251
|
Luning Prak ET, Brooks T, Makhoul W, Beer JC, Zhao L, Girelli T, Skarke C, Sheline YI. No increase in inflammation in late-life major depression screened to exclude physical illness. Transl Psychiatry 2022; 12:118. [PMID: 35332134 PMCID: PMC8948274 DOI: 10.1038/s41398-022-01883-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/18/2022] [Accepted: 03/02/2022] [Indexed: 12/12/2022] Open
Abstract
Depression is a common and debilitating disorder in the elderly. Late-life depression (LLD) has been associated with inflammation and elevated levels of proinflammatory cytokines including interleukin (IL)-1β, tumor necrosis factor-alpha, and IL-6, but often depressed individuals have comorbid medical conditions that are associated with immune dysregulation. To determine whether depression has an association with inflammation independent of medical illness, 1120 adults were screened to identify individuals who had clinically significant depression but not medical conditions associated with systemic inflammation. In total, 66 patients with LLD screened to exclude medical conditions associated with inflammation were studied in detail along with 26 age-matched controls (HC). At baseline, circulating cytokines were low and similar in LLD and HC individuals. Furthermore, cytokines did not change significantly after treatment with either an antidepressant (escitalopram 20 mg/day) or an antidepressant plus a COX-2 inhibitor or placebo, even though depression scores improved in the non-placebo treatment arms. An analysis of cerebrospinal fluid in a subset of individuals for IL-1β using an ultrasensitive digital enzyme-linked immunosorbent assay revealed low levels in both LLD and HC at baseline. Our results indicate that depression by itself does not result in systemic or intrathecal elevations in cytokines and that celecoxib does not appear to have an adjunctive antidepressant role in older patients who do not have medical reasons for having inflammation. The negative finding for increased inflammation and the lack of a treatment effect for celecoxib in this carefully screened depressed population taken together with multiple positive results for inflammation in previous studies that did not screen out physical illness support a precision medicine approach to the treatment of depression that takes the medical causes for inflammation into account.
Collapse
Affiliation(s)
- Eline T. Luning Prak
- grid.25879.310000 0004 1936 8972Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA ,grid.25879.310000 0004 1936 8972Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Thomas Brooks
- grid.25879.310000 0004 1936 8972Center for Neuromodulation in Depression and Stress (CNDS), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA ,grid.25879.310000 0004 1936 8972Institute for Translational Medicine and Therapeutics (ITMAT), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Walid Makhoul
- grid.25879.310000 0004 1936 8972Center for Neuromodulation in Depression and Stress (CNDS), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Joanne C. Beer
- grid.25879.310000 0004 1936 8972Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA USA
| | - Ling Zhao
- grid.25879.310000 0004 1936 8972Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA ,grid.25879.310000 0004 1936 8972Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Tommaso Girelli
- grid.25879.310000 0004 1936 8972Center for Neuromodulation in Depression and Stress (CNDS), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Carsten Skarke
- grid.25879.310000 0004 1936 8972Institute for Translational Medicine and Therapeutics (ITMAT), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA ,grid.25879.310000 0004 1936 8972Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Yvette I. Sheline
- grid.25879.310000 0004 1936 8972Center for Neuromodulation in Depression and Stress (CNDS), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA ,grid.25879.310000 0004 1936 8972Departments of Psychiatry, Radiology, Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| |
Collapse
|
252
|
Herrman H, Patel V, Kieling C, Berk M, Buchweitz C, Cuijpers P, Furukawa TA, Kessler RC, Kohrt BA, Maj M, McGorry P, Reynolds CF, Weissman MM, Chibanda D, Dowrick C, Howard LM, Hoven CW, Knapp M, Mayberg HS, Penninx BWJH, Xiao S, Trivedi M, Uher R, Vijayakumar L, Wolpert M. Time for united action on depression: a Lancet-World Psychiatric Association Commission. Lancet 2022; 399:957-1022. [PMID: 35180424 DOI: 10.1016/s0140-6736(21)02141-3] [Citation(s) in RCA: 474] [Impact Index Per Article: 158.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 09/15/2021] [Accepted: 09/21/2021] [Indexed: 12/12/2022]
Affiliation(s)
- Helen Herrman
- Orygen, The National Centre of Excellence in Youth Mental Health, Parkville, VIC, Australia; Centre for Youth Mental Health, The University of Melbourne, Parkville, VIC, Australia.
| | - Vikram Patel
- Department of Global Health and Social Medicine, Harvard Medical School, Boston, MA, USA; Sangath, Goa, India; Department of Global Health and Population, Harvard T H Chan School of Public Health, Boston, MA, USA
| | - Christian Kieling
- Department of Psychiatry, School of Medicine, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Child & Adolescent Psychiatry Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Michael Berk
- Deakin University, IMPACT Institute, Geelong, VIC, Australia
| | - Claudia Buchweitz
- Graduate Program in Psychiatry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Pim Cuijpers
- Department of Clinical, Neuro and Developmental Psychology, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Toshiaki A Furukawa
- Department of Health Promotion and Human Behavior, Kyoto University Graduate School of Medicine/School of Public Health, Kyoto, Japan
| | - Ronald C Kessler
- Department of Health Care Policy, Harvard Medical School, Boston, MA, USA
| | - Brandon A Kohrt
- Department of Psychiatry and Behavioral Sciences, George Washington University, Washington, DC, USA
| | - Mario Maj
- Department of Psychiatry, University of Campania L Vanvitelli, Naples, Italy
| | - Patrick McGorry
- Orygen, The National Centre of Excellence in Youth Mental Health, Parkville, VIC, Australia; Centre for Youth Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Charles F Reynolds
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Myrna M Weissman
- Columbia University Mailman School of Public Health, New York, NY, USA; Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA; New York State Psychiatric Institute, New York, NY, USA
| | - Dixon Chibanda
- Department of Psychiatry, University of Zimbabwe, Harare, Zimbabwe; Centre for Global Mental Health, The London School of Hygiene and Tropical Medicine, London, UK
| | - Christopher Dowrick
- Department of Primary Care and Mental Health, University of Liverpool, Liverpool, UK
| | - Louise M Howard
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Christina W Hoven
- Columbia University Mailman School of Public Health, New York, NY, USA; Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA; New York State Psychiatric Institute, New York, NY, USA
| | - Martin Knapp
- Care Policy and Evaluation Centre, London School of Economics and Political Science, London, UK
| | - Helen S Mayberg
- Departments of Neurology, Neurosurgery, Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brenda W J H Penninx
- Department of Psychiatry, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Shuiyuan Xiao
- Central South University Xiangya School of Public Health, Changsha, China
| | - Madhukar Trivedi
- Peter O'Donnell Jr Brain Institute and the Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Rudolf Uher
- Department of Psychiatry, Dalhousie University, Halifax, Canada
| | - Lakshmi Vijayakumar
- Sneha, Suicide Prevention Centre and Voluntary Health Services, Chennai, India
| | | |
Collapse
|
253
|
Archer M, Niemelä O, Hämäläinen M, Moilanen E, Leinonen E, Kampman O. The role of alcohol use and adiposity in serum levels of IL-1RA in depressed patients. BMC Psychiatry 2022; 22:158. [PMID: 35232419 PMCID: PMC8889691 DOI: 10.1186/s12888-022-03784-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 01/25/2022] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND The role of Interleukin-1 Receptor antagonist (IL-1Ra), an innate antagonist to pro-inflammatory cytokine IL-1, has attracted increasing attention due to its potential pathogenic and therapeutic implications in depression. However, the role of alcohol and adiposity in modulating IL-1Ra cytokine pathway in depressed patients has remainned unknown. The aim of this study was to follow the changes in IL-1Ra serum levels in depressed patients with or without simultaneous alcohol use disorder (AUD) and different degrees of adiposity during 6 months of follow-up. MATERIALS AND METHODS A total of 242 patients with depression were followed for 6 months. At baseline 99 patients had simultaneous AUD. Levels of serum IL-1Ra and common mediators of inflammation (IL-6, hs-CRP) were measured. Clinical assessments included Body Mass Index (BMI), Montgomery-Asberg Depression Rating Scale (MADRS) and Alcohol Use Disorders Identification Test (AUDIT) scores. RESULTS Significant reductions in clinical symptoms and IL-1Ra were observed during 6-month follow-up. In hierarchical linear regression analysis, the effect of MADRS score, age, gender, and smoking had a combined effect of 2.4% in the model. The effect of AUDIT score increased the effect to 4.2% of variance (p = 0.08), whereas adding BMI increased the effect to 18.5% (p < 0.001). CONCLUSION Adiposity may influence the IL-1Ra anti-inflammatory response in depressed patients, whereas the effect of alcohol consumption in these patients seems insignificant. These findings should be considered in studies on the role of IL-1Ra in depression. TRIAL REGISTRATION Ostrobothnia Depression Study in ClinicalTrials.gov , Identifier NCT02520271 .
Collapse
Affiliation(s)
- Mari Archer
- Tampere University, Faculty of Medicine and Health Technology, P.O. Box 100, 33014, Tampere, Finland.
| | - Onni Niemelä
- grid.415465.70000 0004 0391 502XDepartment of Laboratory Medicine and Medical Research Unit, Seinäjoki Central Hospital and Tampere University, 60220 Seinäjoki, Finland
| | - Mari Hämäläinen
- grid.412330.70000 0004 0628 2985The Immunopharmacology Research Group, Tampere University, Faculty of Medicine and Health Technology, and Tampere University Hospital, 33014 Tampere, Finland
| | - Eeva Moilanen
- grid.412330.70000 0004 0628 2985The Immunopharmacology Research Group, Tampere University, Faculty of Medicine and Health Technology, and Tampere University Hospital, 33014 Tampere, Finland
| | - Esa Leinonen
- grid.412330.70000 0004 0628 2985Tampere University, Faculty of Medicine and Health Technology, P.O. Box 100, 33014 University of Tampere, and Tampere University Hospital, 33014 Tampere, Finland
| | - Olli Kampman
- grid.502801.e0000 0001 2314 6254Tampere University, Faculty of Medicine and Health Technology, P.O. Box 100, 33014 University of Tampere and Pirkanmaa Hospital District, Department of Psychiatry, 33521 Tampere, Finland
| |
Collapse
|
254
|
Baysak E, Guden DS, Aricioglu F, Halaris A. C-reactive protein as a potential biomarker in psychiatric practice: Are we there yet? World J Biol Psychiatry 2022; 23:243-256. [PMID: 34323645 DOI: 10.1080/15622975.2021.1961502] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVES Serum or plasma levels of C-reactive protein (CRP) and high-sensitivity CRP (hsCRP) are widely used clinical markers of inflammation in other branches of medicine, whereas its clinical use in psychiatry has been limited to research studies. We aimed to assess the possibility of using CRP/hsCRP in psychiatric practice. This is a review and evaluation of various lines of evidence supporting the concept of CRP as a biomarker for psychiatric disorders in certain conditions. METHODS We searched the literature for studies which assessed CRP/hsCRP levels in various psychiatric disorders. RESULTS The accumulating evidence from large studies and meta-analyses allows us to understand the role of CRP in major psychiatric disorders and increase our understanding of specific symptoms and subtypes of disorders. CRP may be considered a 'psychiatric biomarker' which can alert clinicians about neuroinflammation, adverse effects of medications, cardiometabolic status, co-morbidities, and may also predict clinical outcomes and guide optimal treatment.selection. CONCLUSION Although the underlying pathophysiological role of CRP and hsCRP is still elusive and the association between CRP and psychiatric disorders is inconsistent, CRP holds promise to become a psychiatric biomarker.
Collapse
Affiliation(s)
- Erensu Baysak
- Department of Psychiatry, Marmara University School of Medicine, Istanbul, Turkey
| | - Demet Sinem Guden
- Department of Basic and Clinical Pharmacology, Istinye University Faculty of Medicine, Istanbul, Turkey
| | - Feyza Aricioglu
- Department of Pharmacology and Psychopharmacology Research Unit, Marmara University School of Pharmacy, Istanbul, Turkey
| | - Angelos Halaris
- Department of Psychiatry and Behavioral Neurosciences, Loyola University Chicago, Stritch School of Medicine, Loyola University Medical Center, Maywood, IL, USA
| |
Collapse
|
255
|
Yu Y, He X, Wang Y, Zhang J, Tang C, Rong P. Transcutaneous auricular vagal nerve stimulation inhibits limbic-regional P2X7R expression and reverses depressive-like behaviors in Zucker diabetic fatty rats. Neurosci Lett 2022; 775:136562. [PMID: 35245625 DOI: 10.1016/j.neulet.2022.136562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 02/25/2022] [Accepted: 02/26/2022] [Indexed: 11/29/2022]
Abstract
Zucker diabetic fatty (ZDF) rats develop type 2 diabetes (T2D) along with depressive-like behaviors. Transcutaneous auricular vagal nerve stimulation (taVNS) has antidiabetic and antidepressant-like effects in ZDF rats; however, the underlying antidepressant-like mechanisms are unclear. The purinergic receptor P2X7R, which is related to inflammation and depression, is upregulated in the limbic brain regions of depressed patients and rodents and is considered as a potential therapeutic target. Thus, this study aimed to provide preliminary evidence at the molecular level of taVNS antidepressant-like effect in ZDF rats through testing their limbic-regional P2X7R expression. ZDF rats were subjected to taVNS and transcutaneous non-vagal nerve stimulation (tnVNS). Body weight and blood glucose levels were monitored weekly. Depressive-like behaviors were evaluated with the open-field test (OFT) and forced swimming test (FST). Limbic-regional P2X7R expression was examined by western blotting (WB). P2X7R expressing cells were detected by immunohistochemistry (IHC). Compared to their lean littermates (ZL rats), ZDF rats developed obesity, hyperglycemia, and depressive-like behaviors with elevated limbic-regional P2X7R expression. taVNS but not tnVNS lowered body weight, reduced and stabilized blood glucose levels, suppressed limbic-regional P2X7R expression, and reversed the depressive-like behaviors. P2X7R was found primarily expressed in ZDF rats' limbic-regional astrocytes. In conclusion, taVNS inhibits ZDF rats' limbic-regional P2X7R expression, which may be one of the taVNS antidepressant-like mechanisms.
Collapse
Affiliation(s)
- Yutian Yu
- Acupuncture Department, Beijing Shijitan Hospital, Capital Medical University, Beijing, China; Ninth School of Clinical Medicine, Peking University, Beijing, China; Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Xun He
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yu Wang
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jinling Zhang
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chunzhi Tang
- Clinical Medical College of Acupuncture, Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Peijing Rong
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
256
|
Drevets WC, Wittenberg GM, Bullmore ET, Manji HK. Immune targets for therapeutic development in depression: towards precision medicine. Nat Rev Drug Discov 2022; 21:224-244. [PMID: 35039676 PMCID: PMC8763135 DOI: 10.1038/s41573-021-00368-1] [Citation(s) in RCA: 148] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/09/2021] [Indexed: 02/08/2023]
Abstract
Over the past two decades, compelling evidence has emerged indicating that immune mechanisms can contribute to the pathogenesis of major depressive disorder (MDD) and that drugs with primary immune targets can improve depressive symptoms. Patients with MDD are heterogeneous with respect to symptoms, treatment responses and biological correlates. Defining a narrower patient group based on biology could increase the treatment response rates in certain subgroups: a major advance in clinical psychiatry. For example, patients with MDD and elevated pro-inflammatory biomarkers are less likely to respond to conventional antidepressant drugs, but novel immune-based therapeutics could potentially address their unmet clinical needs. This article outlines a framework for developing drugs targeting a novel patient subtype within MDD and reviews the current state of neuroimmune drug development for mood disorders. We discuss evidence for a causal role of immune mechanisms in the pathogenesis of depression, together with targets under investigation in randomized controlled trials, biomarker evidence elucidating the link to neural mechanisms, biological and phenotypic patient selection strategies, and the unmet clinical need among patients with MDD.
Collapse
Affiliation(s)
- Wayne C Drevets
- Neuroscience, Janssen Research & Development, LLC, San Diego, CA, USA
| | | | - Edward T Bullmore
- Department of Psychiatry, University of Cambridge, Cambridge, UK
- Cambridgeshire & Peterborough NHS Foundation Trust, Cambridge, UK
| | | |
Collapse
|
257
|
Role of Inflammation in Traumatic Brain Injury-Associated Risk for Neuropsychiatric Disorders: State of the Evidence and Where Do We Go From Here. Biol Psychiatry 2022; 91:438-448. [PMID: 34955170 DOI: 10.1016/j.biopsych.2021.11.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 10/01/2021] [Accepted: 11/02/2021] [Indexed: 02/06/2023]
Abstract
In the past decade, there has been an increasing awareness that traumatic brain injury (TBI) and concussion substantially increase the risk for developing psychiatric disorders. Even mild TBI increases the risk for depression and anxiety disorders such as posttraumatic stress disorder by two- to threefold, predisposing patients to further functional impairment. This strong epidemiological link supports examination of potential mechanisms driving neuropsychiatric symptom development after TBI. One potential mechanism for increased neuropsychiatric symptoms after TBI is via inflammatory processes, as central nervous system inflammation can last years after initial injury. There is emerging preliminary evidence that TBI patients with posttraumatic stress disorder or depression exhibit increased central and peripheral inflammatory markers compared with TBI patients without these comorbidities. Growing evidence has demonstrated that immune signaling in animals plays an integral role in depressive- and anxiety-like behaviors after severe stress or brain injury. In this review, we will 1) discuss current evidence for chronic inflammation after TBI in the development of neuropsychiatric symptoms, 2) highlight potential microglial activation and cytokine signaling contributions, and 3) discuss potential promise and pitfalls for immune-targeted interventions and biomarker strategies to identify and treat TBI patients with immune-related neuropsychiatric symptoms.
Collapse
|
258
|
Oluboka OJ, Katzman MA, Habert J, Khullar A, Oakander MA, McIntosh D, McIntyre RS, Soares CN, Lam RW, Klassen LJ, Tanguay R. Early Optimized Pharmacological Treatment in Patients With Depression and Chronic Pain. CNS Spectr 2022; 28:1-40. [PMID: 35195060 DOI: 10.1017/s1092852922000128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
AbstractMajor depressive disorder (MDD) is the leading cause of disability worldwide. Patients with MDD have high rates of comorbidity with mental and physical conditions, one of which is chronic pain. Chronic pain conditions themselves are also associated with significant disability, and the large number of patients with MDD who have chronic pain drives high levels of disability and compounds healthcare burden. The management of depression in patients who also have chronic pain can be particularly challenging due to underlying mechanisms that are common to both conditions, and because many patients with these conditions are already taking multiple medications. For these reasons, healthcare providers may be reluctant to treat such patients. The Canadian Network for Mood and Anxiety Treatments (CANMAT) guidelines provide evidence-based recommendations for the management of MDD and comorbid psychiatric and medical conditions such as anxiety, substance use disorder, and cardiovascular disease; however, comorbid chronic pain is not addressed. In this article, we provide an overview of the pathophysiological and clinical overlap between depression and chronic pain and review evidence-based pharmacological recommendations in current treatment guidelines for MDD and for chronic pain. Based on clinical experience with MDD patients with comorbid pain, we recommend rapidly and aggressively treating depression according to CANMAT treatment guidelines, using antidepressant medications with analgesic properties, while addressing pain with first-line pharmacotherapy as treatment for depression is optimized. We review options for treating pain symptoms that remain after response to antidepressant treatment is achieved.
Collapse
|
259
|
Lai TL, Au CK, Chung HY, Lau CS. Depression in psoriatic arthritis: Related to socio-demographics, comorbid loads or disease activity? Int J Rheum Dis 2022; 25:474-480. [PMID: 35147299 DOI: 10.1111/1756-185x.14298] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 11/29/2022]
Abstract
AIM Psychological distress commonly occurs in patients with psoriatic arthritis (PsA). The primary objective of this study was to determine the prevalence of depression in PsA. The secondary objective was to explore its associated factors, including socio-demographics, disease activity data and comorbidities. METHODS Patients with PsA fulfilling the Classification Criteria for Psoriatic Arthritis were consecutively recruited from local rheumatology clinics. Depression was assessed by a self-administered Chinese-Cantonese version of the Hospital Anxiety and Depression Scale (HADS). RESULTS Two hundred and eight eligible patients with PsA were recruited, with 82 females and 126 males. Depression was found in 62 (29.8%) of them. The univariate model identified these associated factors: (1) Psoriasis Area and Severity Index score; (2) disease activity measurement, that is tender and swollen joint count, erythrocyte sedimentation rate, C-reactive protein, Disease Activity in Psoriatic Arthritis (DAPSA) score, Leeds Enthesitis Index and tender dactylitis count; (3) quality of life measurement, that is Health Assessment Questionnaire - Disability Index (HAQ-DI), pain and general health perception; (4) PsA duration; and (5) body mass index. The final regression model identified DAPSA and HAQ-DI were closely associated with depression, P = .007 and P = .02 respectively. Moderate and strong correlations with HADS score were found with DAPSA (Kendall's tau-b coefficient [τb] = 0.25) and HAQ-DI (τb = 0.4) respectively. No associations with depression were found between age, living and employment status, gender, demographics, inflammatory markers, disease duration, skin involvement and comorbidities, in term of Charlson's Comorbidity Index. CONCLUSION Depression was prevalent among PsA patients and it was closely correlated with disease activity and physical function impairment. Achieving low disease activity and maintaining physical function in patients with PsA may mitigate the psychological burden. The present study also highlighted the unmet needs of strategies to identify this common phenomenon.
Collapse
Affiliation(s)
- Tin Lok Lai
- Rheumatology Team, Department of Medicine, Tseung Kwan O Hospital, Hong Kong, Hong Kong
| | - Chi Kit Au
- Rheumatology Team, Department of Medicine, Tseung Kwan O Hospital, Hong Kong, Hong Kong
| | - Ho Yin Chung
- Rheumatology & Clinical Immunology Team, Department of Medicine, Queen Mary Hospital, Hong Kong, Hong Kong
| | - Chak Sing Lau
- Rheumatology & Clinical Immunology Team, Department of Medicine, Queen Mary Hospital, Hong Kong, Hong Kong
| |
Collapse
|
260
|
Burden of depression and anxiety among patients with inflammatory bowel disease: results of a nationwide analysis. Int J Colorectal Dis 2022; 37:313-321. [PMID: 34731298 DOI: 10.1007/s00384-021-04056-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/25/2021] [Indexed: 02/07/2023]
Abstract
PURPOSE The burden of psychiatric disorders is on a rise in inflammatory bowel disease (IBD) patients which has shown to effect medication compliance and overall clinical outcomes. We studied the prevalence of depression and anxiety in IBD patients when compared to individuals with other chronic medical conditions. METHODS This is a retrospective cohort study using the United States national inpatient sample of 2016 to 2018. We identified patient encounters with a diagnosis of IBD. Our primary outcome was prevalence of depression and anxiety in IBD patients when compared to general adult population with other chronic medical conditions. We further studied these outcomes in subgroups of patients with ulcerative colitis and Crohn's disease. RESULTS A total of 963,619 patient encounters were identified with the diagnosis of IBD between 2016 and 2018, of them 162,850 (16.9%) had depression and 201,685 (20.9%) had anxiety. The prevalence of depression and anxiety was significantly higher in IBD patients in comparison to general population, (16.9% vs 12.3%) and (20.9% vs 15%) respectively (p < 0.001). Association of depression and anxiety was also higher in IBD patients when compared to patients with other chronic conditions like diabetes, metastatic cancer, and coronary artery disease. Crohn's disease and ulcerative colitis were independently associated with increased odds of depression and anxiety and these results were statistically significant (p < 0.001). CONCLUSIONS IBD is associated with increased prevalence of depression and anxiety when compared to general population. Association of these psychiatric illnesses with IBD is significantly higher when compared to other chronic medical conditions.
Collapse
|
261
|
Rimmerman N, Verdiger H, Goldenberg H, Naggan L, Robinson E, Kozela E, Gelb S, Reshef R, Ryan KM, Ayoun L, Refaeli R, Ashkenazi E, Schottlender N, Ben Hemo-Cohen L, Pienica C, Aharonian M, Dinur E, Lazar K, McLoughlin DM, Zvi AB, Yirmiya R. Microglia and their LAG3 checkpoint underlie the antidepressant and neurogenesis-enhancing effects of electroconvulsive stimulation. Mol Psychiatry 2022; 27:1120-1135. [PMID: 34650207 DOI: 10.1038/s41380-021-01338-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 09/16/2021] [Accepted: 10/01/2021] [Indexed: 12/21/2022]
Abstract
Despite evidence implicating microglia in the etiology and pathophysiology of major depression, there is paucity of information regarding the contribution of microglia-dependent molecular pathways to antidepressant procedures. In this study, we investigated the role of microglia in a mouse model of depression (chronic unpredictable stress-CUS) and its reversal by electroconvulsive stimulation (ECS), by examining the effects of microglia depletion with the colony stimulating factor-1 antagonist PLX5622. Microglia depletion did not change basal behavioral measures or the responsiveness to CUS, but it completely abrogated the therapeutic effects of ECS on depressive-like behavior and neurogenesis impairment. Treatment with the microglia inhibitor minocycline concurrently with ECS also diminished the antidepressant and pro-neurogenesis effects of ECS. Hippocampal RNA-Seq analysis revealed that ECS significantly increased the expression of genes related to neurogenesis and dopamine signaling, while reducing the expression of several immune checkpoint genes, particularly lymphocyte-activating gene-3 (Lag3), which was the only microglial transcript significantly altered by ECS. None of these molecular changes occurred in microglia-depleted mice. Immunohistochemical analyses showed that ECS reversed the CUS-induced changes in microglial morphology and elevation in microglial LAG3 receptor expression. Consistently, either acute or chronic systemic administration of a LAG3 monoclonal antibody, which readily penetrated into the brain parenchyma and was found to serve as a direct checkpoint blocker in BV2 microglia cultures, rapidly rescued the CUS-induced microglial alterations, depressive-like symptoms, and neurogenesis impairment. These findings suggest that brain microglial LAG3 represents a promising target for novel antidepressant therapeutics.
Collapse
Affiliation(s)
- Neta Rimmerman
- Department of Psychology, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Hodaya Verdiger
- Department of Psychology, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Hagar Goldenberg
- Department of Psychology, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Lior Naggan
- Department of Psychology, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Elad Robinson
- Department of Psychology, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ewa Kozela
- Department of Psychology, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Sivan Gelb
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ronen Reshef
- Department of Psychology, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Karen M Ryan
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland.,Department of Psychiatry, Trinity College Dublin, St. Patrick's University Hospital, James Street, Dublin, Ireland
| | - Lily Ayoun
- Department of Psychology, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ron Refaeli
- Edmond & Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Einat Ashkenazi
- Department of Psychology, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nofar Schottlender
- Department of Psychology, The Hebrew University of Jerusalem, Jerusalem, Israel
| | | | - Claudia Pienica
- Department of Psychology, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Maayan Aharonian
- Department of Psychology, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Eyal Dinur
- Department of Psychology, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Koby Lazar
- Department of Psychology, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Declan M McLoughlin
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland.,Department of Psychiatry, Trinity College Dublin, St. Patrick's University Hospital, James Street, Dublin, Ireland
| | - Ayal Ben Zvi
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Raz Yirmiya
- Department of Psychology, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
262
|
Hoang D, Xu Y, Lutz O, Bannai D, Zeng V, Bishop JR, Keshavan M, Lizano P. Inflammatory Subtypes in Antipsychotic-Naïve First-Episode Schizophrenia are Associated with Altered Brain Morphology and Topological Organization. Brain Behav Immun 2022; 100:297-308. [PMID: 34875344 PMCID: PMC8767408 DOI: 10.1016/j.bbi.2021.11.019] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 11/01/2021] [Accepted: 11/26/2021] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Peripheral inflammation is implicated in schizophrenia, however, not all individuals demonstrate inflammatory alterations. Recent studies identified inflammatory subtypes in chronic psychosis with high inflammation having worse cognitive performance and displaying neuroanatomical enlargement compared to low inflammation subtypes. It is unclear if inflammatory subtypes exist earlier in the disease course, thus, we aim to identify inflammatory subtypes in antipsychotic naïve First-Episode Schizophrenia (FES). METHODS 12 peripheral inflammatory markers, clinical, cognitive, and neuroanatomical measures were collected from a naturalistic study of antipsychotic-naïve FES patients. A combination of unsupervised principal component analysis and hierarchical clustering was used to categorize inflammatory subtypes from their cytokine data (17 FES High, 30 FES Low, and 33 healthy controls (HCs)). Linear regression analysis was used to assess subtype differences. Neuroanatomical correlations with clinical and cognitive measures were performed using partial Spearman correlations. Graph theoretical analyses were performed to assess global and local network properties across inflammatory subtypes. RESULTS The FES High group made up 36% of the FES group and demonstrated significantly greater levels of IL1β, IL6, IL8, and TNFα compared to FES Low, and higher levels of IL1β and IL8 compared to HCs. FES High had greater right parahippocampal, caudal anterior cingulate, and bank superior sulcus thicknesses compared to FES Low. Compared to HCs, FES Low showed smaller bilateral amygdala volumes and widespread cortical thickness. FES High and FES Low groups demonstrated less efficient topological organization compared to HCs. Individual cytokines and/or inflammatory signatures were positively associated with cognition and symptom measures. CONCLUSIONS Inflammatory subtypes are present in antipsychotic-naïve FES and are associated with inflammation-mediated cortical expansion. These findings support our previous findings in chronic psychosis and point towards a connection between inflammation and blood-brain barrier disruption. Thus, identifying inflammatory subtypes may provide a novel therapeutic avenue for biomarker-guided treatment involving anti-inflammatory medications.
Collapse
Affiliation(s)
- Dung Hoang
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Yanxun Xu
- Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, MD, USA
| | - Olivia Lutz
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Deepthi Bannai
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Victor Zeng
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Jeffrey R Bishop
- Department of Experimental and Clinical Pharmacology and Psychiatry, University of Minnesota, Minneapolis, MN, USA
| | - Matcheri Keshavan
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Paulo Lizano
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
263
|
Treatment-Resistant Depression with Anhedonia: Integrating Clinical and Preclinical Approaches to Investigate Distinct Phenotypes. Neurosci Biobehav Rev 2022; 136:104578. [DOI: 10.1016/j.neubiorev.2022.104578] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 11/30/2021] [Accepted: 02/11/2022] [Indexed: 12/21/2022]
|
264
|
Ho TC, Kulla A, Teresi GI, Sisk LM, Rosenberg-Hasson Y, Maecker HT, Gotlib IH. Inflammatory cytokines and callosal white matter microstructure in adolescents. Brain Behav Immun 2022; 100:321-331. [PMID: 34896593 PMCID: PMC11849988 DOI: 10.1016/j.bbi.2021.12.003] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 11/29/2021] [Accepted: 12/04/2021] [Indexed: 12/15/2022] Open
Abstract
Adolescent depression is characterized by heightened inflammation and altered connectivity of fronto-cingulate-limbic tracts, including the genu of the corpus callosum (CCG) and the uncinate fasciculus (UF). No studies, however, have yet examined the association between inflammation, measured by peripheral levels of cytokines, and white matter connectivity of fronto-cingulate-limbic tracts in adolescents. Here, 56 depressed adolescents (32 females, 3 non-binary; 16.23 ± 1.28 years) and 19 controls (10 females; 15.72 ± 1.17 years) completed a diffusion-weighted MRI scan at 3 Tesla. We conducted deterministic tractography to segment bilateral corpus callosum (genu and splenium) and UF and computed mean fractional anisotropy (FA) in each tract. A subset of participants (43 depressed and 17 healthy controls) also provided dried blood spot samples from which we assayed interleukin 6 (IL-6) and tumor necrosis factor alpha (TNF-ɑ) using a Luminex multiplex array. Depressed participants did not differ from controls in FA of the corpus callosum or UF (all FDR-corrected ps > 0.056) but exhibited higher levels of inflammation than did controls (IL-6: β = 0.91, FDR-corrected p = 0.006; TNF-α: β = 0.76, FDR-corrected p = 0.006). Although diagnostic group did not moderate the associations between inflammatory cytokines and FA in the CCG and UF, across both groups, greater peripheral inflammation was associated with lower FA in the CCG (IL-6: β = -0.38; FDR-corrected p = 0.044; TNF-ɑ: β = -0.41, FDR-corrected p = 0.044). This study is the first to examine associations between peripheral inflammation and white matter microstructure of fronto-cingulate-limbic tracts in depressed and nondepressed adolescents. Future mechanistic studies are needed to confirm our findings; nevertheless, our results suggest that heightened inflammation is an important component of neurophenotypes that are relevant to adolescent depression.
Collapse
Affiliation(s)
- Tiffany C Ho
- Department of Psychiatry & Behavioral Sciences, Weill Institute of Neurosciences, University of California, San Francisco, San Francisco, CA, United States.
| | - Artenisa Kulla
- Department of Psychology, Stanford University, Stanford, CA, United States; College of Medicine, University of Florida, Gainesville, FL, United States
| | - Giana I Teresi
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Lucinda M Sisk
- Department of Psychology, Yale University, New Haven, CT, United States
| | - Yael Rosenberg-Hasson
- Department of Psychology, Yale University, New Haven, CT, United States; Department of Microbiology and Immunology, Human Immune Monitoring Center, Stanford University, Stanford, CA, United States
| | - Holden T Maecker
- Department of Psychology, Yale University, New Haven, CT, United States; Department of Microbiology and Immunology, Human Immune Monitoring Center, Stanford University, Stanford, CA, United States
| | - Ian H Gotlib
- Department of Psychology, Stanford University, Stanford, CA, United States
| |
Collapse
|
265
|
C-Reactive Protein as a Biomarker for Major Depressive Disorder? Int J Mol Sci 2022; 23:ijms23031616. [PMID: 35163538 PMCID: PMC8836046 DOI: 10.3390/ijms23031616] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/25/2022] [Accepted: 01/28/2022] [Indexed: 11/16/2022] Open
Abstract
The etiopathogenesis of depression is not entirely understood. Several studies have investigated the role of inflammation in major depressive disorder. The present work aims to review the literature on the association between C-Reactive Protein (CRP) and depression. A systematic review was performed for the topics of ‘CRP’ and ‘depression’ using the PubMed database from inception to December 2021. Fifty-six studies were identified and included in the review. Evidence suggested the presence of dysregulation in the inflammation system in individuals with depression. In most studies, higher blood CRP levels were associated with greater symptom severity, a specific pattern of depressive symptoms, and a worse response to treatment. Moreover, about one-third of depressed patients showed a low-grade inflammatory state, suggesting the presence of a different major depressive disorder (MDD) subgroup with a distinct etiopathogenesis, clinical course, treatment response, and prognosis, which could benefit from monitoring of CRP levels and might potentially respond to anti-inflammatory treatments. This work provides robust evidence about the potential role of CRP and its blood levels in depressive disorders. These findings can be relevant to developing new therapeutic strategies and better understanding if CRP may be considered a valuable biomarker for depression.
Collapse
|
266
|
Porter GA, O’Connor JC. Brain-derived neurotrophic factor and inflammation in depression: Pathogenic partners in crime? World J Psychiatry 2022; 12:77-97. [PMID: 35111580 PMCID: PMC8783167 DOI: 10.5498/wjp.v12.i1.77] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/21/2021] [Accepted: 12/02/2021] [Indexed: 02/06/2023] Open
Abstract
Major depressive disorder is a debilitating disorder affecting millions of people each year. Brain-derived neurotrophic factor (BDNF) and inflammation are two prominent biologic risk factors in the pathogenesis of depression that have received considerable attention. Many clinical and animal studies have highlighted associations between low levels of BDNF or high levels of inflammatory markers and the development of behavioral symptoms of depression. However, less is known about potential interaction between BDNF and inflammation, particularly within the central nervous system. Emerging evidence suggests that there is bidirectional regulation between these factors with important implications for the development of depressive symptoms and anti-depressant response. Elevated levels of inflammatory mediators have been shown to reduce expression of BDNF, and BDNF may play an important negative regulatory role on inflammation within the brain. Understanding this interaction more fully within the context of neuropsychiatric disease is important for both developing a fuller understanding of biological pathogenesis of depression and for identifying novel therapeutic opportunities. Here we review these two prominent risk factors for depression with a particular focus on pathogenic implications of their interaction.
Collapse
Affiliation(s)
- Grace A Porter
- Department of Pharmacology, UT Health San Antonio, San Antonio, TX 78229, United States
| | - Jason C O’Connor
- Department of Pharmacology, University of Texas Health San Antonio, San Antonio, TX 78229, United States
- Audie L. Murphy VA Hospital, South Texas Veterans Health System, San Antonio, TX 78229, United States
| |
Collapse
|
267
|
Chen X, Chen Y, Qi D, Cui D. Multifaceted interconnections between macrophage migration inhibitory factor and psychiatric disorders. Prog Neuropsychopharmacol Biol Psychiatry 2022; 112:110422. [PMID: 34358623 DOI: 10.1016/j.pnpbp.2021.110422] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 07/30/2021] [Accepted: 07/31/2021] [Indexed: 01/02/2023]
Abstract
Inflammation is involved in the pathogenesis of psychiatric disorders. Many previous studies have defined the important roles of inflammatory factors in the pathogenesis, diagnosis, and treatment outcomes of psychiatric disorders. Macrophage migration inhibitory factor (MIF), a pro-inflammatory factor, has been gradually recognized to be involved in the development of neurological diseases in recent years. Our current review focuses on discussing the potential beneficial and detrimental roles of MIF in psychiatric disorders. We will provide new mechanistic insights for the development of potential diagnostic and therapeutic biomarkers based on MIF for psychiatric diseases.
Collapse
Affiliation(s)
- Xi Chen
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Psychotic Disorders, Shanghai, China.
| | - Yifan Chen
- Department of Psychology, Tufts University, Medford, MA, USA.
| | - Dake Qi
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada.
| | - Donghong Cui
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Psychotic Disorders, Shanghai, China; Brain Science and Technology Research Center, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
268
|
Cathomas F, Bevilacqua L, Ramakrishnan A, Kronman H, Costi S, Schneider M, Chan KL, Li L, Nestler EJ, Shen L, Charney DS, Russo SJ, Murrough JW. Whole blood transcriptional signatures associated with rapid antidepressant response to ketamine in patients with treatment resistant depression. Transl Psychiatry 2022; 12:12. [PMID: 35013133 PMCID: PMC8748646 DOI: 10.1038/s41398-021-01712-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/22/2021] [Accepted: 10/22/2021] [Indexed: 12/15/2022] Open
Abstract
Ketamine has rapid and sustained antidepressant effects in patients with treatment-resistant depression (TRD). However, the underlying mechanisms of action are not well understood. There is increasing evidence that TRD is associated with a pro-inflammatory state and that ketamine may inhibit inflammatory processes. We thus investigated whole blood transcriptional profiles related to TRD and gene expression changes associated with treatment response to ketamine. Whole blood was collected at baseline (21 healthy controls [HC], 26 patients with TRD) and then again in patients with TRD 24 hours following a single intravenous infusion of ketamine (0.5 mg/kg). We performed RNA-sequencing and analyzed (a) baseline transcriptional profiles between patients with TRD and HC, (b) responders vs. non-responders before ketamine treatment, and (c) gene expression signatures associated with clinical improvement. At baseline, patients with TRD compared to HC showed a gene expression signature indicative of interferon signaling pathway activation. Prior to ketamine administration, the metabotropic glutamate receptor gene GRM2 and the ionotropic glutamate receptor gene GRIN2D were upregulated in responders compared to non-responders. Response to ketamine was associated with a distinct transcriptional signature, however, we did not observe gene expression changes indicative of an anti-inflammatory effect. Future studies are needed to determine the role of the peripheral immune system in the antidepressant effect of ketamine.
Collapse
Affiliation(s)
- Flurin Cathomas
- grid.59734.3c0000 0001 0670 2351Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Laura Bevilacqua
- grid.59734.3c0000 0001 0670 2351Depression and Anxiety Center for Discovery and Treatment, Department of Psychiatry, Icahn School of Medicine of Mount Sinai, New York, NY 10029 USA
| | - Aarthi Ramakrishnan
- grid.59734.3c0000 0001 0670 2351Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Hope Kronman
- grid.59734.3c0000 0001 0670 2351Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Sara Costi
- grid.59734.3c0000 0001 0670 2351Depression and Anxiety Center for Discovery and Treatment, Department of Psychiatry, Icahn School of Medicine of Mount Sinai, New York, NY 10029 USA
| | - Molly Schneider
- grid.59734.3c0000 0001 0670 2351Depression and Anxiety Center for Discovery and Treatment, Department of Psychiatry, Icahn School of Medicine of Mount Sinai, New York, NY 10029 USA
| | - Kenny L. Chan
- grid.59734.3c0000 0001 0670 2351Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Long Li
- grid.59734.3c0000 0001 0670 2351Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Eric J. Nestler
- grid.59734.3c0000 0001 0670 2351Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Li Shen
- grid.59734.3c0000 0001 0670 2351Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Dennis S. Charney
- grid.59734.3c0000 0001 0670 2351Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA ,grid.59734.3c0000 0001 0670 2351Depression and Anxiety Center for Discovery and Treatment, Department of Psychiatry, Icahn School of Medicine of Mount Sinai, New York, NY 10029 USA ,grid.59734.3c0000 0001 0670 2351Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Scott J. Russo
- grid.59734.3c0000 0001 0670 2351Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - James W. Murrough
- grid.59734.3c0000 0001 0670 2351Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA ,grid.59734.3c0000 0001 0670 2351Depression and Anxiety Center for Discovery and Treatment, Department of Psychiatry, Icahn School of Medicine of Mount Sinai, New York, NY 10029 USA
| |
Collapse
|
269
|
The Brain-Skin Axis in Psoriasis-Psychological, Psychiatric, Hormonal, and Dermatological Aspects. Int J Mol Sci 2022; 23:ijms23020669. [PMID: 35054853 PMCID: PMC8776235 DOI: 10.3390/ijms23020669] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/05/2022] [Accepted: 01/06/2022] [Indexed: 02/04/2023] Open
Abstract
Psoriasis is a chronic inflammatory skin disease with systemic manifestation, in which psychological factors play an important role. The etiology of psoriasis is complex and multifactorial, including genetic background and environmental factors such as emotional or physical stress. Psychological stress may also play a role in exacerbation of psoriasis, by dysregulation of the hypothalamic–pituitary–adrenal (HPA) axis, sympathetic–adrenal–medullary axis, peripheral nervous system, and immune system. Skin cells also express various neuropeptides and hormones in response to stress, including the fully functional analog of the HPA axis. The deterioration of psoriatic lesions is accompanied by increased production of inflammatory mediators, which could contribute to the imbalance of neurotransmitters and the development of symptoms of depression and anxiety. Therefore, deregulation of the crosstalk between endocrine, paracrine, and autocrine stress signaling pathways contributes to clinical manifestations of psoriasis, which requires multidisciplinary approaches.
Collapse
|
270
|
Nazir S, Farooq RK, Nasir S, Hanif R, Javed A. Therapeutic effect of Thymoquinone on behavioural response to UCMS and neuroinflammation in hippocampus and amygdala in BALB/c mice model. Psychopharmacology (Berl) 2022; 239:47-58. [PMID: 35029704 DOI: 10.1007/s00213-021-06038-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 11/26/2021] [Indexed: 11/29/2022]
Abstract
RATIONALE Major depressive disorder is the leading cause of disability worldwide. The corticolimbic system plays a critical role in the emotional and cognitive aspects of major depressive disorder. Owing to the unsatisfactory efficacy of conventional antidepressants, there is a need to explore novel therapies. OBJECTIVES The current study aimed to explore the antidepressant potential of thymoquinone, a natural compound with anti-inflammatory activity, and propose its underlying mechanism of action in the unpredictable chronic mild stress (UCMS) mouse model. METHODS Coat state, forced swim test, elevated plus maze test, novelty suppressed feeding test and social interaction test were performed to quantify the behavioural shift induced by UCMS and the effect of thymoquinone and fluoxetine treatment. In addition, messenger RNA (mRNA) expression levels of inflammatory cytokines (IL-1β, IL-6 and TNF-α) and BDNF and NeuN were analysed by a quantitative real-time polymerase chain reaction in the hippocampus and amygdala of experimental and control groups. RESULTS UCMS significantly deteriorated coat state. Thymoquinone reinstated the resignation behaviour and latency to feed affected by UCMS. UCMS induced an increase in inflammatory cytokines (IL-1β, IL-6 and TNF-α) in the hippocampus and amygdala, which was decreased by thymoquinone. UCMS caused an increase in BDNF and NeuN mRNA levels in the amygdala while a decrease in the hippocampus. This opposite effect on BDNF was also compensated by thymoquinone; however, thymoquinone did not significantly change Ki67 and NeuN mRNA levels in the hippocampus. CONCLUSIONS Thymoquinone restored the behavioural changes induced by UCMS. In addition, the antidepressant effect of thymoquinone is in line with changes in inflammatory parameters and changes in BDNF in the hippocampus and amygdala.
Collapse
Affiliation(s)
- Sadia Nazir
- Atta-Ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12 Campus, Islamabad, 44000, Pakistan
| | - Rai Khalid Farooq
- Department of Neuroscience Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam, 31441, Saudi Arabia
| | - Sadia Nasir
- Atta-Ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12 Campus, Islamabad, 44000, Pakistan
| | - Rumeza Hanif
- Atta-Ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12 Campus, Islamabad, 44000, Pakistan
| | - Aneela Javed
- Atta-Ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12 Campus, Islamabad, 44000, Pakistan.
| |
Collapse
|
271
|
Chen PS, Tang LY, Chang HH. Roles of C-reactive protein polymorphisms and life event changes on cognitive function in bipolar patients receiving valproate. Int J Immunopathol Pharmacol 2022; 36:3946320221084835. [PMID: 35377256 PMCID: PMC8984865 DOI: 10.1177/03946320221084835] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Introduction Patients with bipolar disorder (BD) exhibit an inflamed condition that is
associated with metabolic disturbance and cognitive impairment. Whether
inflammation, represented by C-reactive protein (CRP), is causally
associated with BD and influences treatment outcome has not been
established. Methods We examined whether CRP is a causal factor for the risk of BD in drug-naïve,
depressed BD patients and investigated whether polymorphisms in
CRP and life event changes influence cognitive function
in BD patients receiving valproate (VPA) treatment. Results Our results showed that BD patients had significantly higher CRP levels and
worse cognitive function than the controls, while the frequencies of
CRP single nucleotide polymorphisms in BD patients and
in controls were not different. In addition, the life event scale score was
higher for BD patients than for controls. Furthermore, the genotypes of
CRP polymorphisms and the interactions between
polymorphisms of CRP and life event scale score had a
significant influence on cognitive performance in BD patients after 12 weeks
of VPA treatment. Conclusion Our study demonstrated the clinical utility of the application of functional
genetics in clarifying the interactions among CRP, life event stress, and BD
and suggested the important roles of CRP gene–environment
interactions in developing treatment strategies for BD.
Collapse
Affiliation(s)
- Po See Chen
- Department of Psychiatry, National Cheng Kung University Hospital, College of Medicine, 38026National Cheng Kung University, Tainan, Taiwan.,Institute of Behavioral Medicine, College of Medicine, 38026National Cheng Kung University, Tainan, Taiwan
| | - Li-Yi Tang
- Institute of Clinical Pharmacy and Pharmaceutical Sciences, College of Medicine, 38026National Cheng Kung University, Tainan, Taiwan
| | - Hui Hua Chang
- Institute of Clinical Pharmacy and Pharmaceutical Sciences, College of Medicine, 38026National Cheng Kung University, Tainan, Taiwan.,School of Pharmacy, College of Medicine, 38026National Cheng Kung University, Tainan, Taiwan.,Department of Pharmacy, National Cheng Kung University Hospital, College of Medicine, 38026National Cheng Kung University, Tainan, Taiwan.,Department of Pharmacy, National Cheng Kung University Hospital, Dou-Liou Branch, Yunlin, Taiwan
| |
Collapse
|
272
|
Gao W, Xu Y, Liang J, Sun Y, Zhang Y, Shan F, Ge J, Xia Q. Comparison of serum cytokines levels in normal-weight and overweight patients with first-episode drug-naïve major depressive disorder. Front Endocrinol (Lausanne) 2022; 13:1048337. [PMID: 36387880 PMCID: PMC9647627 DOI: 10.3389/fendo.2022.1048337] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 10/18/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE Abnormal levels of blood cytokines have been demonstrated to be associated with both excess weight and major depressive disorder (MDD). However, few studies have addressed the direct effect of body mass index (BMI) on basal serum cytokines in individuals with first-episode drug-naïve MDD. METHODS A total of 49 patients with first-episode drug-naïve MDD were categorized into normal weight (18.5 ≤ BMI < 25 kg/m2) and overweight (25 ≤ BMI < 30 kg/m2) groups according to WHO-criteria. The severity of depressive symptoms was assessed using the 24-items Hamilton Depression Scale (HAMD-24). A total of 37 cytokines were measured using Multiplex Luminex Assays. The scores of HAMD-24 and the levels of serum cytokines between normal weight group and overweight group were compared. Multiple linear regression analysis was performed to evaluate the association between abnormal serum cytokines levels and group after adjusting for HAMD-24 scores. The correlation between BMI and the scores of HAMD-24 and the levels of serum cytokines was evaluated using Pearson correlation analysis. RESULTS The scores of HAMD-24 in overweight group were significantly higher than normal weight group (t = -2.930, P = 0.005). Moreover, the levels of IL-1α, IL-1RA, IL-3, CXCL10, TNF-α, and ICAM-1 in overweight patients with MDD were significantly higher than those in normal-weight patients with MDD (all P < 0.05). Furthermore, after adjustment for HAMD-24 scores, there was a significant correlation between abnormal serum cytokines levels (IL-1α, IL-1RA, IL-3, CXCL10, TNF-α, and ICAM-1) and group (all P < 0.05). Additionally, BMI was positively correlated to the serum levels of IL-1α (r = 0.428, P = 0.002), IL-3 (r = 0.529, P < 0.001), IL-6 (r = 0.285, P = 0.050), IL-10 (r = 0.423, P = 0.003), IL-12 (r = 0.367, P = 0.010), IL-15 (r = 0.300, P = 0.036), CXCL10 (r = 0.316, P = 0.030), TNF-α (r = 0.338, P = 0.021), and ICAM-1 (r = 0.440, P = 0.002) in MDD patients. CONCLUSIONS These results provide direct evidence, probably for the first time, that overweight may be associated with several serum cytokines in patients with first-episode drug-naïve MDD. The underlying mechanisms are unclear and require further investigation.
Collapse
Affiliation(s)
- Wenfan Gao
- Affiliated Psychological Hospital of Anhui Medical University, Hefei, China
- Department of Pharmacy, Hefei Fourth People’s Hospital, Hefei, China
- Psychopharmacology Research Laboratory, Anhui Mental Health Center, Hefei, China
- Department of Science and Education, Anhui Clinical Research Center for Mental Disorders, Hefei, China
| | - Yayun Xu
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
- The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China
| | - Jun Liang
- Affiliated Psychological Hospital of Anhui Medical University, Hefei, China
- Department of Pharmacy, Hefei Fourth People’s Hospital, Hefei, China
- Psychopharmacology Research Laboratory, Anhui Mental Health Center, Hefei, China
- Department of Science and Education, Anhui Clinical Research Center for Mental Disorders, Hefei, China
| | - Yanhong Sun
- Affiliated Psychological Hospital of Anhui Medical University, Hefei, China
- Department of Pharmacy, Hefei Fourth People’s Hospital, Hefei, China
- Psychopharmacology Research Laboratory, Anhui Mental Health Center, Hefei, China
- Department of Science and Education, Anhui Clinical Research Center for Mental Disorders, Hefei, China
| | - Yuanyuan Zhang
- Affiliated Psychological Hospital of Anhui Medical University, Hefei, China
- Department of Pharmacy, Hefei Fourth People’s Hospital, Hefei, China
- Psychopharmacology Research Laboratory, Anhui Mental Health Center, Hefei, China
- Department of Science and Education, Anhui Clinical Research Center for Mental Disorders, Hefei, China
| | - Feng Shan
- Affiliated Psychological Hospital of Anhui Medical University, Hefei, China
- Department of Pharmacy, Hefei Fourth People’s Hospital, Hefei, China
- Psychopharmacology Research Laboratory, Anhui Mental Health Center, Hefei, China
- Department of Science and Education, Anhui Clinical Research Center for Mental Disorders, Hefei, China
| | - Jinfang Ge
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, China
- The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China
- School of Pharmacy, Anhui Medical University, Hefei, China
- *Correspondence: Jinfang Ge, ; Qingrong Xia,
| | - Qingrong Xia
- Affiliated Psychological Hospital of Anhui Medical University, Hefei, China
- Department of Pharmacy, Hefei Fourth People’s Hospital, Hefei, China
- Psychopharmacology Research Laboratory, Anhui Mental Health Center, Hefei, China
- Department of Science and Education, Anhui Clinical Research Center for Mental Disorders, Hefei, China
- *Correspondence: Jinfang Ge, ; Qingrong Xia,
| |
Collapse
|
273
|
Brás JP, Guillot de Suduiraut I, Zanoletti O, Monari S, Meijer M, Grosse J, Barbosa MA, Santos SG, Sandi C, Almeida MI. Stress-induced depressive-like behavior in male rats is associated with microglial activation and inflammation dysregulation in the hippocampus in adulthood. Brain Behav Immun 2022; 99:397-408. [PMID: 34793941 DOI: 10.1016/j.bbi.2021.10.018] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 10/22/2021] [Accepted: 10/31/2021] [Indexed: 12/27/2022] Open
Abstract
Neuroinflammation is increasingly recognized as playing a critical role in depression. Early-life stress exposure and constitutive differences in glucocorticoid responsiveness to stressors are two key risk factors for depression, but their impacts on the inflammatory status of the brain is still uncertain. Moreover, there is a need to identify specific molecules involved in these processes with the potential to be used as alternative therapeutic targets in inflammation-related depression. Here, we studied how peripubertal stress (PPS) combined with differential corticosterone (CORT)-stress responsiveness (CSR) influences depressive-like behaviors and brain inflammatory markers in male rats in adulthood, and how these alterations relate to microglia activation and miR-342 expression. We found that high-CORT stress-responsive (H-CSR) male rats that underwent PPS exhibited increased anhedonia and passive coping responses in adulthood. Also, animals exposed to PPS showed increased hippocampal TNF-α expression, which positively correlated with passive coping responses. In addition, PPS caused long-term effects on hippocampal microglia, particularly in H-CSR rats, with increased hippocampal IBA-1 expression and morphological alterations compatible with a higher degree of activation. H-CSR animals also showed upregulation of hippocampal miR-342, a mediator of TNF-α-driven microglial activation, and its expression was positively correlated with TNF-α expression, microglial activation and passive coping responses. Our findings indicate that individuals with constitutive H-CSR are particularly sensitive to developing protracted depression-like behaviors following PPS exposure. In addition, they show neuro-immunological alterations in adulthood, such as increased hippocampal TNF-α expression, microglial activation and miR-342 expression. Our work highlights miR-342 as a potential therapeutic target in inflammation-related depression.
Collapse
Affiliation(s)
- João Paulo Brás
- Instituto de Investigação e Inovação em Saúde/Instituto de Engenharia Biomédica (i3S/INEB), University of Porto (UP), Rua Alfredo Allen 208, 4200-135 Porto, Portugal; Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto (UP), Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal; Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Station 19, CH-1015 Lausanne, Switzerland
| | | | - Olivia Zanoletti
- Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Station 19, CH-1015 Lausanne, Switzerland
| | - Silvia Monari
- Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Station 19, CH-1015 Lausanne, Switzerland
| | - Mandy Meijer
- Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Station 19, CH-1015 Lausanne, Switzerland; Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, Netherlands
| | - Jocelyn Grosse
- Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Station 19, CH-1015 Lausanne, Switzerland
| | - Mário Adolfo Barbosa
- Instituto de Investigação e Inovação em Saúde/Instituto de Engenharia Biomédica (i3S/INEB), University of Porto (UP), Rua Alfredo Allen 208, 4200-135 Porto, Portugal; Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto (UP), Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Susana Gomes Santos
- Instituto de Investigação e Inovação em Saúde/Instituto de Engenharia Biomédica (i3S/INEB), University of Porto (UP), Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Carmen Sandi
- Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Station 19, CH-1015 Lausanne, Switzerland
| | - Maria Inês Almeida
- Instituto de Investigação e Inovação em Saúde/Instituto de Engenharia Biomédica (i3S/INEB), University of Porto (UP), Rua Alfredo Allen 208, 4200-135 Porto, Portugal; Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto (UP), Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal.
| |
Collapse
|
274
|
Abstract
In the field of neuropsychiatry, neuroinflammation is one of the prevailing hypotheses to explain the pathophysiology of mood and psychotic disorders. Neuroinflammation encompasses an ill-defined set of pathophysiological processes in the central nervous system that cause neuronal or glial atrophy or death and disruptions in neurotransmitter signaling, resulting in cognitive and behavioral changes. Positron emission tomography for the brain-based translocator protein has been shown to be a useful tool to measure glial activation in neuropsychiatric disorders. Recent neuroimaging studies also indicate a potential disruption in the choroid plexus and blood-brain barrier, which modulate the transfer of ions, molecules, toxins, and cells from the periphery into the brain. Simultaneously, peripheral inflammatory markers have consistently been shown to be altered in mood and psychotic disorders. The crosstalk (i.e., the communication between peripheral and central inflammatory pathways) is not well understood in these disorders, however, and neuroimaging studies hold promise to shed light on this complex process. In the current Perspectives article, we discuss the neuroimaging insights into neuroimmune crosstalk offered in selected works. Overall, evidence exists for peripheral immune cell infiltration into the central nervous system in some patients, but the reason for this is unknown. Future neuroimaging studies should aim to extend our knowledge of this system and the role it likely plays in symptom onset and recurrence.
Collapse
|
275
|
Influences of dopaminergic system dysfunction on late-life depression. Mol Psychiatry 2022; 27:180-191. [PMID: 34404915 PMCID: PMC8850529 DOI: 10.1038/s41380-021-01265-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/28/2021] [Accepted: 08/04/2021] [Indexed: 12/15/2022]
Abstract
Deficits in cognition, reward processing, and motor function are clinical features relevant to both aging and depression. Individuals with late-life depression often show impairment across these domains, all of which are moderated by the functioning of dopaminergic circuits. As dopaminergic function declines with normal aging and increased inflammatory burden, the role of dopamine may be particularly salient for late-life depression. We review the literature examining the role of dopamine in the pathogenesis of depression, as well as how dopamine function changes with aging and is influenced by inflammation. Applying a Research Domain Criteria (RDoC) Initiative perspective, we then review work examining how dopaminergic signaling affects these domains, specifically focusing on Cognitive, Positive Valence, and Sensorimotor Systems. We propose a unified model incorporating the effects of aging and low-grade inflammation on dopaminergic functioning, with a resulting negative effect on cognition, reward processing, and motor function. Interplay between these systems may influence development of a depressive phenotype, with an initial deficit in one domain reinforcing decline in others. This model extends RDoC concepts into late-life depression while also providing opportunities for novel and personalized interventions.
Collapse
|
276
|
Jolink TA, Fendinger NJ, Alvarez GM, Feldman MJ, Gaudier-Diaz MM, Muscatell KA. Inflammatory reactivity to the influenza vaccine is associated with changes in automatic social behavior. Brain Behav Immun 2022; 99:339-349. [PMID: 34748895 PMCID: PMC9041378 DOI: 10.1016/j.bbi.2021.10.019] [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: 07/09/2021] [Revised: 10/14/2021] [Accepted: 10/31/2021] [Indexed: 01/03/2023] Open
Abstract
Recent evidence suggests differential patterns of social behavior following an inflammatory challenge, such that increases in inflammation may not uniformly lead to social withdrawal. Indeed, increases in inflammation have been associated with enhanced self-reported motivation to approach a specific close other, and greater neural sensitivity to positive social cues. However, no known studies have examined the association between inflammation in response to an inflammatory challenge and social behavior in humans, nor has past research examined specifically how approach and withdrawal behavior may differ based on whether the target is a close other or stranger. To address this, 31 participants (ages 18-24) received the influenza vaccine to elicit a low-grade inflammatory response. The morning before and approximately 24 h after the vaccine, participants provided a blood sample and completed a computer task assessing automatic (implicit) approach and withdrawal behavior toward a social support figure and strangers. Greater increases in the inflammatory cytokine interleukin-6 (IL-6) in response to the vaccine were associated with an increase in accuracy in avoiding strangers and a decrease in accuracy in approaching them. Increases in IL-6 were also associated with a decrease in reaction time to approach a support figure, but only when controlling for baseline IL-6 levels. There were no associations between change in IL-6 and changes in self-reported motivation to engage in social behavior with either close others, or strangers. Together, these findings reveal that increases in inflammation following the influenza vaccine are associated with automatic social behavior, especially behavior suggesting avoidance of unfamiliar social targets and ease in approaching a support figure. These data add to the growing literature suggesting that the association between inflammation and social behavior includes both social withdrawal and social approach, depending on the specific target.
Collapse
Affiliation(s)
- Tatum A. Jolink
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - Nicholas J. Fendinger
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - Gabriella M. Alvarez
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - Mallory J. Feldman
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - Monica M. Gaudier-Diaz
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| | - Keely A. Muscatell
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC USA,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC USA,Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
| |
Collapse
|
277
|
Guo XJ, Wu P, Jia X, Dong YM, Zhao CM, Chen NN, Zhang ZY, Miao YT, Yun KM, Gao CR, Ren Y. Mapping the structure of depression biomarker research: A bibliometric analysis. Front Psychiatry 2022; 13:943996. [PMID: 36186850 PMCID: PMC9523516 DOI: 10.3389/fpsyt.2022.943996] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 08/29/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Depression is a common mental disorder and the diagnosis is still based on the descriptions of symptoms. Biomarkers can reveal disease characteristics for diagnosis, prognosis, and treatment. In recent years, many biomarkers relevant to the mechanisms of depression have been identified. This study uses bibliometric methods and visualization tools to analyse the literature on depression biomarkers and its hot topics, and research frontiers to provide references for future research. METHODS Scientific publications related to depression biomarkers published between 2009 and 2022 were obtained from the Web of Science database. The BICOMB software was used to extract high-frequency keywords and to construct binary word-document and co-word matrices. gCLUTO was used for bicluster and visual analyses of high-frequency keywords. Further graphical visualizations were generated using R, CiteSpace and VOSviewer software. RESULTS A total of 14,403 articles related to depression biomarkers were identified. The United States (34.81%) and China (15.68%), which together account for more than half of all publications, can be considered the research base for the field. Among institutions, the University of California, University of London, and Harvard University are among the top in terms of publication number. Three authors (Maes M, Penninx B.W.J.H., and Berk M) emerged as eminent researchers in the field. Finally, eight research hotspots for depression biomarkers were identified using reference co-citation analysis. CONCLUSION This study used bibliometric methods to characterize the body of literature and subject knowledge in the field of depression biomarker research. Among the core biomarkers of depression, functional magnetic resonance imaging (fMRI), cytokines, and oxidative stress are relatively well established; however, research on machine learning, metabolomics, and microRNAs holds potential for future development. We found "microRNAs" and "gut microbiota" to be the most recent burst terms in the study of depression biomarkers and the likely frontiers of future research.
Collapse
Affiliation(s)
- Xiang-Jie Guo
- Department of Forensic Medicine, Shanxi Medical University, Taiyuan, China
| | - Peng Wu
- Department of Forensic Medicine, Shanxi Medical University, Taiyuan, China
| | - Xiao Jia
- College of Pharmacy, Nankai University, Tianjin, China
| | - Yi-Ming Dong
- Department of Forensic Medicine, Shanxi Medical University, Taiyuan, China
| | - Chun-Mei Zhao
- Department of Forensic Medicine, Shanxi Medical University, Taiyuan, China
| | - Nian-Nian Chen
- Department of Forensic Medicine, Shanxi Medical University, Taiyuan, China
| | - Zhi-Yong Zhang
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Yu-Ting Miao
- Department of Psychology, School of Humanities and Social Sciences, Shanxi Medical University, Taiyuan, China
| | - Ke-Ming Yun
- Department of Forensic Medicine, Shanxi Medical University, Taiyuan, China
| | - Cai-Rong Gao
- Department of Forensic Medicine, Shanxi Medical University, Taiyuan, China
| | - Yan Ren
- Department of Psychiatry, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China.,Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
278
|
Aschbacher K, Cole S, Hagan M, Rivera L, Baccarella A, Wolkowitz OM, Lieberman AF, Bush NR. An immunogenomic phenotype predicting behavioral treatment response: Toward precision psychiatry for mothers and children with trauma exposure. Brain Behav Immun 2022; 99:350-362. [PMID: 34298096 DOI: 10.1016/j.bbi.2021.07.012] [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: 02/01/2021] [Revised: 06/30/2021] [Accepted: 07/16/2021] [Indexed: 12/27/2022] Open
Abstract
Inflammatory pathways predict antidepressant treatment non-response among individuals with major depression; yet, this phenomenon may have broader transdiagnostic and transtherapeutic relevance. Among trauma-exposed mothers (Mage = 32 years) and their young children (Mage = 4 years), we tested whether genomic and proteomic biomarkers of pro-inflammatory imbalance prospectively predicted treatment response (PTSD and depression) to an empirically-supported behavioral treatment. Forty-three mother-child dyads without chronic disease completed Child Parent Psychotherapy (CPP) for roughly 9 months. Maternal blood was drawn pre-treatment, CD14 + monocytes isolated, gene expression derived from RNA sequencing (n = 34; Illumina HiSeq 4000;TruSeqcDNA library), and serum assayed (n = 43) for C-Reactive Protein (CRP) and interleukin-1ß (IL-1ß). Symptoms of PTSD and depression decreased significantly from pre- to post-treatment for both mothers and children (all p's < 0.01). Nonetheless, a higher pre-treatment maternal pro-inflammatory imbalance of M1-like versus M2-like macrophage-associated RNA expression (M1/M2) (ß = 0.476, p = .004) and IL-1ß (ß=0.333, p = .029), but not CRP, predicted lesser improvements in maternal PTSD symptoms, unadjusted and adjusting for maternal age, BMI, ethnicity, antidepressant use, income, education, and US birth. Only higher pre-treatment M1/M2 predicted a clinically-relevant threshold of PTSD non-response among mothers (OR = 3.364, p = .015; ROC-AUC = 0.78). Additionally, higher M1/M2 predicted lesser decline in maternal depressive symptoms (ß = 0.556, p = .001), though not independent of PTSD symptoms. For child outcomes, higher maternal IL-1ß significantly predicted poorer PTSD and depression symptom trajectories (ß's = 0.318-0.429, p's < 0.01), while M1/M2 and CRP were marginally associated with poorer PTSD symptom improvement (ß's = 0.295-0.333, p's < 0.056). Pre-treatment pro-inflammatory imbalance prospectively predicts poorer transdiagnostic symptom response to an empirically-supported behavioral treatment for trauma-exposed women and their young children.
Collapse
Affiliation(s)
- Kirstin Aschbacher
- Department of Psychiatry and Behavioral Sciences, Weill Institute for Neurosciences, University of California San Francisco, United States; Division of Cardiology, Department of Medicine, University of California San Francisco, United States; The Institute for Integrative Health, United States.
| | - Steve Cole
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, United States
| | - Melissa Hagan
- Department of Psychology, College of Science & Engineering, San Francisco State University, United States
| | - Luisa Rivera
- Department of Anthropology, Emory University, United States
| | | | - Owen M Wolkowitz
- Department of Psychiatry and Behavioral Sciences, Weill Institute for Neurosciences, University of California San Francisco, United States
| | - Alicia F Lieberman
- Department of Psychiatry and Behavioral Sciences, Weill Institute for Neurosciences, University of California San Francisco, United States
| | - Nicole R Bush
- Department of Psychiatry and Behavioral Sciences, Weill Institute for Neurosciences, University of California San Francisco, United States; Center for Health and Community, University of California San Francisco, United States; Department of Pediatrics, Division of Developmental Medicine, University of California San Francisco, United States.
| |
Collapse
|
279
|
Upadhyay J, Verrico CD, Cay M, Kodele S, Yammine L, Koob GF, Schreiber R. Neurocircuitry basis of the opioid use disorder-post-traumatic stress disorder comorbid state: conceptual analyses using a dimensional framework. Lancet Psychiatry 2022; 9:84-96. [PMID: 34774203 DOI: 10.1016/s2215-0366(21)00008-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/11/2020] [Accepted: 01/06/2021] [Indexed: 12/17/2022]
Abstract
Understanding the interface between opioid use disorder (OUD) and post-traumatic stress disorder (PTSD) is challenging. By use of a dimensional framework, such as research domain criteria, convergent and targetable neurobiological processes in OUD-PTSD comorbidity can be identified. We hypothesise that, in OUD-PTSD, circuitry that is implicated in two research domain criteria systems (ie, negative valence and cognitive control) underpins dysregulation of incentive salience, negative emotionality, and executive function. We also propose that the OUD-PTSD state might be systematically investigated with approaches outlined within a neuroclinical assessment framework for addictions and PTSD. Our dimensional analysis of the OUD-PTSD state shows how first-line therapeutic approaches (ie, partial μ-type opioid receptor [MOR1] agonism) modulate overlapping neurobiological and clinical features and also provides mechanistic rationale for evaluating polytherapeutic strategies (ie, partial MOR1 agonism, κ-type opioid receptor [KOR1] antagonism, and α-2A adrenergic receptor [ADRA2A] agonism). A combination of these therapeutic mechanisms is projected to facilitate recovery in patients with OUD-PTSD by mitigating negative valence states and enhancing executive control.
Collapse
Affiliation(s)
- Jaymin Upadhyay
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, USA.
| | - Christopher D Verrico
- Department of Psychiatry and Behavioral Sciences and Department of Pharmacology, Baylor College of Medicine, Houston, TX, USA
| | - Mariesa Cay
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, USA
| | - Sanda Kodele
- Faculty of Psychology and Neuroscience, Section Neuropsychology and Psychopharmacology, Maastricht University, Maastricht, Netherlands
| | - Luba Yammine
- Louis A Faillace Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - George F Koob
- National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA
| | - Rudy Schreiber
- Faculty of Psychology and Neuroscience, Section Neuropsychology and Psychopharmacology, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
280
|
Abstract
ABSTRACT Inflammatory phenomena are found in many psychiatric disorders-notably, depression, schizophrenia, and posttraumatic stress disorder. Inflammation has been linked to severity and treatment resistance, and may both contribute to, and result from, the pathophysiology of some psychiatric illnesses. Emerging research suggests that inflammation may contribute to symptom domains of reward, motor processing, and threat reactivity across different psychiatric diagnoses. Reward-processing deficits contribute to motivational impairments in depression and schizophrenia, and motor-processing deficits contribute to psychomotor slowing in both depression and schizophrenia. A number of experimental models and clinical trials suggest that inflammation produces deficits in reward and motor processing through common pathways connecting the cortex and the striatum, which includes the nucleus accumbens, caudate nucleus, and putamen.The observed effects of inflammation on psychiatric disorders may cut across traditional conceptualizations of psychiatric diagnoses. Further study may lead to targeted immunomodulating treatments that address difficult-to-treat symptoms in a number of psychiatric disorders. In this review, we use a Research Domain Criteria framework to discuss proposed mechanisms for inflammation and its effects on the domains of reward processing, psychomotor slowing, and threat reactivity. We also discuss data that support contributing roles of metabolic dysregulation and sex differences on the behavioral outcomes of inflammation. Finally, we discuss ways that future studies can help disentangle this complex topic to yield fruitful results that will help advance the field of psychoneuroimmunology.
Collapse
Affiliation(s)
- David S Thylur
- From the Department of Psychiatry and Behavioral Sciences, Emory University
| | | |
Collapse
|
281
|
Treatment resistance in psychiatry: state of the art and new directions. Mol Psychiatry 2022; 27:58-72. [PMID: 34257409 PMCID: PMC8960394 DOI: 10.1038/s41380-021-01200-3] [Citation(s) in RCA: 170] [Impact Index Per Article: 56.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/26/2021] [Accepted: 06/10/2021] [Indexed: 02/06/2023]
Abstract
Treatment resistance affects 20-60% of patients with psychiatric disorders; and is associated with increased healthcare burden and costs up to ten-fold higher relative to patients in general. Whilst there has been a recent increase in the proportion of psychiatric research focussing on treatment resistance (R2 = 0.71, p < 0.0001), in absolute terms this is less than 1% of the total output and grossly out of proportion to its prevalence and impact. Here, we provide an overview of treatment resistance, considering its conceptualisation, assessment, epidemiology, impact, and common neurobiological models. We also review new treatments in development and future directions. We identify 23 consensus guidelines on its definition, covering schizophrenia, major depressive disorder, bipolar affective disorder, and obsessive compulsive disorder (OCD). This shows three core components to its definition, but also identifies heterogeneity and lack of criteria for a number of disorders, including panic disorder, post-traumatic stress disorder, and substance dependence. We provide a reporting check-list to aid comparisons across studies. We consider the concept of pseudo-resistance, linked to poor adherence or other factors, and provide an algorithm for the clinical assessment of treatment resistance. We identify nine drugs and a number of non-pharmacological approaches being developed for treatment resistance across schizophrenia, major depressive disorder, bipolar affective disorder, and OCD. Key outstanding issues for treatment resistance include heterogeneity and absence of consensus criteria, poor understanding of neurobiology, under-investment, and lack of treatments. We make recommendations to address these issues, including harmonisation of definitions, and research into the mechanisms and novel interventions to enable targeted and personalised therapeutic approaches.
Collapse
|
282
|
Bekhbat M, Treadway MT, Felger JC. Inflammation as a Pathophysiologic Pathway to Anhedonia: Mechanisms and Therapeutic Implications. Curr Top Behav Neurosci 2022; 58:397-419. [PMID: 34971449 DOI: 10.1007/7854_2021_294] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Anhedonia, characterized by a lack of motivation, interest, or ability to experience pleasure, is a prominent symptom of depression and other psychiatric disorders and has been associated with poor response to standard therapies. One pathophysiologic pathway receiving increased attention for its potential role in anhedonia is inflammation and its effects on the brain. Exogenous administration of inflammatory stimuli to humans and laboratory animals has reliably been found to affect neurotransmitters and neurocircuits involved in reward processing, including the ventral striatum and ventromedial prefrontal cortex, in association with reduced motivation. Moreover, a rich literature including meta-analyses describes increased inflammation in a significant proportion of patients with depression and other psychiatric illnesses involving anhedonia, as evident by elevated inflammatory cytokines, acute phase proteins, chemokines, and adhesion molecules in both the periphery and central nervous system. This endogenous inflammation may arise from numerous sources including stress, obesity or metabolic dysfunction, genetics, and lifestyle factors, many of which are also risk factors for psychiatric illness. Consistent with laboratory studies involving exogenous administration of peripheral inflammatory stimuli, neuroimaging studies have further confirmed that increased endogenous inflammation in depression is associated with decreased activation of and reduced functional connectivity within reward circuits involving ventral striatum and ventromedial prefrontal cortex in association with anhedonia. Here, we review recent evidence of relationships between inflammation and anhedonia, while highlighting translational and mechanistic work describing the impact of inflammation on synthesis, release, and reuptake of neurotransmitters like dopamine and glutamate that affects circuits to drive motivational deficits. We will then present insight into novel pharmacological strategies that target either inflammation or its downstream effects on the brain and behavior. The meaningful translation of these concepts through appropriately designed trials targeting therapies for psychiatric patients with high inflammation and transdiagnostic symptoms of anhedonia is also discussed.
Collapse
Affiliation(s)
- Mandakh Bekhbat
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Michael T Treadway
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Atlanta, GA, USA
- Department of Psychology, Emory University, Atlanta, GA, USA
| | - Jennifer C Felger
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA.
- Winship Cancer Institute, Atlanta, GA, USA.
| |
Collapse
|
283
|
Abstract
Depression and psychosis have a developmental component to their origin. Epidemiologic evidence, which we synthesize in this nonsystematic review, suggests that early-life infection, inflammation, and metabolic alterations could play a role in the etiology of these psychiatric disorders. The risk of depression and psychosis is associated with prenatal maternal and childhood infections, which could be mediated by impaired neurodevelopment. Evidence suggests linear dose-response associations between elevated concentrations of circulating inflammatory markers in childhood, particularly the inflammatory cytokine interleukin 6, and the risk for depression and psychosis subsequently in early adulthood. Childhood inflammatory markers are also associated with persistence of depressive symptoms subsequently in adolescence and early adulthood. Developmental trajectories reflecting persistently high insulin levels during childhood and adolescence are associated with a higher risk of psychosis in adulthood, whereas increased adiposity during and after puberty is associated with the risk of depression. Together, these findings suggest that higher levels of infection, inflammation, and metabolic alterations commonly seen in people with depression and psychosis could be a cause for, rather than simply a consequence of, these disorders. Therefore, early-life immuno-metabolic alterations, as well as factors influencing these alterations such as adversity or maltreatment, could represent targets for prevention of these psychiatric disorders. Inflammation could also be an important treatment target for depression and psychosis. The field requires further research to examine sensitive periods when exposure to such immuno-metabolic alterations is most harmful. Interventional studies are also needed to test the potential usefulness of targeting early-life immuno-metabolic alterations for preventing adult depression and psychosis.
Collapse
|
284
|
McFarland DC, Riba M, Grassi L. Clinical Implications of Cancer Related Inflammation and Depression: A Critical Review. Clin Pract Epidemiol Ment Health 2021; 17:287-294. [PMID: 35444703 PMCID: PMC8985467 DOI: 10.2174/1745017902117010287] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 09/07/2021] [Accepted: 10/01/2021] [Indexed: 12/27/2022]
Abstract
Background: Neuropsychiatric symptoms are problematic in cancer settings. In addition to poor quality of life, depression is associated with worsened survival. Patients who develop depression that responds to treatment have the same cancer-related survival as those patients who never had depression. Although depression in patients with cancer is common, it is often unrecognized, untreated, or at best, undertreated. There remains untapped potential for underlying cancer-related biology associated with depression to help clinicians correctly identify depressed cancer patients and orchestrate appropriate treatments to address cancer-related depression. Biologically, inflammation has been most vigorously described in its association with depression in otherwise healthy patients and to a significant extent in patients with medical illness. This association is especially relevant to patients with cancer since so many aspects of cancer induce inflammation. In addition to cancer itself, its treatments (e.g., surgery, radiation, chemotherapy, and systemic therapies) and associated factors (e.g., smoking, obesity, aging) are all associated with increased inflammation that can drive immunological changes in the brain followed by depression. This critical review investigates the relationship between depression and cancer-related inflammation. It investigates several hypotheses that support these relationships in cancer patients. Special attention is given to the data that support certain inflammatory markers specific to both cancer and depression, the neurobiological mechanisms by which inflammation can impact neurotransmitters and neurocircuits in the brain, and the data addressing interventions that reduce inflammation and depression in cancer patients, and future directions.
Collapse
|
285
|
Li W, Xu Y, Liu Z, Shi M, Zhang Y, Deng Y, Zhong X, Chen L, He J, Zeng J, Luo M, Cao W, Wan W. TRPV4 inhibitor HC067047 produces antidepressant-like effect in LPS-induced depression mouse model. Neuropharmacology 2021; 201:108834. [PMID: 34637786 DOI: 10.1016/j.neuropharm.2021.108834] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 10/05/2021] [Accepted: 10/07/2021] [Indexed: 01/07/2023]
Abstract
Inflammation is a crucial component that contributes to the pathogenesis of major depressive disorder. It has been revealed that the nonselective cation channel transient receptor potential vanilloid 4 (TRPV4) profoundly affects a variety of physiological processes, including inflammation. However, its roles and mechanisms in LPS-induced depression are still unclear. Here, for the first time, we found that there was a significant increase in TRPV4 in the hippocampus in a depression mouse model induced by LPS. TRPV4 inhibitor HC067047 or knockdown the hippocampal TRPV4 with TRPV4 shRNA could effectively rescue the aberrant behaviors. Furthermore, TRPV4 inhibitor HC067047 reduced the activation of astrocyte and microglia, decreased expression of CaMKII-NLRP3 inflammasome and increased the expression of neurogenesis marker DCX in the hippocampus. In addition, enhanced neuroinflammation in the serum was also reversed by TRPV4 inhibitor HC067047. Thus, we consider that TRPV4 has an important role in contributing to the depression-like behavior following LPS-induced systemic inflammation.
Collapse
Affiliation(s)
- Wei Li
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, 421001, Hengyang, Hunan, China
| | - Yang Xu
- Institute of Neuroscience, Hengyang Medical School, University of South China, 421001, Hengyang, Hunan, China
| | - Zhenghai Liu
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, 421001, Hengyang, Hunan, China
| | - Mengmeng Shi
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, 421001, Hengyang, Hunan, China
| | - Yuan Zhang
- Department of Pathology, Hengyang Medical School, University of South China, 421001, Hengyang, Hunan, China
| | - Yingcheng Deng
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, 421001, Hengyang, Hunan, China
| | - Xiaolin Zhong
- Institute of Clinical Medicine, The First Affiliated Hospital of University of South China, 421001, Hengyang, Hunan, China
| | - Ling Chen
- Institute of Clinical Medicine, The First Affiliated Hospital of University of South China, 421001, Hengyang, Hunan, China
| | - Jie He
- Department of Pathology, Hengyang Medical School, University of South China, 421001, Hengyang, Hunan, China
| | - Jiayu Zeng
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, 421001, Hengyang, Hunan, China
| | - Mingying Luo
- Department of Anatomy & Histology & Embryology, Kunming Medical University, 650500, Kunming, Yunnan, China
| | - Wenyu Cao
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, 421001, Hengyang, Hunan, China.
| | - Wei Wan
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical School, University of South China, 421001, Hengyang, Hunan, China; Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, 571199, Haikou, China.
| |
Collapse
|
286
|
A new experimental design to study inflammation-related versus non-inflammation-related depression in mice. J Neuroinflammation 2021; 18:290. [PMID: 34895261 PMCID: PMC8666053 DOI: 10.1186/s12974-021-02330-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 11/24/2021] [Indexed: 12/28/2022] Open
Abstract
Background Major depressive disorder (MDD) represents a major public health concern, particularly due to its steadily rising prevalence and the poor responsiveness to standard antidepressants notably in patients afflicted with chronic inflammatory conditions, such as obesity. This highlights the need to improve current therapeutic strategies, including by targeting inflammation based on its role in the pathophysiology and treatment responsiveness of MDD. Nevertheless, dissecting the relative contribution of inflammation in the development and treatment of MDD remains a major issue, further complicated by the lack of preclinical depression models suitable to experimentally dissociate inflammation-related vs. inflammation-unrelated depression. Methods While current models usually focus on one particular MDD risk factor, we compared in male C57BL/6J mice the behavioral, inflammatory and neurobiological impact of chronic exposure to high-fat diet (HFD), a procedure known to induce inflammation-related depressive-like behaviors, and unpredictable chronic mild stress (UCMS), a stress-induced depression model notably renowned for its responsivity to antidepressants. Results While both paradigms induced neurovegetative, depressive-like and anxiety-like behaviors, inflammation and downstream neurobiological pathways contributing to inflammation-driven depression were specifically activated in HFD mice, as revealed by increased circulating levels of inflammatory factors, as well as brain expression of microglial activation markers and enzymes from the kynurenine and tetrahydrobiopterin (BH4) pathways. In addition, serotoninergic and dopaminergic systems were differentially impacted, depending on the experimental condition. Conclusions These data validate an experimental design suitable to deeply study the mechanisms underlying inflammation-driven depression comparatively to non-inflammatory depression. This design could help to better understand the pathophysiology of treatment resistant depression. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02330-9.
Collapse
|
287
|
Tran AA, De Smet M, Grant GD, Khoo TK, Pountney DL. Investigating the Convergent Mechanisms between Major Depressive Disorder and Parkinson's Disease. Complex Psychiatry 2021; 6:47-61. [PMID: 34883500 DOI: 10.1159/000512657] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 10/23/2020] [Indexed: 12/21/2022] Open
Abstract
Major depressive disorder (MDD) affects more than cognition, having a temporal relationship with neuroinflammatory pathways of Parkinson's disease (PD). Although this association is supported by epidemiological and clinical studies, the underlying mechanisms are unclear. Microglia and astrocytes play crucial roles in the pathophysiology of both MDD and PD. In PD, these cells can be activated by misfolded forms of the protein α-synuclein to release cytokines that can interact with multiple different physiological processes to produce depressive symptoms, including monoamine transport and availability, the hypothalamus-pituitary axis, and neurogenesis. In MDD, glial cell activation can be induced by peripheral inflammatory agents that cross the blood-brain barrier and/or c-Fos signalling from neurons. The resulting neuroinflammation can cause neurodegeneration due to oxidative stress and glutamate excitotoxicity, contributing to PD pathology. Astrocytes are another major link due to their recognized role in the glymphatic clearance mechanism. Research suggesting that MDD causes astrocytic destruction or structural atrophy highlights the possibility that accumulation of α-synuclein in the brain is facilitated as the brain cannot adequately clear the protein aggregates. This review examines research into the overlapping pathophysiology of MDD and PD with particular focus on the roles of glial cells and neuroinflammation.
Collapse
Affiliation(s)
- Angela A Tran
- School of Medical Science, Griffith University, Southport, Queensland, Australia.,School of Medicine, Griffith University, Southport, Queensland, Australia
| | - Myra De Smet
- School of Medical Science, Griffith University, Southport, Queensland, Australia
| | - Gary D Grant
- School of Pharmacy and Pharmacology, Griffith University, Southport, Queensland, Australia
| | - Tien K Khoo
- School of Medicine, Griffith University, Southport, Queensland, Australia.,Menzies Health Institute Queensland, Griffith University, Southport, Queensland, Australia.,School of Medicine, University of Wollongong, Wollongong, New South Wales, Australia
| | - Dean L Pountney
- School of Medical Science, Griffith University, Southport, Queensland, Australia
| |
Collapse
|
288
|
Avan R, Sahebnasagh A, Hashemi J, Monajati M, Faramarzi F, Henney NC, Montecucco F, Jamialahmadi T, Sahebkar A. Update on Statin Treatment in Patients with Neuropsychiatric Disorders. Life (Basel) 2021; 11:1365. [PMID: 34947895 PMCID: PMC8703562 DOI: 10.3390/life11121365] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/26/2021] [Accepted: 12/04/2021] [Indexed: 02/06/2023] Open
Abstract
Statins are widely accepted as first-choice agents for the prevention of lipid-related cardiovascular diseases. These drugs have both anti-inflammatory and anti-oxidant properties, which may also make them effective as potential treatment marked by perturbations in these pathways, such as some neuropsychiatric disorders. In this narrative review, we have investigated the effects of statin therapy in individuals suffering from major depressive disorder (MDD), schizophrenia, anxiety, obsessive-compulsive disorder (OCD), bipolar disorder (BD), delirium, and autism spectrum disorders using a broad online search of electronic databases. We also explored the adverse effects of these drugs to obtain insights into the benefits and risks associated with their use in the treatment of these disorders. Lipophilic statins (including simvastatin) because of better brain penetrance may have greater protective effects against MDD and schizophrenia. The significant positive effects of statins in the treatment of anxiety disorders without any serious adverse side effects were shown in numerous studies. In OCD, BD, and delirium, limitations, and contradictions in the available data make it difficult to draw conclusions on any positive effect of statins. The positive effects of simvastatin in autism disorders have been evaluated in only a small number of clinical trials. Although some studies showed positive effect of statins in some neuropsychiatric disorders, further prospective studies are needed to confirm this and define the most effective doses and treatment durations.
Collapse
Affiliation(s)
- Razieh Avan
- Department of Clinical Pharmacy, Medical Toxicology and Drug Abuse Research Center (MTDRC), School of Pharmacy, Birjand University of Medical Sciences, Birjand 9717853577, Iran;
| | - Adeleh Sahebnasagh
- Clinical Research Center, Department of Internal Medicine, North Khorasan University of Medical Sciences, Bojnurd 9453155166, Iran;
| | - Javad Hashemi
- Department of Pathobiology and Laboratory Sciences, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd 9453155166, Iran;
| | - Mahila Monajati
- Department of Internal Medicine, Golestan University of Medical Sciences, Gorgan 4934174515, Iran;
| | - Fatemeh Faramarzi
- Clinical Pharmacy Research Center, Iran University of Medical Sciences, Tehran 1445613131, Iran;
| | - Neil C. Henney
- Pharmacy & Biomolecular Sciences, Liverpool John Moores University, Liverpool L3 5UX, UK;
| | - Fabrizio Montecucco
- IRCCS Ospedale Policlinico San Martino Genoa-Italian Cardiovascular Network, 10 Largo Benzi, 16132 Genoa, Italy;
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132 Genoa, Italy
| | - Tannaz Jamialahmadi
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran;
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 9177948954, Iran
| |
Collapse
|
289
|
Baune BT, Sampson E, Louise J, Hori H, Schubert KO, Clark SR, Mills NT, Fourrier C. No evidence for clinical efficacy of adjunctive celecoxib with vortioxetine in the treatment of depression: A 6-week double-blind placebo controlled randomized trial. Eur Neuropsychopharmacol 2021; 53:34-46. [PMID: 34375789 DOI: 10.1016/j.euroneuro.2021.07.092] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 07/19/2021] [Accepted: 07/21/2021] [Indexed: 12/22/2022]
Abstract
Given the role of low-grade inflammation in the pathophysiology of major depressive disorder (MDD), anti-inflammatory strategies may improve treatment outcomes in some patients. However, it is controversial whether they can be used as adjunctive treatments and whether pre-treatment levels of inflammation can predict treatment outcomes. This study was conducted to measure the efficacy of anti-inflammatory augmentation of antidepressant treatment in MDD patients; and to investigate whether treatment response was dependent on baseline inflammation levels. This parallel-group randomised, double-blind, placebo-controlled trial was conducted at the University of Adelaide (Australia). Participants with MDD were randomised to receive vortioxetine with celecoxib or vortioxetine with placebo for six weeks, and baseline blood high sensitivity C reactive protein levels were measured. Primary outcome was change in depressive symptoms (Montgomery-Åsberg Depression Rating Scale) and secondary outcomes included change in cognition (THINC-integrated tool - Codebreaker task) and functioning (Functioning Assessment Short Test) over 6 weeks. There was no evidence of superior efficacy of celecoxib augmentation over placebo on depressive symptom severity, response and remission rates, cognition and psychosocial functioning. There was also no evidence that pre-treatment inflammation levels modified the effect of celecoxib augmentation versus placebo. This observed lack of efficacy of celecoxib add-on does not support the use of celecoxib augmentation of antidepressants in the treatment of MDD in a cohort that mostly comprises treatment-resistant individuals. Additionally, C-reactive protein may not be suitable to predict treatment selection and response in MDD. The study was registered on the Australian New Zealand Clinical Trials Registry: ACTRN12617000527369 (www.anzctr.org.au/Trial/Registration/TrialReview.aspx?ACTRN=12617000527369p).
Collapse
Affiliation(s)
- Bernhard T Baune
- Discipline of Psychiatry, Adelaide Medical School, University of Adelaide, Adelaide, Australia; Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Melbourne, Australia; The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia; Department of Psychiatry and Psychotherapy, University Hospital Münster, University of Münster, Münster, Germany.
| | - Emma Sampson
- Discipline of Psychiatry, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Jennie Louise
- 'Faculty of Health Sciences, University of Adelaide, Adelaide, Australia
| | - Hikaru Hori
- Department of Psychiatry, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - K Oliver Schubert
- Discipline of Psychiatry, Adelaide Medical School, University of Adelaide, Adelaide, Australia; Northern Adelaide Mental Health Service, Salisbury, Australia
| | - Scott R Clark
- Discipline of Psychiatry, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Natalie T Mills
- Discipline of Psychiatry, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Célia Fourrier
- Discipline of Psychiatry, Adelaide Medical School, University of Adelaide, Adelaide, Australia; Lysosomal Health in Ageing, Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| |
Collapse
|
290
|
Transcriptomic signatures of psychomotor slowing in peripheral blood of depressed patients: evidence for immunometabolic reprogramming. Mol Psychiatry 2021; 26:7384-7392. [PMID: 34535767 PMCID: PMC8881295 DOI: 10.1038/s41380-021-01258-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 07/25/2021] [Accepted: 07/30/2021] [Indexed: 02/08/2023]
Abstract
Inflammation impacts basal ganglia motor circuitry in association with psychomotor retardation, a key symptom of major depression (MD). We previously reported associations between circulating protein inflammatory biomarkers and psychomotor slowing as measured by neuropsychological tests probing psychomotor speed in patients with MD. To discover novel transcriptional signatures in peripheral blood immune cells related to psychomotor slowing, microarray data were analyzed in a primary cohort of 88 medically-stable, unmedicated, ambulatory MD patients. Results were confirmed and extended in a second cohort of 57 patients with treatment resistant depression (TRD) before and after anti-inflammatory challenge with the tumor necrosis factor antagonist infliximab versus placebo. Composite scores reflecting pure motor and cognitive-motor processing speed were linearly associated with 403 and 266 gene transcripts in each cohort, respectively (|R| > 0.30, p < 0.01), that were enriched for cytokine signaling and glycolysis-related pathways (p < 0.05). Unsupervised clustering in the primary cohort revealed two psychomotor slowing-associated gene co-expression modules that were enriched for interferon, interleukin-6, aerobic glycolysis, and oxidative phosphorylation pathways (p < 0.05, q < 0.1). Transcripts were predominantly derived from monocytes, plasmacytoid dendritic cells, and natural killer cells (p's < 0.05). In infliximab-treated TRD patients with high plasma C-reactive protein concentrations (>5 mg/L), two differential co-expression modules enriched for oxidative stress and mitochondrial degradation were associated with improvements in psychomotor reaction time (p < 0.05). These results indicate that inflammatory signaling and associated metabolic reprogramming in peripheral blood immune cells are associated with systemic inflammation in depression and may affect relevant brain circuits to promote psychomotor slowing.
Collapse
|
291
|
Carta MG, Kalcev G, Fornaro M, Nardi AE. Novel experimental and early investigational drugs for the treatment of bipolar disorder. Expert Opin Investig Drugs 2021; 30:1081-1087. [PMID: 34844484 DOI: 10.1080/13543784.2021.2000965] [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] [Indexed: 01/19/2023]
Abstract
INTRODUCTION The quest toward more effective treatments for bipolar disorder (BD) solicits novel drugs and further research on the underpinning neurobiology. The present review aims to critically appraise the existing evidence about the pharmacological treatment of BD toward the development of novel treatment avenues. AREAS COVERED The present review appraises animal and human studies concerning both the currently available psychotropic drugs, and the general medicine drugs which may represent a path toward the development of novel drugs for BD. PubMed and Scopus were last accessed on February 20th, 2021 for records indexed upon inception relevant to the pharmacological treatment of BD. Immune-modulating agents, anti-inflammatory agents, and glutamate antagonists represent the most intriguing potential targets for the development of new drugs for BD, thus receiving critical appraisal in the present text. EXPERT OPINION Regardless of the neurobiological pathways worthy of investigation toward the development of experimental drugs for BD, several unmet needs need to be addressed first. In particular, several biomarkers are altered in BD. However, it is the opinion herein expressed by the authors that it remains uncertain what comes first, that is peripheral changes or the disease.
Collapse
Affiliation(s)
- Mauro Giovanni Carta
- Department of Public Health, Clinical and Molecular Medicine, University of Cagliari, Cagliari Italy
| | - Goce Kalcev
- Department of Mechanical, Chemical and Materials Engineering, International Ph.D. In Innovation Sciences and Technologies, University of Cagliari, Cagliari Italy
| | - Michele Fornaro
- Department of Psychiatry, University of Federico II of Naples, Italy
| | - Antonio Egidio Nardi
- Laboratory Panic and Respiration, Institute of Psychiatry (Ipub), Federal University of Rio De Janeiro (Ufrj), Rio De Janeiro, Brazil
| |
Collapse
|
292
|
Abstract
Neuropsychiatric diseases have traditionally been studied from brain, and mind-centric perspectives. However, mounting epidemiological and clinical evidence shows a strong correlation of neuropsychiatric manifestations with immune system activation, suggesting a likely mechanistic interaction between the immune and nervous systems in mediating neuropsychiatric disease. Indeed, immune mediators such as cytokines, antibodies, and complement proteins have been shown to affect various cellular members of the central nervous system in multitudinous ways, such as by modulating neuronal firing rates, inducing cellular apoptosis, or triggering synaptic pruning. These observations have in turn led to the exciting development of clinical therapies aiming to harness this neuro-immune interaction for the treatment of neuropsychiatric disease and symptoms. Besides the clinic, important theoretical fundamentals can be drawn from the immune system and applied to our understanding of the brain and neuropsychiatric disease. These new frameworks could lead to novel insights in the field and further potentiate the development of future therapies to treat neuropsychiatric disease.
Collapse
|
293
|
De Giorgi R, Martens M, Rizzo Pesci N, Cowen PJ, Harmer CJ. The effects of atorvastatin on emotional processing, reward learning, verbal memory and inflammation in healthy volunteers: An experimental medicine study. J Psychopharmacol 2021; 35:1479-1487. [PMID: 34872404 PMCID: PMC8652357 DOI: 10.1177/02698811211060307] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Growing evidence from clinical trials and epidemiological studies suggests that statins can have clinically significant antidepressant effects, potentially related to anti-inflammatory action on several neurobiological structures. However, the underlying neuropsychological mechanisms of these effects remain unexplored. AIMS In this experimental medicine trial, we investigated the 7-day effects of the lipophilic statin, atorvastatin on a battery of neuropsychological tests and inflammation in healthy volunteers. METHODS Fifty healthy volunteers were randomised to either 7 days of atorvastatin 20 mg or placebo in a double-blind design. Participants were assessed with psychological questionnaires and a battery of well-validated behavioural tasks assessing emotional processing, which is sensitive to putative antidepressant effects, reward learning and verbal memory, as well as the inflammatory marker, C-reactive protein. RESULTS Compared to placebo, 7-day atorvastatin increased the recognition (p = 0.006), discriminability (p = 0.03) and misclassifications (p = 0.04) of fearful facial expression, independently from subjective states of mood and anxiety, and C-reactive protein levels. Otherwise, atorvastatin did not significantly affect any other psychological and behavioural measure, nor peripheral C-reactive protein. CONCLUSIONS Our results reveal for the first time the early influence of atorvastatin on emotional cognition by increasing the processing of anxiety-related stimuli (i.e. increased recognition, discriminability and misclassifications of fearful facial expression) in healthy volunteers, in the absence of more general effects on negative affective bias. Further studies exploring the effects of statins in depressed patients, especially with raised inflammatory markers, may clarify this finding and inform future clinical trials.
Collapse
Affiliation(s)
- Riccardo De Giorgi
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, UK
- Warneford Hospital, Oxford Health NHS Foundation Trust, Oxford, UK
| | - Marieke Martens
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, UK
| | - Nicola Rizzo Pesci
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, UK
| | - Philip J Cowen
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, UK
- Warneford Hospital, Oxford Health NHS Foundation Trust, Oxford, UK
| | - Catherine J Harmer
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, UK
- Warneford Hospital, Oxford Health NHS Foundation Trust, Oxford, UK
| |
Collapse
|
294
|
Matt SM. Targeting neurotransmitter-mediated inflammatory mechanisms of psychiatric drugs to mitigate the double burden of multimorbidity and polypharmacy. Brain Behav Immun Health 2021; 18:100353. [PMID: 34647105 PMCID: PMC8495104 DOI: 10.1016/j.bbih.2021.100353] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 09/16/2021] [Accepted: 09/18/2021] [Indexed: 12/12/2022] Open
Abstract
The increased incidence of multimorbidities and polypharmacy is a major concern, particularly in the growing aging population. While polypharmacy can be beneficial, in many cases it can be more harmful than no treatment, especially in individuals suffering from psychiatric disorders, who have elevated risks of multimorbidity and polypharmacy. Age-related chronic inflammation and immunopathologies might contribute to these increased risks in this population, but the optimal clinical management of drug-drug interactions and the neuro-immune mechanisms that are involved warrants further investigation. Given that neurotransmitter systems, which psychiatric medications predominantly act on, can influence the development of inflammation and the regulation of immune function, it is important to better understand these interactions to develop more successful strategies to manage these comorbidities and complicated polypharmacy. I propose that expanding upon research in translationally relevant human in vitro models, in tandem with other preclinical models, is critical to defining the neurotransmitter-mediated mechanisms by which psychiatric drugs alter immune function. This will define more precisely the interactions of psychiatric drugs and other immunomodulatory drugs, used in combination, enabling identification of novel targets to be translated into more efficacious diagnostic, preventive, and therapeutic interventions. This interdisciplinary approach will aid in better precision polypharmacy for combating adverse events associated with multimorbidity and polypharmacy in the future.
Collapse
Affiliation(s)
- Stephanie M. Matt
- Drexel University College of Medicine, Department of Pharmacology and Physiology, Philadelphia, PA, USA
| |
Collapse
|
295
|
Yavi M, Henter ID, Park LT, Zarate C. Key considerations in the pharmacological management of treatment-resistant depression. Expert Opin Pharmacother 2021; 22:2405-2415. [PMID: 34252320 PMCID: PMC8648908 DOI: 10.1080/14656566.2021.1951225] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 06/30/2021] [Indexed: 10/20/2022]
Abstract
Introduction: Treatment-resistant depression (TRD) is a complex, multifactorial, and biologically heterogeneous disorder with debilitating outcomes. Understanding individual reasons why patients do not respond to treatment is necessary for improving clinical recommendations regarding medication regimens, augmentation strategies, and alternative treatments.Areas covered: This manuscript reviews evidence-based treatment strategies for the clinical management of TRD. Current developments in the field and potential future recommendations for personalized treatment of TRD are also discussed.Expert opinion: Treatment guidelines for TRD are limited by the heterogeneous nature of the disorder. Furthermore, current strategies reflect this heterogeneity by emphasizing disease characteristics as well as drug trial response or failure. Developing robust biomarkers that could one day be integrated into clinical practice has the potential to advance specific treatment targets and ultimately improve treatment and remission outcomes.
Collapse
Affiliation(s)
- Mani Yavi
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental HealthNational Institutes of Health Bethesda, MD, USA
| | | | | | | |
Collapse
|
296
|
Affiliation(s)
- Carmine M Pariante
- Department of Psychological Medicine, Institute of Psychiatry, Psychology, and Neuroscience, King's College London
| |
Collapse
|
297
|
Czysz AH, Mason BL, Li Q, Chin-Fatt C, Minhajuddin A, Carmody T, Trivedi MH. Comparison of inflammatory markers as moderators of depression outcomes: A CO-MED study. J Affect Disord 2021; 295:1066-1071. [PMID: 34706415 DOI: 10.1016/j.jad.2021.08.116] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 07/30/2021] [Accepted: 08/28/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Prior work suggests some individual immunomarkers may be useful moderators of treatment response between antidepressant medications. The relative moderating effect of individual immunomarkers remains unclear. It is also unknown whether combinations of immunomarkers have a superior moderating effect compared to any individual immunomarker. METHODS Baseline immunomarker levels were assayed using multiplex from a subset of participants in the CO-MED trial (n = 143). Individual and combinations of 19 immunomarkers were modeled as moderators between the three treatment arms (escitalopram monotherapy, escitalopram-bupropion and venlafaxine-mirtazapine) across a variety of depression outcomes. RESULTS Only IL-13 demonstrated a consistent moderating effect across all depression outcome measures. High IL-13 (>20 pg/ml) was associated with higher remission rates to bupropion-escitalopram (67%) versus escitalopram (24%) whereas low IL-13 was associated higher remission rates to escitalopram (59%) versus bupropion-escitalopram (38%). A similar, but weaker moderating effect was observed with venlafaxine-mirtazapine versus escitalopram. The addition of multiple immunomarkers did not consistently improve predictive modeling. LIMITATIONS This is a secondary analysis of a single clinical trial with a relatively small sample size per treatment arm. The testing of specific individual and combinations of biomarkers was data-driven. CONCLUSIONS Among 19 immunomarkers, Il-13 was the best single moderator of treatment outcome. Combinations of immunomarkers were not meaningfully superior to Il-13.
Collapse
Affiliation(s)
- Andrew H Czysz
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | - Brittany L Mason
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | - Qiwei Li
- Department of Mathematical Sciences, University of Texas Dallas, Richardson, TX, United States of America
| | - Cherise Chin-Fatt
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | - Abu Minhajuddin
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | - Thomas Carmody
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | - Madhukar H Trivedi
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, United States of America.
| |
Collapse
|
298
|
Hölsken S, Krefting F, Schedlowski M, Sondermann W. Common Fundamentals of Psoriasis and Depression. Acta Derm Venereol 2021; 101:adv00609. [PMID: 34806760 PMCID: PMC9455336 DOI: 10.2340/actadv.v101.565] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Psoriasis is an inflammatory, immune-mediated disease that is frequently associated with psychological comorbidities such as depression. The stigma patients feel because of the appearance of their skin may contribute to the high psycho-social burden of psoriasis. However, there is emerging evidence that overlapping biological mechanisms are, to a substantial degree, responsible for the close interaction between psoriasis and depression. Increased proinflammatory mediators, such as C-reactive protein or interleukin-6, are present in both psoriasis and depression, indicating that inflammation may represent a pathophysiological link between the diseases. Anti-inflammatory biologic therapies treat the clinical manifestations of psoriasis, but might also play a significant role in reducing associated depressive symptoms in patients with psoriasis. Comparison between single studies focusing on the change in depressive symptoms in psoriasis is limited by inconsistency in the depression screening tools applied.
Collapse
Affiliation(s)
| | | | | | - Wiebke Sondermann
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, University Duisburg-Essen, Hufelandstr. 55, DE-45122 Essen, Germany.
| |
Collapse
|
299
|
Borbély É, Simon M, Fuchs E, Wiborg O, Czéh B, Helyes Z. Novel drug developmental strategies for treatment-resistant depression. Br J Pharmacol 2021; 179:1146-1186. [PMID: 34822719 PMCID: PMC9303797 DOI: 10.1111/bph.15753] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 10/17/2021] [Accepted: 11/14/2021] [Indexed: 11/30/2022] Open
Abstract
Major depressive disorder is a leading cause of disability worldwide. Because conventional therapies are ineffective in many patients, novel strategies are needed to overcome treatment‐resistant depression (TRD). Limiting factors of successful drug development in the last decades were the lack of (1) knowledge of pathophysiology, (2) translational animal models and (3) objective diagnostic biomarkers. Here, we review novel drug targets and drug candidates currently investigated in Phase I–III clinical trials. The most promising approaches are inhibition of glutamatergic neurotransmission by NMDA and mGlu5 receptor antagonists, modulation of the opioidergic system by κ receptor antagonists, and hallucinogenic tryptamine derivates. The only registered drug for TRD is the NMDA receptor antagonist, S‐ketamine, but add‐on therapies with second‐generation antipsychotics, certain nutritive, anti‐inflammatory and neuroprotective agents seem to be effective. Currently, there is an intense research focus on large‐scale, high‐throughput omics and neuroimaging studies. These results might provide new insights into molecular mechanisms and potential novel therapeutic strategies.
Collapse
Affiliation(s)
- Éva Borbély
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Hungary.,Molecular Pharmacology Research Group, Szentágothai János Research Centre, University of Pécs, Pécs, Hungary
| | - Mária Simon
- Department of Psychiatry and Psychotherapy, Clinical Centre, Medical School, University of Pécs, Hungary
| | - Eberhard Fuchs
- German Primate Center, Leibniz Institute for Primate Research, Göttingen, Germany
| | - Ove Wiborg
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Boldizsár Czéh
- Neurobiology of Stress Research Group, Szentágothai János Research Centre, University of Pécs, Pécs, Hungary.,Department of Laboratory Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Hungary.,Molecular Pharmacology Research Group, Szentágothai János Research Centre, University of Pécs, Pécs, Hungary
| |
Collapse
|
300
|
Appleton KM, Voyias PD, Sallis HM, Dawson S, Ness AR, Churchill R, Perry R. Omega-3 fatty acids for depression in adults. Cochrane Database Syst Rev 2021; 11:CD004692. [PMID: 34817851 PMCID: PMC8612309 DOI: 10.1002/14651858.cd004692.pub5] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BACKGROUND Major depressive disorder (MDD) is highly debilitating, difficult to treat, has a high rate of recurrence, and negatively impacts the individual and society as a whole. One potential treatment for MDD is n-3 polyunsaturated fatty acids (n-3PUFAs), also known as omega-3 oils, naturally found in fatty fish, some other seafood, and some nuts and seeds. Various lines of evidence suggest a role for n-3PUFAs in MDD, but the evidence is far from conclusive. Reviews and meta-analyses clearly demonstrate heterogeneity between studies. Investigations of heterogeneity suggest different effects of n-3PUFAs, depending on the severity of depressive symptoms, where no effects of n-3PUFAs are found in studies of individuals with mild depressive symptomology, but possible benefit may be suggested in studies of individuals with more severe depressive symptomology. Hence it is important to establish their effectiveness in treating MDD. This review updates and incorporates an earlier review with the same research objective (Appleton 2015). OBJECTIVES To assess the effects of n-3 polyunsaturated fatty acids (also known as omega-3 fatty acids) versus a comparator (e.g. placebo, antidepressant treatment, standard care, no treatment, wait-list control) for major depressive disorder (MDD) in adults. SEARCH METHODS We searched the Cochrane Central Register of Controlled trials (CENTRAL), Ovid MEDLINE, Embase and PsycINFO together with trial registries and grey literature sources (to 9 January 2021). We checked reference lists and contacted authors of included studies for additional information when necessary. SELECTION CRITERIA We included studies in the review if they: used a randomised controlled trial design; provided n-3PUFAs as an intervention; used a comparator; measured depressive symptomology as an outcome; and were conducted in adults with MDD. Primary outcomes were depressive symptomology (continuous data collected using a validated rating scale) and adverse events. Secondary outcomes were depressive symptomology (dichotomous data on remission and response), quality of life, and non-completion of studies. DATA COLLECTION AND ANALYSIS We used standard methodological procedures as expected by Cochrane. We assessed the certainty of the evidence using GRADE criteria. MAIN RESULTS The review includes 35 relevant studies: 34 studies involving a total of 1924 participants investigated the impact of n-3PUFA supplementation compared to placebo, and one study involving 40 participants investigated the impact of n-3PUFA supplementation compared to antidepressant treatment. For the placebo comparison, n-3PUFA supplementation resulted in a small to modest benefit for depressive symptomology, compared to placebo: standardised mean difference (SMD) (random-effects model) -0.40 (95% confidence interval (CI) -0.64 to -0.16; 33 studies, 1848 participants; very low-certainty evidence), but this effect is unlikely to be clinically meaningful. An SMD of 0.40 represents a difference between groups in scores on the HDRS (17-item) of approximately 2.5 points (95% CI 1.0 to 4.0), where the minimal clinically important change score on this scale is 3.0 points. The confidence intervals include both a possible clinically important effect and a possible negligible effect, and there is considerable heterogeneity between studies. Sensitivity analyses, funnel plot inspection and comparison of our results with those of large well-conducted trials also suggest that this effect estimate may be biased towards a positive finding for n-3PUFAs. Although the numbers of individuals experiencing adverse events were similar in intervention and placebo groups (odds ratio (OR) 1.27, 95% CI 0.99 to 1.64; 24 studies, 1503 participants; very low-certainty evidence), the confidence intervals include a small decrease to a modest increase in adverse events with n-3PUFAs. There was no evidence for a difference between n-3PUFA and placebo groups in remission rates (OR 1.13, 95% CI 0.74 to 1.72; 8 studies, 609 participants, low-certainty evidence), response rates (OR 1.20, 95% CI 0.80 to 1.79; 17 studies, 794 participants; low-certainty evidence), quality of life (SMD -0.38 (95% CI -0.82 to 0.06), 12 studies, 476 participants, very low-certainty evidence), or trial non-completion (OR 0.92, 95% CI 0.70 to 1.22; 29 studies, 1777 participants, very low-certainty evidence). The evidence on which these results are based was also very limited, highly heterogeneous, and potentially biased. Only one study, involving 40 participants, was available for the antidepressant comparison. This study found no differences between treatment with n-3PUFAs and treatment with antidepressants in depressive symptomology (mean difference (MD) -0.70, 95% CI -5.88 to 4.48), rates of response to treatment (OR 1.23, 95% CI 0.35 to 4.31), or trial non-completion (OR 1.00, 95% CI 0.21 to 4.71). Confidence intervals are however very wide in all analyses, and do not rule out important beneficial or detrimental effects of n-3PUFAs compared to antidepressants. Adverse events were not reported in a manner suitable for analysis, and rates of depression remission and quality of life were not reported. AUTHORS' CONCLUSIONS At present, we do not have sufficient high-certainty evidence to determine the effects of n-3PUFAs as a treatment for MDD. Our primary analyses may suggest a small-to-modest, non-clinically beneficial effect of n-3PUFAs on depressive symptomology compared to placebo; however the estimate is imprecise, and we judged the certainty of the evidence on which this result is based to be low to very low. Our data may also suggest similar rates of adverse events and trial non-completion in n-3PUFA and placebo groups, but again our estimates are very imprecise. Effects of n-3PUFAs compared to antidepressants are very imprecise and uncertain. More complete evidence is required for both the potential positive and negative effects of n-3PUFAs for MDD.
Collapse
Affiliation(s)
| | - Philip D Voyias
- North Bristol NHS Trust, Bristol, UK
- University of Bristol, Bristol, UK
| | - Hannah M Sallis
- Centre for Academic Mental Health, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- MRC Integrative Epidemiology Unit (IEU) at the University of Bristol, Bristol, UK
- School of Psychological Science, University of Bristol, Bristol, UK
| | - Sarah Dawson
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Cochrane Common Mental Disorders, University of York, York, UK
| | - Andrew R Ness
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust and University of Bristol, Bristol, UK
| | - Rachel Churchill
- Cochrane Common Mental Disorders, University of York, York, UK
- Centre for Reviews and Dissemination, University of York, York, UK
| | - Rachel Perry
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust and University of Bristol, Bristol, UK
| |
Collapse
|