration. MSCs and recipient neutrophils were cleared by recipient macrophages promoting a switch toward a regulatory phenotype and systemic resolution of inflammation. CONCLUSIONS Peripheral delivery of MSCs modulates central nervous system inflammatory processes and aberrant behavioral patterns in a stress-based rodent model of MDD and anxiety. Recent studies suggest that host immune cell-mediated phagocytosis of MSCs in vivo can trigger an immunomodulatory cascade, resulting in resolution of inflammation. Our data suggest that similar mechanisms may protect distal organs, including the brain, from systemic, stress-induced proinflammatory spikes and may uncover unexpected targets in the periphery for novel or adjunct treatment for a subset of patients with MDD.
Collapse
|
95
|
Verdonk F, Petit AC, Abdel-Ahad P, Vinckier F, Jouvion G, de Maricourt P, De Medeiros GF, Danckaert A, Van Steenwinckel J, Blatzer M, Maignan A, Langeron O, Sharshar T, Callebert J, Launay JM, Chrétien F, Gaillard R. Microglial production of quinolinic acid as a target and a biomarker of the antidepressant effect of ketamine. Brain Behav Immun 2019; 81:361-373. [PMID: 31255681 DOI: 10.1016/j.bbi.2019.06.033] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 06/21/2019] [Accepted: 06/22/2019] [Indexed: 12/27/2022] Open
Abstract
Major depressive disorder is a complex multifactorial condition with a so far poorly characterized underlying pathophysiology. Consequently, the available treatments are far from satisfactory as it is estimated that up to 30% of patients are resistant to conventional treatment. Recent comprehensive evidence has been accumulated which suggests that inflammation may be implied in the etiology of this disease. Here we investigated ketamine as an innovative treatment strategy due to its immune-modulating capacities. In a murine model of LPS-induced depressive-like behavior we demonstrated that a single dose of ketamine restores the LPS-induced depressive-like alterations. These behavioral effects are associated with i/ a reversal of anxiety and reduced self-care, ii/ a decrease in parenchymal cytokine production, iii/ a modulation of the microglial reactivity and iv/ a decrease in microglial quinolinic acid production that is correlated with plasmatic peripheral production. In a translational approach, we show that kynurenic acid to quinolinic acid ratio is a predictor of ketamine response in treatment-resistant depressed patients and that the reduction in quinolinic acid after a ketamine infusion is a predictor of the reduction in MADRS score. Our results suggest that microglia is a key therapeutic target and that quinolinic acid is a biomarker of ketamine response in major depressive disorder.
Collapse
Affiliation(s)
- Franck Verdonk
- Institut Pasteur, Experimental Neuropathology Unit, Infection and Epidemiology Department, Paris, France; Department of Anaesthesiology and Intensive Care, Saint Antoine Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France; Sorbonne University, Paris, France
| | - Anne-Cécile Petit
- Institut Pasteur, Experimental Neuropathology Unit, Infection and Epidemiology Department, Paris, France; Service Hospitalo Universitaire, Centre Hospitalier Sainte-Anne, Paris, France; Paris Descartes University, Sorbonne Paris Cité, Paris, France
| | - Pierre Abdel-Ahad
- Service Hospitalo Universitaire, Centre Hospitalier Sainte-Anne, Paris, France; Paris Descartes University, Sorbonne Paris Cité, Paris, France; INSERM, Laboratoire de "Physiopathologie des maladies Psychiatriques", Centre de psychiatrie et neurosciences, CPN U894, Institut de psychiatrie (GDR 3557), Paris, France
| | - Fabien Vinckier
- Service Hospitalo Universitaire, Centre Hospitalier Sainte-Anne, Paris, France; Paris Descartes University, Sorbonne Paris Cité, Paris, France; INSERM, Laboratoire de "Physiopathologie des maladies Psychiatriques", Centre de psychiatrie et neurosciences, CPN U894, Institut de psychiatrie (GDR 3557), Paris, France
| | - Gregory Jouvion
- Institut Pasteur, Experimental Neuropathology Unit, Infection and Epidemiology Department, Paris, France
| | - Pierre de Maricourt
- Service Hospitalo Universitaire, Centre Hospitalier Sainte-Anne, Paris, France; Paris Descartes University, Sorbonne Paris Cité, Paris, France; INSERM, Laboratoire de "Physiopathologie des maladies Psychiatriques", Centre de psychiatrie et neurosciences, CPN U894, Institut de psychiatrie (GDR 3557), Paris, France
| | | | - Anne Danckaert
- Institut Pasteur, Experimental Neuropathology Unit, Infection and Epidemiology Department, Paris, France; Institut Pasteur, UtechS Photonic BioImaging (Imagopole) - C2RT, Paris, France
| | - Juliette Van Steenwinckel
- Inserm, U1141 Paris, France; Paris Diderot University, Sorbonne Paris Cité, UMRS 1141, F-75019 Paris, France
| | - Michael Blatzer
- Institut Pasteur, Experimental Neuropathology Unit, Infection and Epidemiology Department, Paris, France
| | - Anna Maignan
- Service Universitaire de Psychiatrie d'adultes, Centre Hospitalier de Versailles, Le Chesnay, France
| | - Olivier Langeron
- Institut Pasteur, Experimental Neuropathology Unit, Infection and Epidemiology Department, Paris, France; Sorbonne University, Paris, France; Multidisciplinary Intensive Care Unit, Department of Anesthesiology and Critical Care, La Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Tarek Sharshar
- Institut Pasteur, Experimental Neuropathology Unit, Infection and Epidemiology Department, Paris, France; Paris Descartes University, Sorbonne Paris Cité, Paris, France; Department of Intensive Care, Centre Hospitalier Sainte Anne, Paris, France
| | - Jacques Callebert
- Service de Biochimie et Biologie Moléculaire, INSERM U942, Hôpital Lariboisière, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Jean-Marie Launay
- Service de Biochimie et Biologie Moléculaire, INSERM U942, Hôpital Lariboisière, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Fabrice Chrétien
- Institut Pasteur, Experimental Neuropathology Unit, Infection and Epidemiology Department, Paris, France; Paris Descartes University, Sorbonne Paris Cité, Paris, France; Laboratoire hospitalo-universitaire de Neuropathologie, Centre Hospitalier Sainte Anne, Paris, France.
| | - Raphael Gaillard
- Institut Pasteur, Experimental Neuropathology Unit, Infection and Epidemiology Department, Paris, France; Service Hospitalo Universitaire, Centre Hospitalier Sainte-Anne, Paris, France; Paris Descartes University, Sorbonne Paris Cité, Paris, France.
| |
Collapse
|
96
|
Hashimoto K. Rapid-acting antidepressant ketamine, its metabolites and other candidates: A historical overview and future perspective. Psychiatry Clin Neurosci 2019; 73:613-627. [PMID: 31215725 PMCID: PMC6851782 DOI: 10.1111/pcn.12902] [Citation(s) in RCA: 228] [Impact Index Per Article: 45.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 05/28/2019] [Accepted: 06/06/2019] [Indexed: 12/11/2022]
Abstract
Major depressive disorder (MDD) is one of the most disabling psychiatric disorders. Approximately one-third of the patients with MDD are treatment resistant to the current antidepressants. There is also a significant therapeutic time lag of weeks to months. Furthermore, depression in patients with bipolar disorder (BD) is typically poorly responsive to antidepressants. Therefore, there exists an unmet medical need for rapidly acting antidepressants with beneficial effects in treatment-resistant patients with MDD or BD. Accumulating evidence suggests that the N-methyl-D-aspartate receptor (NMDAR) antagonist ketamine produces rapid and sustained antidepressant effects in treatment-resistant patients with MDD or BD. Ketamine is a racemic mixture comprising equal parts of (R)-ketamine (or arketamine) and (S)-ketamine (or esketamine). Because (S)-ketamine has higher affinity for NMDAR than (R)-ketamine, esketamine was developed as an antidepressant. On 5 March 2019, esketamine nasal spray was approved by the US Food and Drug Administration. However, preclinical data suggest that (R)-ketamine exerts greater potency and longer-lasting antidepressant effects than (S)-ketamine in animal models of depression and that (R)-ketamine has less detrimental side-effects than (R,S)-ketamine or (S)-ketamine. In this article, the author reviews the historical overview of the antidepressant actions of enantiomers of ketamine and its major metabolites norketamine and hydroxynorketamine. Furthermore, the author discusses the other potential rapid-acting antidepressant candidates (i.e., NMDAR antagonists and modulators, low-voltage-sensitive T-type calcium channel inhibitor, potassium channel Kir4.1 inhibitor, negative modulators of γ-aminobutyric acid, and type A [GABAA ] receptors) to compare them with ketamine. Moreover, the molecular and cellular mechanisms of ketamine's antidepressant effects are discussed.
Collapse
Affiliation(s)
- Kenji Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| |
Collapse
|
97
|
Yang C, Wardenaar KJ, Bosker FJ, Li J, Schoevers RA. Inflammatory markers and treatment outcome in treatment resistant depression: A systematic review. J Affect Disord 2019; 257:640-649. [PMID: 31357161 DOI: 10.1016/j.jad.2019.07.045] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 06/03/2019] [Accepted: 07/04/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND A substantial percentage of depressed patients do not respond satisfactorily to conventional antidepressant treatment. This treatment resistant depression (TRD) may be partly related to inflammatory processes in the central nervous system. Accordingly, peripheral inflammatory markers might serve to predict treatment response with novel but still experimental forms of antidepressant treatment. METHODS A literature search on treatment of TRD and inflammatory markers was performed using the PubMed/Medline database on November 8th 2018, and 95 articles were retrieved initially, which were subsequently screened and selected only when the inclusion and exclusion criteria were met. RESULTS Ten studies were recruited. In five studies higher baseline interleukin-6 (IL-6) or C-reactive protein (CRP)/high-sensitivity-CRP (hsCRP) in blood predicted better response to medication with anti-inflammatory characteristics, such as ketamine and infliximab. One study found that higher IL-6 predicted worse response to antidepressant treatment in patients with TRD. No evidence was found for the predictive value of other inflammatory markers (e.g., Tumor Necrosis Factor-α, Interferon-γ). LIMITATIONS The number of available studies was limited; included studies showed considerable methodological variation and used different definitions for TRD. CONCLUSION The inflammatory markers IL-6 and CRP/hsCRP could hold promise as markers for the prediction of treatment response in TRD. Clearly, this field of research is still far from mature but it could pave the way for novel and efficacious treatments for at least the inflammatory type of TRD with more well-designed studies and more convincing results.
Collapse
Affiliation(s)
- Chenghao Yang
- Tianjin Mental Health Institute, Tianjin Anding Hospital, Tianjin, China; University Centre of Psychiatry, University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands; University of Groningen, Research School Behavioral and Cognitive Neurosciences (BCN), Groningen, the Netherlands
| | - Klaas J Wardenaar
- University of Groningen, University Medical Center Groningen, Department of Psychiatry, Interdisciplinary Center Psychopathology and Emotion regulation (ICPE), Groningen, the Netherlands
| | - Fokko J Bosker
- University Centre of Psychiatry, University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands; University of Groningen, Research School Behavioral and Cognitive Neurosciences (BCN), Groningen, the Netherlands
| | - Jie Li
- Tianjin Mental Health Institute, Tianjin Anding Hospital, Tianjin, China
| | - Robert A Schoevers
- University Centre of Psychiatry, University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands; University of Groningen, Research School Behavioral and Cognitive Neurosciences (BCN), Groningen, the Netherlands.
| |
Collapse
|
98
|
Yuan N, Chen Y, Xia Y, Dai J, Liu C. Inflammation-related biomarkers in major psychiatric disorders: a cross-disorder assessment of reproducibility and specificity in 43 meta-analyses. Transl Psychiatry 2019; 9:233. [PMID: 31534116 PMCID: PMC6751188 DOI: 10.1038/s41398-019-0570-y] [Citation(s) in RCA: 239] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 05/24/2019] [Indexed: 12/15/2022] Open
Abstract
Inflammation is a natural defence response of the immune system against environmental insult, stress and injury, but hyper- and hypo-inflammatory responses can trigger diseases. Accumulating evidence suggests that inflammation is involved in multiple psychiatric disorders. Using inflammation-related factors as biomarkers of psychiatric disorders requires the proof of reproducibility and specificity of the changes in different disorders, which remains to be established. We performed a cross-disorder study by systematically evaluating the meta-analysis results of inflammation-related factors in eight major psychiatric disorders, including schizophrenia (SCZ), bipolar disorder (BD), autism spectrum disorder (ASD), major depression disorder (MDD), post-trauma stress disorder (PTSD), sleeping disorder (SD), obsessive-compulsive disorder (OCD) and suicide. A total of 43 meta-analyses involving 704 publications on 44 inflammation-related factors were included in the study. We calculated the effect size and statistical power for every inflammation-related factor in each disorder. Our analyses showed that well-powered case-control studies provided more consistent results than underpowered studies when one factor was meta-analysed by different researchers. After removing underpowered studies, 30 of the 44 inflammation-related factors showed significant alterations in at least one disorder based on well-powered meta-analyses. Eleven of them changed in patients of more than two disorders when compared with the controls. A few inflammation-related factors showed unique changes in specific disorders (e.g., IL-4 increased in BD, decreased in suicide, but had no change in MDD, ASD, PTSD and SCZ). MDD had the largest number of changes while SD has the least. Clustering analysis showed that closely related disorders share similar patterns of inflammatory changes, as genome-wide genetic studies have found. According to the effect size obtained from the meta-analyses, 13 inflammation-related factors would need <50 cases and 50 controls to achieve 80% power to show significant differences (p < 0.0016) between patients and controls. Changes in different states of MDD, SCZ or BD were also observed in various comparisons. Studies comparing first-episode SCZ to controls may have more reproducible findings than those comparing pre- and post-treatment results. Longitudinal, system-wide studies of inflammation regulation that can differentiate trait- and state-specific changes will be needed to establish valuable biomarkers.
Collapse
Affiliation(s)
- Ning Yuan
- Department of Psychiatry, The Second Xiangya Hospital; Mental health Institute of the Second Xiangya Hospital; National Clinical Research Center on Mental Disorders; National Technology Institute on Mental Disorders, Central South University, Changsha, Hunan, China
- Department of Psychiatry, Hunan Provincial Brain Hospital; Clinical Research Center for Mental Behavioral Disorder in Hunan Province, Clinical Medical School of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yu Chen
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Yan Xia
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Jiacheng Dai
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Chunyu Liu
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China.
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, USA.
- School of Psychology, Shaanxi Normal University, Xi'an, Shaanxi, China.
| |
Collapse
|
99
|
Xu YH, Zhu Y, Zhu YY, Wei H, Zhang NN, Qin JS, Zhu XL, Yu M, Li YF. Abnormalities in FGF family members and their roles in modulating depression-related molecules. Eur J Neurosci 2019; 53:140-150. [PMID: 31491043 DOI: 10.1111/ejn.14570] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 08/22/2019] [Accepted: 09/02/2019] [Indexed: 12/22/2022]
Abstract
The role of the fibroblast growth factor (FGF) system in depression has received considerable attention in recent years. To understand the role of this system, it is important to identify the specific members of the FGF family that have been implicated and the various mechanisms that they modulated. Here, we review the role of FGFs in depression and integrate evidence from clinical and basic research. These data suggest that changes in the FGF family are involved in depression and possibly in a wider range of psychiatric disorders. We analyse the abnormalities of FGF family members in depression and their roles in modulating depression-related molecules. The role of the FGF family in depression and related disorders needs to be studied in more detail.
Collapse
Affiliation(s)
- Yu-Hao Xu
- Department of Neurology, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China.,Department of Neuroimaging laboratory, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yan Zhu
- Department of Neuroimaging laboratory, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China.,Department of Radiology, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yuan-Yuan Zhu
- Department of Neurology, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China.,Department of Neuroimaging laboratory, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Hong Wei
- Department of Neurology, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China.,Department of Neuroimaging laboratory, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Ning-Ning Zhang
- Department of Neuroimaging laboratory, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China.,Department of Radiology, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Jia-Sheng Qin
- Department of Neuroimaging laboratory, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China.,Department of Radiology, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xiao-Lan Zhu
- Department of Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Ming Yu
- Department of Neurology, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yue-Feng Li
- Department of Neuroimaging laboratory, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China.,Department of Radiology, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
100
|
Carboni L, McCarthy DJ, Delafont B, Filosi M, Ivanchenko E, Ratti E, Learned SM, Alexander R, Domenici E. Biomarkers for response in major depression: comparing paroxetine and venlafaxine from two randomised placebo-controlled clinical studies. Transl Psychiatry 2019; 9:182. [PMID: 31375659 PMCID: PMC6677721 DOI: 10.1038/s41398-019-0521-7] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 04/19/2019] [Accepted: 06/20/2019] [Indexed: 02/08/2023] Open
Abstract
The identification of biomarkers of response might speed drug development and set the premises to assist clinical practice in psychiatry. In this work, we evaluated a panel of peripheral biomarkers (including IL-6, IL-10, TNF-α, TNFRII, BDNF, CRP, MMP9 and PAI1) in depressed patients receiving paroxetine, venlafaxine, or placebo. Samples were obtained from two randomised placebo-controlled studies evaluating the efficacy and tolerability of a novel drug candidate, using either paroxetine or venlafaxine as active comparators. In both studies, the biomarker candidates were analysed in plasma collected at randomization and after 10 weeks of treatment with either placebo or active comparator (for a total of 106 and 108 subjects in the paroxetine and venlafaxine study, respectively). Data were obtained by multiplexing sandwich-ELISA system. Data were subjected to statistical analysis to assess their correlation with baseline severity and with response outcome. Increases in biomarker levels were correlated with reduction in depression severity for TNF-α, IL-6 IL-10 and CRP. Response to paroxetine treatment correlated with baseline IL-10, IL-6 and TNF-α levels, with the strongest signal being observed in males. In the venlafaxine study, a correlation was observed only between CRP level at randomisation and response, suggesting differences between the two active treatments and the two studies. Our investigations suggest that a combination of pro- and anti-inflammatory cytokines may predict response outcome in patients treated with paroxetine. The potential for IL-10, IL-6 and TNF-α as response biomarkers for a wider range of antidepressants warrants further investigations in clinical trials with other monoamine reuptake inhibitors.
Collapse
Affiliation(s)
- Lucia Carboni
- 0000 0004 1757 1758grid.6292.fDepartment of Pharmacy and Biotechnology, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Dennis J. McCarthy
- Indipendent Consultant, Clinical Pharmacology and Translational Science, Newark, DE USA
| | | | - Michele Filosi
- 0000 0004 1937 0351grid.11696.39Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | | | - Emiliangelo Ratti
- 0000 0004 0447 7762grid.419849.9Neuroscience Therapeutic Area Unit, Takeda, Boston, MA USA
| | - Susan M. Learned
- grid.504165.3Global Medicines Development, Indivior, Inc., Richmond, VA USA
| | - Robert Alexander
- 0000 0004 0447 7762grid.419849.9Neuroscience Therapeutic Area Unit, Takeda, Boston, MA USA
| | - Enrico Domenici
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy. .,Fondazione The Microsoft Research-University of Trento Centre for Computational and Systems Biology (COSBI), Rovereto, TN, Italy.
| |
Collapse
|