1
|
Martin-Garcia O, da Silva PHR, De Smet S, De Witte S, Brunoni AR, Vanderhasselt MA, Razza LB. Baseline gray matter volume associates with working memory performance after prefrontal transcranial direct current stimulation. Behav Brain Res 2025; 481:115416. [PMID: 39761752 DOI: 10.1016/j.bbr.2025.115416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 12/18/2024] [Accepted: 01/02/2025] [Indexed: 01/13/2025]
Abstract
Working memory is crucial for daily life and is often impaired in neuropsychiatric conditions. Attempts to enhance it using transcranial direct current stimulation (tDCS) have shown mixed results, possibly due to large inter-individual variability. This study assessed whether baseline regional brain volume was associated with working memory performance following tDCS. Healthy participants were randomly assigned to three bilateral tDCS protocols (sham, 1.5 mA, and 3 mA) over the dorsolateral prefrontal cortex (dlPFC) (anode-left, cathode-right) for 20 minutes, in a within-subjects design with a 2-week interval, followed by emotional and non-emotional 3-back tasks. Baseline volumetric data were used to extract gray matter volumes of defined regions of interest; the dlPFC, the medial PFC (mPFC), and the posterior cingulate cortex (PCC) bilaterally. Data from thirty-nine participants (69.2 % female, mean age: 24.56 years) across 112 tDCS sessions were analyzed. Findings revealed no significant association between working memory performance post sham-tDCS and gray matter volume. However, larger baseline cortical volumes across all regions were associated with slower reaction times and lower accuracy for the non-emotional task at 1.5 mA, whereas non-significant results were observed at 3 mA. For the emotional task, only a significant association for reaction time after 3 mA and left dlPFC and right PCC were found. Findings highlight not only the association between individual baseline gray matter, but also the impact of methodological choices, such as current intensity and outcome, on the effect of tDCS. Future research should aim to further explore individual variability and methodological factors to deepen our understanding of the mechanisms underlying tDCS.
Collapse
Affiliation(s)
- Oscar Martin-Garcia
- Department of Personality, Evaluation and Clinical Psychology, Complutense University of Madrid, Madrid, Spain.
| | - Pedro Henrique Rodrigues da Silva
- Departamento e Instituto de Psiquiatria, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil; Division of Neuropsychiatry and Neuromodulation, Massachusetts General Hospital, Harvard Medical School, Boston, USA.
| | - Stefanie De Smet
- Department of Head and Skin, Psychiatry and Medical Psychology, Ghent University Hospital, Ghent University, Ghent, Belgium; Ghent Experimental Psychiatry (GHEP) Lab, Ghent University, Ghent, Belgium; Brain Stimulation and Cognition (BSC) Lab, Department of Cognitive Neuroscience, Faculty of Psychology & Neuroscience, Maastricht University, Maastricht, the Netherlands.
| | - Sara De Witte
- Ghent Experimental Psychiatry (GHEP) Lab, Ghent University, Ghent, Belgium; Department of Neurology and Bru-BRAIN, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium; Neuroprotection and Neuromodulation (NEUR) Research Group, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Laarbeeklaan 103, Brussels, Belgium.
| | - Andre R Brunoni
- Departamento e Instituto de Psiquiatria, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil; Departamento de Clínica Médica, Faculdade de Medicina da Universidade de São Paulo & Hospital Universitário, Universidade de São Paulo, São Paulo, Brazil; Hospital Universitário, Universidade de São Paulo, São Paulo, Brazil.
| | - Marie-Anne Vanderhasselt
- Department of Head and Skin, Psychiatry and Medical Psychology, Ghent University Hospital, Ghent University, Ghent, Belgium; Ghent Experimental Psychiatry (GHEP) Lab, Ghent University, Ghent, Belgium.
| | - Lais B Razza
- Department of Head and Skin, Psychiatry and Medical Psychology, Ghent University Hospital, Ghent University, Ghent, Belgium; Ghent Experimental Psychiatry (GHEP) Lab, Ghent University, Ghent, Belgium.
| |
Collapse
|
2
|
Cabreira V, Alty J, Antic S, Araujo R, Aybek S, Ball HA, Baslet G, Bhome R, Coebergh J, Dubois B, Edwards M, Filipovic SR, Frederiksen KS, Harbo T, Hayhow B, Howard R, Huntley J, Isaacs JD, LaFrance C, Larner A, Di Lorenzo F, Main J, Mallam E, Marra C, Massano J, McGrath ER, Portela Moreira I, Nobili F, Pal S, Pennington CM, Tábuas-Pereira M, Perez D, Popkirov S, Rayment D, Rossor M, Russo M, Santana I, Schott J, Scott EP, Taipa R, Teodoro T, Tinazzi M, Tomic S, Toniolo S, Tørring CW, Wilkinson T, Zeidler M, Frostholm L, McWhirter L, Stone J, Carson A. Development of a diagnostic checklist to identify functional cognitive disorder versus other neurocognitive disorders. BMJ Neurol Open 2025; 7:e000918. [PMID: 40034653 PMCID: PMC11873336 DOI: 10.1136/bmjno-2024-000918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 01/31/2025] [Indexed: 03/05/2025] Open
Abstract
Background Functional cognitive disorder (FCD) poses a diagnostic challenge due to its resemblance to other neurocognitive disorders and limited biomarker accuracy. We aimed to develop a new diagnostic checklist to identify FCD versus other neurocognitive disorders. Methods The clinical checklist was developed through mixed methods: (1) a literature review, (2) a three-round Delphi study with 45 clinicians from 12 countries and (3) a pilot discriminative accuracy study in consecutive patients attending seven memory services across the UK. Items gathering consensus were incorporated into a pilot checklist. Item redundancy was evaluated with phi coefficients. A briefer checklist was produced by removing items with >10% missing data. Internal validity was tested using Cronbach's alpha. Optimal cut-off scores were determined using receiver operating characteristic curve analysis. Results A full 11-item checklist and a 7-item briefer checklist were produced. Overall, 239 patients (143 FCD, 96 non-FCD diagnoses) were included. The checklist scores were significantly different across subgroups (FCD and other neurocognitive disorders) (F(2, 236)=313.3, p<0.001). The area under the curve was excellent for both the full checklist (0.97, 95% CI 0.95 to 0.99) and its brief version (0.96, 95% CI 0.93 to 0.98). Optimal cut-off scores corresponded to a specificity of 97% and positive predictive value of 91% for identifying FCD. Both versions showed good internal validity (>0.80). Conclusions This pilot study shows that a brief clinical checklist may serve as a quick complementary tool to differentiate patients with neurodegeneration from those with FCD. Prospective blind large-scale validation in diverse populations is warranted.Cite Now.
Collapse
Affiliation(s)
- Verónica Cabreira
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Jane Alty
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - Sonja Antic
- Neurology, Aarhus Universitetshospital, Aarhus, Denmark
| | - Rui Araujo
- Neurology, Centro Hospitalar Universitario de Sao Joao, Porto, Portugal
- Clinical Neurosciences and Mental Health, University of Porto Faculty of Medicine, Porto, Portugal
| | - Selma Aybek
- Neurology, University of Fribourg Faculty of Science and Medicine, Fribourg, Switzerland
| | - Harriet A Ball
- University of Bristol Faculty of Health Sciences, Bristol Medical School, Bristol, UK
| | - Gaston Baslet
- Psychiatry, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Rohan Bhome
- Dementia Research Centre, University College London, London, UK
- Centre for Medical Image Computing, University College London, London, UK
| | - Jan Coebergh
- St George’s University of London, London, London, UK
| | - Bruno Dubois
- Department of Neurology, Institut de la mémoire et de la maladie d’Alzheimer, Centre de Référence ‘Démences Rares’, Hôpital de la Pitié-Salpêtrière, AP-HP, Paris, France
- ICM-INSERM 1127, FrontLab, Institut du Cerveau et de la Moelle Epinière (ICM), Paris, France
| | - Mark Edwards
- Department of Basic and Clinical Neuroscience, King’s College London Institute of Psychiatry Psychology and Neuroscience, London, UK
| | - Sasa R Filipovic
- Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Kristian Steen Frederiksen
- Clinical Trial Unit, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Kobenhavn, Denmark
| | - Thomas Harbo
- Neurology, Aarhus Universitetshospital, Aarhus, Denmark
| | - Bradleigh Hayhow
- Neurology, Fiona Stanley Hospital, Murdoch, Western Australia, Australia
- School of Medicine, The University of Notre Dame Australia, Perth, Western Australia, Australia
| | - Robert Howard
- Division of Psychiatry, University College London, London, UK
| | - Jonathan Huntley
- Division of Psychiatry, University College London, London, UK
- Camden and Islington NHS Foundation Trust, London, UK
| | | | - Curt LaFrance
- Alpert Medical School Area Health Education Centre, Providence, Rhode Island, USA
- Neuropsychiatry and Behavioral Neurology, Rhode Island Hospital, Providence, Rhode Island, USA
| | - Andrew Larner
- Cognitive Function Clinic, Walton Centre for Neurology and Neurosurgery, Liverpool, UK
| | - Francesco Di Lorenzo
- Department of Clinical and Behavioural Neurology, Fondazione Santa Lucia Istituto di Ricovero e Cura a Carattere Scientifico, Roma, Italy
| | - James Main
- Bristol Dementia Wellbeing Service, Devon Partnership NHS Trust, Bristol, UK
| | | | - Camillo Marra
- Universita Cattolica del Sacro Cuore Sede di Roma, Roma, Italy
| | - João Massano
- Neurology, Centro Hospitalar Universitario de Sao Joao, Porto, Portugal
- Clinical Neurosciences and Mental Health, University of Porto Faculty of Medicine, Porto, Portugal
| | - Emer R McGrath
- University of Galway School of Medicine, Galway, Ireland
| | - Isabel Portela Moreira
- Neurology Department, Private Hospital of Gaia of the Trofa Saúde Group, Vila Nova de Gaia, Portugal
| | - Flavio Nobili
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Suvankar Pal
- Anne Rowling Regenerative Neurology Clinic, University of Edinburgh, Edinburgh, UK
- Neurology, NHS Forth Valley, Stirling, UK
| | - Catherine M Pennington
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- Neurology, NHS Forth Valley, Stirling, UK
| | - Miguel Tábuas-Pereira
- Neurology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- University of Coimbra Faculty of Medicine, Coimbra, Portugal
| | - David Perez
- Neurology and Psychiatry, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Stoyan Popkirov
- Department of Neurology, University Hospital Essen, Essen, Germany
| | - Dane Rayment
- Rosa Burden Centre for Neuropsychiatry, Southmead Hospital, Bristol, UK
| | - Martin Rossor
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Mirella Russo
- Department of Sciences, Department of Neuroscience, Imaging and Clinical Sciences, Gabriele d’Annunzio University of Chieti and Pescara, Chieti, Italy
| | - Isabel Santana
- Neurology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Jonathan Schott
- Dementia Research Centre, Institute of Neurology, London, UK
| | - Emmi P Scott
- Medical University of South Carolina, Charleston, South Carolina, USA
| | - Ricardo Taipa
- Neuropathology Unit, Centro Hospitalar Universitário de Santo António, Porto, Portugal
| | - Tiago Teodoro
- Neurology, St George’s University of London, London, UK
| | - Michele Tinazzi
- Department of Neurosciences, Biomedicine and Movement, University of Verona, Verona, Italy
| | | | - Sofia Toniolo
- Cognitive Disorder Clinic, Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, UK
| | | | - Tim Wilkinson
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | | | - Lisbeth Frostholm
- Department of Clinical Medicine, Aarhus Universitetshospital, Aarhus, Denmark
- Department of Functional Disorders and Psychosomatics, Aarhus Universitetshospital, Aarhus, Denmark
| | - Laura McWhirter
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Jon Stone
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Alan Carson
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
3
|
Niu C, Jiang Y, Li Y, Wang X, Zhao H, Cheng Z, Li X, Zhang X, Liu Z, Yu X, Peng Y. A network analysis of the heterogeneity and associated risk and protective factors of depression and anxiety among college students. Sci Rep 2025; 15:6699. [PMID: 40000716 PMCID: PMC11861700 DOI: 10.1038/s41598-025-91025-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 02/18/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Comorbidity of depression and anxiety is common among adolescents and can lead to adverse outcomes. However, there is limited understanding of the latent characteristics and mechanisms governing these disorders and their interactions. Moreover, few studies have examined the impacts of relevant risk and protective factors. METHODS This cross-sectional study involved 1,719 students. Mplus 8.0 software was used to conduct latent profile analysis to explore the potential categories of depression and anxiety comorbidities. R4.3.2 software was used to explore the network of core depression and anxiety symptoms, bridge these disorders, and evaluate the effects of risk and protective factors. RESULTS Three categories were established: "healthy" (57.8%), "mild depression-mild anxiety" (36.6%), and "moderately severe depression-moderate anxiety" (5.6%). "Depressed mood", "nervousness", and "difficulty relaxing" were core symptoms in both the depression-anxiety comorbidity network and the network of risk and protective factors. Stress perception and neuroticism serve as bridging nodes connecting some symptoms of depression and anxiety and are thus considered the most prominent risk factors. CONCLUSIONS According to the core and bridging symptoms identified in this study, targeted intervention and treatment can be provided to groups with comorbid depression and anxiety, thereby reducing the risk of these comorbidities in adolescents.
Collapse
Affiliation(s)
- Chunjuan Niu
- School of Psychology and Mental Health, Hebei Key Laboratory of Mental Health and Brain Science, North China University of Science and Technology, Tangshan, China
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang, Malaysia
| | - Yaye Jiang
- School of Psychology and Mental Health, Hebei Key Laboratory of Mental Health and Brain Science, North China University of Science and Technology, Tangshan, China
| | - Yihui Li
- School of Psychology and Mental Health, Hebei Key Laboratory of Mental Health and Brain Science, North China University of Science and Technology, Tangshan, China
| | - Xudong Wang
- School of Psychology and Mental Health, Hebei Key Laboratory of Mental Health and Brain Science, North China University of Science and Technology, Tangshan, China
| | - Huiyuan Zhao
- School of Psychology and Mental Health, Hebei Key Laboratory of Mental Health and Brain Science, North China University of Science and Technology, Tangshan, China
| | - Zhengshu Cheng
- School of Psychology and Mental Health, Hebei Key Laboratory of Mental Health and Brain Science, North China University of Science and Technology, Tangshan, China
| | - Xiaoran Li
- School of Psychology and Mental Health, Hebei Key Laboratory of Mental Health and Brain Science, North China University of Science and Technology, Tangshan, China
| | - Xu Zhang
- School of Psychology and Mental Health, Hebei Key Laboratory of Mental Health and Brain Science, North China University of Science and Technology, Tangshan, China
| | - Zhiwei Liu
- School of Psychology and Mental Health, Hebei Key Laboratory of Mental Health and Brain Science, North China University of Science and Technology, Tangshan, China
| | - Xiaoyu Yu
- School of Psychology and Mental Health, Hebei Key Laboratory of Mental Health and Brain Science, North China University of Science and Technology, Tangshan, China
| | - Yan Peng
- School of Psychology and Mental Health, Hebei Key Laboratory of Mental Health and Brain Science, North China University of Science and Technology, Tangshan, China.
- North China University of Science and Technology Affiliated Hospital, Tangshan, China.
- School of Social Sciences, Universiti Sains Malaysia, Penang, Malaysia.
| |
Collapse
|
4
|
Mattoni M, Fisher AJ, Gates KM, Chein J, Olino TM. Group-to-individual generalizability and individual-level inferences in cognitive neuroscience. Neurosci Biobehav Rev 2025; 169:106024. [PMID: 39889869 PMCID: PMC11835466 DOI: 10.1016/j.neubiorev.2025.106024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/14/2025] [Accepted: 01/21/2025] [Indexed: 02/03/2025]
Abstract
Much of cognitive neuroscience research is focused on group-averages and interindividual brain-behavior associations. However, many theories core to the goal of cognitive neuroscience, such as hypothesized neural mechanisms for a behavior, are inherently based on intraindividual processes. To accommodate this mismatch between study design and theory, research frequently relies on an implicit assumption that group-level, between-person inferences extend to individual-level, within-person processes. The assumption of group-to-individual generalizability, formally referred to as ergodicity, requires that a process be both homogenous within a population and stationary within individuals over time. Our goal in this review is to assess this assumption and provide an accessible introduction to idiographic science (study of the individual) for the cognitive neuroscientist, ultimately laying a foundation for increased focus on the study of intraindividual processes. We first review the history of idiographic science in psychology to connect this longstanding literature with recent individual-level research goals in cognitive neuroscience. We then consider two requirements of group-to-individual generalizability, pattern homogeneity and stationarity, and suggest that most processes in cognitive neuroscience do not meet these assumptions. Consequently, interindividual findings are inappropriate for the intraindividual inferences that many theories are based on. To address this challenge, we suggest precision imaging as an ideal path forward for intraindividual study and present a research framework for complementary interindividual and intraindividual study.
Collapse
Affiliation(s)
- Matthew Mattoni
- Temple University, Department of Psychology and Neuroscience, 1801 N Broad St., Philadelphia, PA, USA.
| | - Aaron J Fisher
- University of California-Berkeley, Department of Psychology, 2121 Berkeley Way, Berkeley, CA, USA
| | - Kathleen M Gates
- University of North Carolina at Chapel Hill, Department of Psychology and Neuroscience, 235 E. Cameron Avenue, Chapel Hill, NC, USA
| | - Jason Chein
- Temple University, Department of Psychology and Neuroscience, 1801 N Broad St., Philadelphia, PA, USA
| | - Thomas M Olino
- Temple University, Department of Psychology and Neuroscience, 1801 N Broad St., Philadelphia, PA, USA
| |
Collapse
|
5
|
Cabreira V, Alty J, Antic S, Araújo R, Aybek S, Ball HA, Baslet G, Bhome R, Coebergh J, Dubois B, Edwards M, Filipović SR, Frederiksen KS, Harbo T, Hayhow B, Howard R, Huntley J, Isaacs J, LaFrance WC, Larner AJ, Di Lorenzo F, Main J, Mallam E, Marra C, Massano J, McGrath ER, McWhirter L, Moreira IP, Nobili F, Pennington C, Tábuas‐Pereira M, Perez DL, Popkirov S, Rayment D, Rossor M, Russo M, Santana I, Schott J, Scott EP, Taipa R, Tinazzi M, Tomic S, Toniolo S, Tørring CW, Wilkinson T, Frostholm L, Stone J, Carson A. Perspectives on the diagnosis and management of functional cognitive disorder: An international Delphi study. Eur J Neurol 2025; 32:e16318. [PMID: 38700361 PMCID: PMC11617961 DOI: 10.1111/ene.16318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 03/18/2024] [Accepted: 04/11/2024] [Indexed: 05/05/2024]
Abstract
BACKGROUND Current proposed criteria for functional cognitive disorder (FCD) have not been externally validated. We sought to analyse the current perspectives of cognitive specialists in the diagnosis and management of FCD in comparison with neurodegenerative conditions. METHODS International experts in cognitive disorders were invited to assess seven illustrative clinical vignettes containing history and bedside characteristics alone. Participants assigned a probable diagnosis and selected the appropriate investigation and treatment. Qualitative, quantitative and inter-rater agreement analyses were undertaken. RESULTS Eighteen diagnostic terminologies were assigned by 45 cognitive experts from 12 countries with a median of 13 years of experience, across the seven scenarios. Accurate discrimination between FCD and neurodegeneration was observed, independently of background and years of experience: 100% of the neurodegenerative vignettes were correctly classified and 75%-88% of the FCD diagnoses were attributed to non-neurodegenerative causes. There was <50% agreement in the terminology used for FCD, in comparison with 87%-92% agreement for neurodegenerative syndromes. Blood tests and neuropsychological evaluation were the leading diagnostic modalities for FCD. Diagnostic communication, psychotherapy and psychiatry referral were the main suggested management strategies in FCD. CONCLUSIONS Our study demonstrates the feasibility of distinguishing between FCD and neurodegeneration based on relevant patient characteristics and history details. These characteristics need further validation and operationalisation. Heterogeneous labelling and framing pose clinical and research challenges reflecting a lack of agreement in the field. Careful consideration of FCD diagnosis is advised, particularly in the presence of comorbidities. This study informs future research on diagnostic tools and evidence-based interventions.
Collapse
Affiliation(s)
- Verónica Cabreira
- Centre for Clinical Brain SciencesUniversity of EdinburghEdinburghUK
| | - Jane Alty
- Wicking Dementia Research and Education CentreUniversity of TasmaniaHobartTasmaniaAustralia
| | - Sonja Antic
- Department of NeurologyAarhus University HospitalAarhusDenmark
| | - Rui Araújo
- Department of NeurologyCentro Hospitalar Universitário São JoãoPortoPortugal
- Department of Clinical Neurosciences and Mental HealthFaculty of Medicine University of PortoPortoPortugal
| | - Selma Aybek
- Neurology, Faculty of Sciences and MedicineFribourg UniversityFribourgSwitzerland
| | | | - Gaston Baslet
- Department of PsychiatryBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Rohan Bhome
- Dementia Research CentreUCL Queen Square Institute of NeurologyLondonUK
- Centre for Medical Image ComputingUniversity College LondonLondonUK
| | - Jan Coebergh
- Department of NeurologySt George's University of LondonLondonUK
| | - Bruno Dubois
- Department of NeurologyInstitut de la mémoire et de la maladie d'Alzheimer (IM2A), AP‐HP, Brain Institute, Sorbonne UniversityParisFrance
| | - Mark Edwards
- Department of Basic and Clinical NeurosciencesInstitute of Psychiatry Psychology and Neurosciences, Kings College LondonLondonUK
| | | | - Kristian Steen Frederiksen
- Clinical Trial Unit, RigshospitaletUniversity of CopenhagenCopenhagenDenmark
- Department of Clinical MedicineUniversity of CopenhagenCopenhagenDenmark
| | - Thomas Harbo
- Department of NeurologyAarhus University HospitalAarhusDenmark
| | - Bradleigh Hayhow
- Department of NeurologyFiona Stanley HospitalMurdochWestern AustraliaAustralia
- School of MedicineUniversity of Notre Dame AustraliaFremantleWestern AustraliaAustralia
| | - Robert Howard
- Division of PsychiatryUniversity College LondonLondonUK
| | - Jonathan Huntley
- Division of PsychiatryUniversity College LondonLondonUK
- Camden and Islington NHS Foundation TrustLondonUK
| | - Jeremy Isaacs
- Department of NeurologySt George's University of LondonLondonUK
| | - William Curt LaFrance
- Alpert Medical SchoolBrown UniversityProvidenceRhode IslandUSA
- Neuropsychiatry and Behavioral NeurologyRhode Island HospitalProvidenceRhode IslandUSA
| | - Andrew J. Larner
- Cognitive Function ClinicWalton Centre for Neurology and NeurosurgeryLiverpoolUK
| | - Francesco Di Lorenzo
- Department of Clinical and Behavioural NeurologySanta Lucia Foundation IRCCSRomeItaly
| | - James Main
- Bristol Dementia Wellbeing Service, Devon Partnership NHS TrustBristolUK
| | | | - Camillo Marra
- Department of NeuroscienceCatholic University of the Sacred Heart, Memory Clinic ‐ Fondazione Policlinico Agostino Gemelli IRCCSRomeItaly
| | - João Massano
- Department of NeurologyCentro Hospitalar Universitário São JoãoPortoPortugal
- Department of Clinical Neurosciences and Mental HealthFaculty of Medicine University of PortoPortoPortugal
| | | | - Laura McWhirter
- Centre for Clinical Brain SciencesUniversity of EdinburghEdinburghUK
| | - Isabel Portela Moreira
- Neurology DepartmentPrivate Hospital of Gaia of the Trofa Saúde GroupVila Nova de GaiaPortugal
| | | | - Catherine Pennington
- Clinical LecturerUniversity of EdinburghEdinburghUK
- Neurology DepartmentNHS Forth ValleyLarbertUK
- Department of Clinical NeurosciencesNHS LothianEdinburghUK
| | - Miguel Tábuas‐Pereira
- Neurology DepartmentCentro Hospitalar e Universitário de Coimbra, Praceta Prof. Mota PintoCoimbraPortugal
- Faculty of MedicineUniversity of CoimbraCoimbraPortugal
- Center for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
| | - David L. Perez
- Department of Neurology and Psychiatry, Massachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Stoyan Popkirov
- Department of NeurologyUniversity Hospital EssenEssenGermany
| | - Dane Rayment
- Rosa Burden Centre for NeuropsychiatrySouthmead HospitalBristolUK
| | - Martin Rossor
- Dementia Research CentreUCL Queen Square Institute of NeurologyLondonUK
| | - Mirella Russo
- Department of NeuroscienceImaging and Clinical Sciences G. d'Annunzio University of Chieti‐PescaraChietiItaly
| | - Isabel Santana
- Faculty of MedicineUniversity of CoimbraCoimbraPortugal
- Center for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
| | - Jonathan Schott
- Dementia Research CentreUCL Queen Square Institute of NeurologyLondonUK
| | - Emmi P. Scott
- Medical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Ricardo Taipa
- Neuropathology DepartmentCentro Hospitalar Universitário de Santo AntónioPortoPortugal
| | - Michele Tinazzi
- Department of Neurosciences, Biomedicine and MovementUniversity of VeronaVeronaItaly
| | - Svetlana Tomic
- Department of NeurologyUniversity Hospital Center Osijek, Medical School on University of OsijekOsijekCroatia
| | - Sofia Toniolo
- Cognitive Disorder Clinic, Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | | | - Tim Wilkinson
- Centre for Clinical Brain SciencesUniversity of EdinburghEdinburghUK
| | - Lisbeth Frostholm
- Department of Clinical MedicineAarhus University HospitalAarhusDenmark
- Department of Functional Disorders and PsychosomaticsAarhus University HospitalAarhusDenmark
| | - Jon Stone
- Centre for Clinical Brain SciencesUniversity of EdinburghEdinburghUK
| | - Alan Carson
- Centre for Clinical Brain SciencesUniversity of EdinburghEdinburghUK
| |
Collapse
|
6
|
Sindermann L, Dannlowski U, Leehr EJ, Hahn T, Grotegerd D, Meinert S, Lemke H, Winter A, Thiel K, Flinkenflügel K, Kircher T, Nenadić I, Straube B, Alexander N, Jamalabadi H, Jansen A, Stein F, Brosch K, Thomas-Odenthal F, Usemann P, Teutenberg L, Krug A, Andlauer TFM, David FS, Federmann LM, Beins E, Nöthen MM, Forstner AJ. Neurobiological correlates of comorbidity in disorders across the affective disorders-psychosis spectrum. J Psychiatr Res 2024; 180:462-472. [PMID: 39541637 DOI: 10.1016/j.jpsychires.2024.09.052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 09/18/2024] [Accepted: 09/30/2024] [Indexed: 11/16/2024]
Abstract
Disorders across the affective disorders-psychosis spectrum such as major depressive disorder (MDD), bipolar disorder (BD), schizoaffective disorder (SCA), and schizophrenia (SCZ), have overlapping symptomatology and high comorbidity rates with other mental disorders. So far, however, it is largely unclear why some of the patients develop comorbidities. In particular, the specific genetic architecture of comorbidity and its relationship with brain structure remain poorly understood. Therefore, we performed systematic analyses of clinical, genetics and brain structural measures to gain further insights into the neurobiological correlates of mental disorder's comorbidity. We investigated a sub-sample of the Marburg/Münster Cohort Study (MACS), comprising DSM-IV-TR diagnosed patients with a single disorder in the affective disorders-psychosis spectrum (SD, n = 470, MDD; BD; SCA; SCZ), with additional mental disorder's comorbidities (COM, n = 310), and healthy controls (HC, n = 649). We investigated group differences regarding a) the global severity index (based on SCL90-R), b) a cross-disorder polygenic risk score (PRS) calculated with PRS-continuous shrinkage (PRS-CS) using the summary statistics of a large genome-wide association study across mental disorders, and c) whole brain grey matter volume (GMV). The SCL90-R score significantly differed between groups (COM > SD > HC). While SD and COM did not differ in cross-disorder PRS and GMV, SD and COM versus HC displayed increased cross-disorder PRS and decreased GMV in the bilateral insula, the left middle temporal, the left inferior parietal, and several frontal gyri. Our results thus suggest that disorders in the affective disorders-psychosis spectrum with or without additional comorbidities differ in self-reported clinical data, but not on genetic or brain structural levels.
Collapse
Affiliation(s)
- Lisa Sindermann
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany; Institute for Translational Psychiatry, University Münster, Münster, Germany; Department of Psychiatry and Psychotherapy, University Hospital Bonn, Bonn, Germany.
| | - Udo Dannlowski
- Institute for Translational Psychiatry, University Münster, Münster, Germany
| | - Elisabeth J Leehr
- Institute for Translational Psychiatry, University Münster, Münster, Germany
| | - Tim Hahn
- Institute for Translational Psychiatry, University Münster, Münster, Germany
| | - Dominik Grotegerd
- Institute for Translational Psychiatry, University Münster, Münster, Germany
| | - Susanne Meinert
- Institute for Translational Psychiatry, University Münster, Münster, Germany; Institute for Translational Neuroscience, University Münster, Münster, Germany
| | - Hannah Lemke
- Institute for Translational Psychiatry, University Münster, Münster, Germany
| | - Alexandra Winter
- Institute for Translational Psychiatry, University Münster, Münster, Germany
| | - Katharina Thiel
- Institute for Translational Psychiatry, University Münster, Münster, Germany
| | - Kira Flinkenflügel
- Institute for Translational Psychiatry, University Münster, Münster, Germany
| | - Tilo Kircher
- Department of Psychiatry and Psychotherapy, Philipps-University and University Hospital Marburg, Marburg, Germany; Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University Giessen, Germany
| | - Igor Nenadić
- Department of Psychiatry and Psychotherapy, Philipps-University and University Hospital Marburg, Marburg, Germany; Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University Giessen, Germany
| | - Benjamin Straube
- Department of Psychiatry and Psychotherapy, Philipps-University and University Hospital Marburg, Marburg, Germany; Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University Giessen, Germany
| | - Nina Alexander
- Department of Psychiatry and Psychotherapy, Philipps-University and University Hospital Marburg, Marburg, Germany; Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University Giessen, Germany
| | - Hamidreza Jamalabadi
- Department of Psychiatry and Psychotherapy, Philipps-University and University Hospital Marburg, Marburg, Germany; Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University Giessen, Germany
| | - Andreas Jansen
- Department of Psychiatry and Psychotherapy, Philipps-University and University Hospital Marburg, Marburg, Germany; Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University Giessen, Germany
| | - Frederike Stein
- Department of Psychiatry and Psychotherapy, Philipps-University and University Hospital Marburg, Marburg, Germany; Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University Giessen, Germany
| | - Katharina Brosch
- Department of Psychiatry and Psychotherapy, Philipps-University and University Hospital Marburg, Marburg, Germany; Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University Giessen, Germany; Institute of Behavioral Science, Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Florian Thomas-Odenthal
- Department of Psychiatry and Psychotherapy, Philipps-University and University Hospital Marburg, Marburg, Germany; Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University Giessen, Germany
| | - Paula Usemann
- Department of Psychiatry and Psychotherapy, Philipps-University and University Hospital Marburg, Marburg, Germany; Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University Giessen, Germany
| | - Lea Teutenberg
- Department of Psychiatry and Psychotherapy, Philipps-University and University Hospital Marburg, Marburg, Germany; Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University Giessen, Germany
| | - Axel Krug
- Department of Psychiatry and Psychotherapy, University Hospital Bonn, Bonn, Germany; Institute for Translational Neuroscience, University Münster, Münster, Germany
| | - Till F M Andlauer
- Department of Neurology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Friederike S David
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Lydia M Federmann
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany; Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany
| | - Eva Beins
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Markus M Nöthen
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Andreas J Forstner
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany; Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany; Centre for Human Genetics, Philipps-University Marburg, Marburg, Germany.
| |
Collapse
|
7
|
Niu Z, Haley AP, Clark AL, Duarte A. Age exacerbates the negative effect of depression on executive functioning in racial and ethnic minorities. Brain Imaging Behav 2024; 18:1064-1074. [PMID: 38850388 PMCID: PMC11582303 DOI: 10.1007/s11682-024-00898-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2024] [Indexed: 06/10/2024]
Abstract
Age and depression may interact to produce a "double jeopardy" for cognitive impairment, and executive functioning, in cognitively unimpaired aging. Few studies have considered middle age or the ethnoracial diversity of subjects, despite evidence of more severe cognitive outcomes in historically minoritized people. In this pilot study, we investigated the impact of age on depression-related cognitive impairment and the underlying brain volumes in middle-aged non-Hispanic White adults (116), and Hispanic and Black adults (60), with a total number of 176 adults. The result shows a significant interaction between age and depression for executive functioning, specifically for middle-aged Hispanic and Black adults, but not non-Hispanic White adults. Prefrontal cortex volumes, which were reduced in the Black and Hispanic compared to the non-Hispanic White adults, partially mediated the relationship between depression level and executive functioning, across age and ethnoracial group. Collectively, these results suggest that the negative impact of depression on executive functioning and Prefrontal cortex volumes integrity may be exacerbated by age and that historically minoritized people may be particularly sensitive to this double jeopardy.
Collapse
Affiliation(s)
- Zhimei Niu
- Department of Psychology, University of Texas at Austin, 108 Dean Keeton Street, Austin, TX, 78712, USA.
| | - Andreana P Haley
- Department of Psychology, University of Texas at Austin, 108 Dean Keeton Street, Austin, TX, 78712, USA
| | - Alexandra L Clark
- Department of Psychology, University of Texas at Austin, 108 Dean Keeton Street, Austin, TX, 78712, USA
| | - Audrey Duarte
- Department of Psychology, University of Texas at Austin, 108 Dean Keeton Street, Austin, TX, 78712, USA
| |
Collapse
|
8
|
Noor AAM. Exploring the Therapeutic Potential of Terpenoids for Depression and Anxiety. Chem Biodivers 2024; 21:e202400788. [PMID: 38934531 DOI: 10.1002/cbdv.202400788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 06/19/2024] [Accepted: 06/19/2024] [Indexed: 06/28/2024]
Abstract
This review focus on the terpenoids as potential therapeutic agents for depression and anxiety disorders, which naturally found in a variety of plants and exhibit a wide range of biological activities. Among the terpenoids discussed in this review are α-pinene, β-caryophyllene, α-phellandrene, limonene, β-linalool, 1, 8-cineole, β-pinene, caryophyllene oxide, p-cymene, and eugenol. All of these compounds have been studied extensively regarding their pharmacological properties, such as neuroprotective effect, anti-inflammation, antibacterial, regulation of neurotransmitters and antioxidant effect. Preclinical evidence are reviewed to highlight their diverse mechanisms of action and therapeutic potential to support antidepressant and anxiolytic properties. Additionally, challenges and future directions are also discussed to emphasize therapeutic utility of terpenoids for mental health disorders. Overall, this review provides a promising role of terpenoids as novel therapeutic agents for depression and anxiety, with potential implications for the development of more effective and well-tolerated treatments in the field of psychopharmacology.
Collapse
Affiliation(s)
- Arif Azimi Md Noor
- Harvard Medical School, Department of Biomedical Informatics, 10 Shattuck Street Suite 514, Boston MA, 02115, United States of America
- Eyes Specialist Clinic, Raja Perempuan Zainab 2 Hospital, 15586, Kota Bharu, Kelantan, Malaysia
| |
Collapse
|
9
|
Karim HT, Lee S, Gerlach A, Stinley M, Berta R, Mahbubani R, Tudorascu DL, Butters MA, Gross JJ, Andreescu C. Hippocampal subfield volume in older adults with and without mild cognitive impairment: Effects of worry and cognitive reappraisal. Neurobiol Aging 2024; 141:55-65. [PMID: 38823204 PMCID: PMC11246796 DOI: 10.1016/j.neurobiolaging.2024.02.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 02/16/2024] [Accepted: 02/19/2024] [Indexed: 06/03/2024]
Abstract
Studies have confirmed that anxiety, especially worry and rumination, are associated with increased risk for cognitive decline, including Alzheimer's disease and related dementias (ADRD). Hippocampal atrophy is a hallmark of ADRD. We investigated the association between hippocampus and its subfield volumes and late-life global anxiety, worry, and rumination, and emotion regulation strategies. We recruited 110 participants with varying worry severity who underwent magnetic resonance imaging and clinical interviews. We conducted cross-sectional regression analysis between each subfield and anxiety, worry, rumination, reappraisal, and suppression while adjusting for age, sex, race, education, cumulative illness burden, stress, neuroticism, and intracranial volume. We imputed missing data and corrected for multiple comparisons across regions. Greater worry was associated with smaller subiculum volume, whereas greater use of reappraisal was associated with larger subiculum and CA1 volume. Greater worry may be detrimental to the hippocampus and to subfields involved in early ADRD pathology. Use of reappraisal appears protective of hippocampal structure. Worry and reappraisal may be modifiable targets for ADRD prevention.
Collapse
Affiliation(s)
- Helmet T Karim
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
| | - Soyoung Lee
- Department of Psychiatry, University of Maryland, Baltimore, MD, United States
| | - Andrew Gerlach
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
| | - Mark Stinley
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
| | - Rachel Berta
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
| | - Rebecca Mahbubani
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
| | - Dana L Tudorascu
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States; Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, United States
| | - Meryl A Butters
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
| | - James J Gross
- Department of Psychology, Stanford University, Stanford, CA, United States
| | - Carmen Andreescu
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States.
| |
Collapse
|
10
|
Liu W, Su JP, Zeng LL, Shen H, Hu DW. Gene expression and brain imaging association study reveals gene signatures in major depressive disorder. Brain Commun 2024; 6:fcae258. [PMID: 39185029 PMCID: PMC11342243 DOI: 10.1093/braincomms/fcae258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 06/03/2024] [Accepted: 08/09/2024] [Indexed: 08/27/2024] Open
Abstract
Major depressive disorder is often characterized by changes in the structure and function of the brain, which are influenced by modifications in gene expression profiles. How the depression-related genes work together within the scope of time and space to cause pathological changes remains unclear. By integrating the brain-wide gene expression data and imaging data in major depressive disorder, we identified gene signatures of major depressive disorder and explored their temporal-spatial expression specificity, network properties, function annotations and sex differences systematically. Based on correlation analysis with permutation testing, we found 345 depression-related genes significantly correlated with functional and structural alteration of brain images in major depressive disorder and separated them by directional effects. The genes with negative effect for grey matter density and positive effect for functional indices are enriched in downregulated genes in the post-mortem brain samples of patients with depression and risk genes identified by genome-wide association studies than genes with positive effect for grey matter density and negative effect for functional indices and control genes, confirming their potential association with major depressive disorder. By introducing a parameter of dispersion measure on the gene expression data of developing human brains, we revealed higher spatial specificity and lower temporal specificity of depression-related genes than control genes. Meanwhile, we found depression-related genes tend to be more highly expressed in females than males, which may contribute to the difference in incidence rate between male and female patients. In general, we found the genes with negative effect have lower network degree, more specialized function, higher spatial specificity, lower temporal specificity and more sex differences than genes with positive effect, indicating they may play different roles in the occurrence and development of major depressive disorder. These findings can enhance the understanding of molecular mechanisms underlying major depressive disorder and help develop tailored diagnostic and treatment strategies for patients of depression of different sex.
Collapse
Affiliation(s)
- Wei Liu
- College of Intelligence Science and Technology, National University of Defense Technology, Changsha, Hunan 410073, P.R. China
| | - Jian-Po Su
- College of Intelligence Science and Technology, National University of Defense Technology, Changsha, Hunan 410073, P.R. China
| | - Ling-Li Zeng
- College of Intelligence Science and Technology, National University of Defense Technology, Changsha, Hunan 410073, P.R. China
| | - Hui Shen
- College of Intelligence Science and Technology, National University of Defense Technology, Changsha, Hunan 410073, P.R. China
| | - De-Wen Hu
- College of Intelligence Science and Technology, National University of Defense Technology, Changsha, Hunan 410073, P.R. China
| |
Collapse
|
11
|
Hilbert K, Boeken OJ, Langhammer T, Groenewold NA, Bas-Hoogendam JM, Aghajani M, Zugman A, Åhs F, Arolt V, Beesdo-Baum K, Björkstrand J, Blackford JU, Blanco-Hinojo L, Böhnlein J, Bülow R, Cano M, Cardoner N, Caseras X, Dannlowski U, Domschke K, Fehm L, Feola B, Fredrikson M, Goossens L, Grabe HJ, Grotegerd D, Gur RE, Hamm AO, Harrewijn A, Heinig I, Herrmann MJ, Hofmann D, Jackowski AP, Jansen A, Kaczkurkin AN, Kindt M, Kingsley EN, Kircher T, Klahn AL, Koelkebeck K, Krug A, Kugel H, Larsen B, Leehr EJ, Leonhardt L, Lotze M, Margraf J, Michałowski J, Muehlhan M, Nenadić I, Pan PM, Pauli P, Peñate W, Pittig A, Plag J, Pujol J, Richter J, Rivero FL, Salum GA, Satterthwaite TD, Schäfer A, Schäfer J, Schienle A, Schneider S, Schrammen E, Schruers K, Schulz SM, Seidl E, Stark RM, Stein F, Straube B, Straube T, Ströhle A, Suchan B, Thomopoulos SI, Ventura-Bort C, Visser R, Völzke H, Wabnegger A, Wannemüller A, Wendt J, Wiemer J, Wittchen HU, Wittfeld K, Wright B, Yang Y, Zilverstand A, Zwanzger P, Veltman DJ, Winkler AM, Pine DS, Jahanshad N, Thompson PM, Stein DJ, Van der Wee NJ, Lueken U. Cortical and Subcortical Brain Alterations in Specific Phobia and Its Animal and Blood-Injection-Injury Subtypes: A Mega-Analysis From the ENIGMA Anxiety Working Group. Am J Psychiatry 2024; 181:728-740. [PMID: 38859702 PMCID: PMC11979901 DOI: 10.1176/appi.ajp.20230032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
OBJECTIVE Specific phobia is a common anxiety disorder, but the literature on associated brain structure alterations exhibits substantial gaps. The ENIGMA Anxiety Working Group examined brain structure differences between individuals with specific phobias and healthy control subjects as well as between the animal and blood-injection-injury (BII) subtypes of specific phobia. Additionally, the authors investigated associations of brain structure with symptom severity and age (youths vs. adults). METHODS Data sets from 31 original studies were combined to create a final sample with 1,452 participants with phobia and 2,991 healthy participants (62.7% female; ages 5-90). Imaging processing and quality control were performed using established ENIGMA protocols. Subcortical volumes as well as cortical surface area and thickness were examined in a preregistered analysis. RESULTS Compared with the healthy control group, the phobia group showed mostly smaller subcortical volumes, mixed surface differences, and larger cortical thickness across a substantial number of regions. The phobia subgroups also showed differences, including, as hypothesized, larger medial orbitofrontal cortex thickness in BII phobia (N=182) compared with animal phobia (N=739). All findings were driven by adult participants; no significant results were observed in children and adolescents. CONCLUSIONS Brain alterations associated with specific phobia exceeded those of other anxiety disorders in comparable analyses in extent and effect size and were not limited to reductions in brain structure. Moreover, phenomenological differences between phobia subgroups were reflected in diverging neural underpinnings, including brain areas related to fear processing and higher cognitive processes. The findings implicate brain structure alterations in specific phobia, although subcortical alterations in particular may also relate to broader internalizing psychopathology.
Collapse
Affiliation(s)
- Kevin Hilbert
- Department of Psychology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ole Jonas Boeken
- Department of Psychology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Till Langhammer
- Department of Psychology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Nynke A. Groenewold
- Neuroscience Institute, Department of Psychiatry and Mental Health, University of Cape Town
- South African Medical Research Council (SA-MRC) Unit on Child and Adolescent Health, Department of Paediatrics and Child Health, Red Cross War Memorial Children’s Hospital, University of Cape Town, Cape Town, South Africa
| | - Janna Marie Bas-Hoogendam
- Department of Psychiatry, Leiden University Medical Center, Leiden, The Netherlands
- Department of Developmental and Educational Psychology, Institute of Psychology, Leiden University, Leiden, The Netherlands
- Leiden Institute for Brain and Cognition, Leiden, The Netherlands
| | - Moji Aghajani
- Leiden University, Institute of Education & Child Studies, Section Forensic Family & Youth Care, Leiden, The Netherlands
- Department of Psychiatry, Amsterdam UMC location VUMC, Amsterdam, The Netherlands
| | - André Zugman
- Emotion and Development Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Fredrik Åhs
- Department of Psychology and Social Work, Mid Sweden University, Östersund, Sweden
| | - Volker Arolt
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Katja Beesdo-Baum
- Institute for Clinical Psychology und Psychotherapy, Behavioral Epidemiology, Technische Universität Dresden, Dresden, Germany
| | | | | | - Laura Blanco-Hinojo
- MRI Reseach Unit, Department of Radiology, Hospital del Mar, Barcelona, Spain
- IMIM-CIBER de Salud Mental, Instituto de Salud Carlos III., Barcelona, Spain
| | - Joscha Böhnlein
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Robin Bülow
- Institute of Diagnostic Radiology and Neuroradiology, University Medicine Greifswald, Greifswald, Germany
| | - Marta Cano
- Mental Health Department, Unitat de Neurociència Traslacional, Parc Tauli University Hospital, Institut d’Investigació i Innovació Sanitària Parc Taulí (I3PT), Barcelona, Spain
- Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Department of Psychobiology and Methodology of Health Sciences, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Salud Mental, Carlos III Health Institute, Madrid, Spain
| | - Narcis Cardoner
- Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Salud Mental, Carlos III Health Institute, Madrid, Spain
- Department of Psychiatry and Forensic Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Xavier Caseras
- Department of Psychological Medicine and Clinical Neurosciences, Cardiff University Cardiff United Kingdom
| | - Udo Dannlowski
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Katharina Domschke
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Lydia Fehm
- Department of Psychology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Brandee Feola
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mats Fredrikson
- Department of Psychology, Uppsala University, Uppsala, Sweden
| | - Liesbet Goossens
- Department of Psychiatry and Neuropsychology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Hans J. Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- German Center for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, Greifswald, Germany
| | - Dominik Grotegerd
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Raquel E. Gur
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
| | - Alfons O. Hamm
- Department of Biological and Clinical Psychology, University of Greifswald, Greifswald, Germany
| | - Anita Harrewijn
- Emotion and Development Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
- Department of Psychology, Education and Child Studies, Erasmus University Rotterdam Bethesda, MD, USA
| | - Ingmar Heinig
- Institute of Clinical Psychology & Psychotherapy, Technische Universität Dresden, Dresden, Germany
| | - Martin J. Herrmann
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital Würzburg, Würzburg, Germany
| | - David Hofmann
- Institute of Medical Psychology and Systems Neuroscience, University of Münster, Münster, Germany
| | - Andrea P. Jackowski
- LiNC, Department of Psychiatry, Federal University of São Paulo, São Paulo, Brazil
- Department of Education, Information and Communications Technology (ICT) and Learning, Østfold University College, Halden, Norway
| | - Andreas Jansen
- Core-Facility Brainimaging, Faculty of Medicine, University of Marburg, Marburg, Germany
| | | | - Merel Kindt
- Department of Psychology, University of Amsterdam, Amsterdam, The Netherlands
| | - Ellen N. Kingsley
- COMIC Research, Leeds and York Partnership NHS Foundation Trust, Leeds, United Kingdom
| | - Tilo Kircher
- Department of Psychiatry, University of Marburg, Marburg, Germany
| | - Anna L. Klahn
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Katja Koelkebeck
- LVR-Hospital Essen, Department of Psychiatry and Psychotherapy, Institute and Hospital of the University of Duisburg-Essen, Essen, Germany
| | - Axel Krug
- Department of Psychiatry, University of Marburg, Marburg, Germany
- Department of Psychiatry, University Hospital of Bonn, Bonn, Germany
| | - Harald Kugel
- University Clinic for Radiology, University of Münster, Münster, Germany
| | - Bart Larsen
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
| | - Elisabeth J. Leehr
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Lieselotte Leonhardt
- Institute of Clinical Psychology & Psychotherapy, Technische Universität Dresden, Dresden, Germany
| | - Martin Lotze
- Functional Imaging Unit. Diagnostic Radiology and Neuroradiology, University Medicine Greifswald, Greifswald, Germany
| | - Jürgen Margraf
- Mental Health Research and Treatment Center, Faculty of Psychology, Ruhr-Universität Bochum, Bochum, Germany
| | - Jarosław Michałowski
- Laboratory of Affective Neuroscience in Poznan, SWPS University of Social Sciences and Humanities, Warszawa, Poland
| | - Markus Muehlhan
- Department of Psychology, Faculty of Human Sciences, Medical School Hamburg, Hamburg, Germany
- ICAN Institute for Cognitive and Affective Neuroscience, Medical School Hamburg, Hamburg, Germany
| | - Igor Nenadić
- Department of Psychiatry, University of Marburg, Marburg, Germany
| | - Pedro M. Pan
- LiNC, Department of Psychiatry, Federal University of São Paulo, São Paulo, Brazil
| | - Paul Pauli
- Department of Psychology, University of Würzburg, Würzburg, Germany
| | - Wenceslao Peñate
- Department of Clinical Psychology, Psychobiology and Methodology, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Andre Pittig
- Translational Psychotherapy, Institute of Psychology, University of Goettingen, Goettingen, Germany
| | - Jens Plag
- Department of Psychiatry and Psychotherapy, Campus Mitte, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Department of Psychiatry, Psychotherapy and Psychosomatics, Alexianer Krankenhaus Hedwigshoehe, St. Hedwig Kliniken, Berlin, Germany
| | - Jesus Pujol
- IMIM-CIBER de Salud Mental, Instituto de Salud Carlos III., Barcelona, Spain
- MRI Reseach Unit, Department of Radiology, Hospital del Mar, Barcelona, Spain
| | - Jan Richter
- Department of Biological and Clinical Psychology, University of Greifswald, Greifswald, Germany
- Department of Experimental Psychopathology, University of Hildesheim, Hildesheim, Germany
| | - Francisco L. Rivero
- Department of Clinical Psychology, Psychobiology and Methodology, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
- Departament os Psychology, Faculty of Health Science, Universidad Europea de Canarias, La Orotava, Spain
| | - Giovanni A. Salum
- Section on Negative Affect and Social Processes, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | | | - Axel Schäfer
- Bender Institute of Neuroimaging (BION), Justus-Liebig University Gießen, Center of Mind, Brain and Behavior, Universities of Marburg and Gießen, Gießen, Germany
| | - Judith Schäfer
- Institute of Clinical Psychology & Psychotherapy, Technische Universität Dresden, Dresden, Germany
| | - Anne Schienle
- Department of Psychology, University of Graz, Graz, Austria
| | - Silvia Schneider
- Faculty of Psychology, Clinical Child and Adolescent Psychology, Mental Health Research and Treatment Center, Ruhr-Universität Bochum, Bochum, Germany
| | - Elisabeth Schrammen
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Koen Schruers
- Department of Psychiatry and Neuropsychology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Stefan M. Schulz
- Department of Psychology, University of Würzburg, Würzburg, Germany
- Department of Behavioural Medicine and Principles of Human Biology for the Health Sciences, Universität Trier, Trier, Germany
| | - Esther Seidl
- Research Group Security and Privacy, Faculty of Computer Science, University of Vienna, Vienna, Austria
| | - Rudolf M. Stark
- Department of Psychotherapy and Systems Neuroscience, Bender Institute of Neuroimaging (BION), Justus-Liebig University Gießen, Center of Mind, Brain and Behavior, Universities of Marburg and Gießen, Gießen, Germany
| | - Frederike Stein
- Department of Psychiatry, University of Marburg, Marburg, Germany
| | - Benjamin Straube
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, Marburg, Germany
| | - Thomas Straube
- Institute of Medical Psychology and Systems Neuroscience, University of Münster, Münster, Germany
| | - Andreas Ströhle
- Department of Psychiatry and Neuroscience, Campus Charité Mitte, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Boris Suchan
- Department of Psychology, Ruhr-Universität Bochum, Bochum, Germany
| | - Sophia I. Thomopoulos
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California Marina del Rey, CA, USA
| | - Carlos Ventura-Bort
- Department of Biological Psychology and Affective Science, Faculty of Human Sciences, University of Potsdam, Potsdam, Germany
| | - Renee Visser
- Department of Psychology, University of Amsterdam, Amsterdam, The Netherlands
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | | | | | - Julia Wendt
- Department of Biological Psychology and Affective Science, Faculty of Human Sciences, University of Potsdam, Potsdam, Germany
| | - Julian Wiemer
- Department of Psychology, University of Würzburg, Würzburg, Germany
| | | | - Katharina Wittfeld
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- German Center for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, Greifswald, Germany
| | - Barry Wright
- Health Sciences, University of York, York, United Kingdom
| | - Yunbo Yang
- Department of Psychiatry and Psychotherapy, Philipps-University Marburg, Marburg, Germany
| | - Anna Zilverstand
- Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Peter Zwanzger
- KBO-Inn-Salzach-Klinikum, Wasserburg am Inn, Germany
- Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Dick J. Veltman
- Department of Psychiatry, Amsterdam UMC location VUMC, Amsterdam, The Netherlands
| | - Anderson M. Winkler
- Emotion and Development Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Daniel S. Pine
- Emotion and Development Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Neda Jahanshad
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California Marina del Rey, CA, USA
| | - Paul M. Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California Marina del Rey, CA, USA
| | - Dan J. Stein
- South African Medical Research Council Unit on Risk & Resilience in Mental Disorders, Neuroscience Institute, Department of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa
| | - Nic J.A. Van der Wee
- Department of Psychiatry, Leiden University Medical Center, Leiden, The Netherlands
- Leiden Institute for Brain and Cognition, Leiden, The Netherlands
| | - Ulrike Lueken
- Department of Psychology, Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
12
|
Chaudhary S, Zhang S, Chen Y, Dominguez JC, Chao HH, Li CSR. Age-related reduction in anxiety and neural encoding of negative emotional memory. Front Aging Neurosci 2024; 16:1375435. [PMID: 39021704 PMCID: PMC11252031 DOI: 10.3389/fnagi.2024.1375435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 06/24/2024] [Indexed: 07/20/2024] Open
Abstract
Introduction Older adults experience less anxiety. We examined how memory of negative emotional images varied with age and may reflect age-related differences in anxiety. Methods Fifty-one adults, age 22-80 years, underwent imaging with a memory task where negative and neutral images were displayed pseudo-randomly. They were queried post-scan about the images inter-mixed with an equal number of images never displayed. Sensitivity (d') and reporting bias (Z-score of false alarm rate; Z[FAR]) were quantified with signal detection theory. Results Age was negatively correlated with both Spielberg State Trait Anxiety Inventory (STAI) state score and d' (negative - neutral) and positively with Z[FAR] (negative - neutral). However, STAI score and d' or Z[FAR] (negative - neutral) were not significantly correlated. In whole-brain regression, STAI score was correlated with higher activity of the right middle/superior temporal gyri/temporal parietal junction (MTG/STG/TPJ) for "negative correct - incorrect" - "neutral correct - incorrect" trials. Further, the MTG/STG/TPJ activity (β) was also negatively correlated with age. Mediation analyses supported a complete mediation model of age → less anxiety → less MTG/STG/TPJ β. Discussion Together, the findings demonstrated age-related changes in negative emotional memory and how age-related reduction in anxiety is reflected in diminished temporoparietal cortical activities during encoding of negative emotional memory.
Collapse
Affiliation(s)
- Shefali Chaudhary
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - Sheng Zhang
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - Yu Chen
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | | | - Herta H. Chao
- VA Connecticut Healthcare System, West Haven, CT, United States
- Department of Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Chiang-Shan R. Li
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, United States
- Wu Tsai Institute, Yale University, New Haven, CT, United States
| |
Collapse
|
13
|
Ge MJ, Chen G, Zhang ZQ, Yu ZH, Shen JX, Pan C, Han F, Xu H, Zhu XL, Lu YP. Chronic restraint stress induces depression-like behaviors and alterations in the afferent projections of medial prefrontal cortex from multiple brain regions in mice. Brain Res Bull 2024; 213:110981. [PMID: 38777132 DOI: 10.1016/j.brainresbull.2024.110981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 05/06/2024] [Accepted: 05/16/2024] [Indexed: 05/25/2024]
Abstract
INTRODUCTION The medial prefrontal cortex (mPFC) forms output pathways through projection neurons, inversely receiving adjacent and long-range inputs from other brain regions. However, how afferent neurons of mPFC are affected by chronic stress needs to be clarified. In this study, the effects of chronic restraint stress (CRS) on the distribution density of mPFC dendrites/dendritic spines and the projections from the cortex and subcortical brain regions to the mPFC were investigated. METHODS In the present study, C57BL/6 J transgenic (Thy1-YFP-H) mice were subjected to CRS to establish an animal model of depression. The infralimbic (IL) of mPFC was selected as the injection site of retrograde AAV using stereotactic technique. The effects of CRS on dendrites/dendritic spines and afferent neurons of the mPFC IL were investigaed by quantitatively assessing the distribution density of green fluorescent (YFP) positive dendrites/dendritic spines and red fluorescent (retrograde AAV recombinant protein) positive neurons, respectively. RESULTS The results revealed that retrograde tracing virus labeled neurons were widely distributed in ipsilateral and contralateral cingulate cortex (Cg1), second cingulate cortex (Cg2), prelimbic cortex (PrL), infralimbic cortex, medial orbital cortex (MO), and dorsal peduncular cortex (DP). The effects of CRS on the distribution density of mPFC red fluorescence positive neurons exhibited regional differences, ranging from rostral to caudal or from top to bottom. Simultaneously, CRS resulted a decrease in the distribution density of basal, proximal and distal dendrites, as well as an increase in the loss of dendritic spines of the distal dendrites in the IL of mPFC. Furthermore, varying degrees of red retrograde tracing virus fluorescence signals were observed in other cortices, amygdala, hippocampus, septum/basal forebrain, hypothalamus, thalamus, mesencephalon, and brainstem in both ipsilateral and contralateral brain. CRS significantly reduced the distribution density of red fluorescence positive neurons in other cortices, hippocampus, septum/basal forebrain, hypothalamus, and thalamus. Conversely, CRS significantly increased the distribution density of red fluorescence positive neurons in amygdala. CONCLUSION Our results suggest a possible mechanism that CRS leads to disturbances in synaptic plasticity by affecting multiple inputs to the mPFC, which is characterized by a decrease in the distribution density of dendrites/dendritic spines in the IL of mPFC and a reduction in input neurons of multiple cortices to the IL of mPFC as well as an increase in input neurons of amygdala to the IL of mPFC, ultimately causing depression-like behaviors.
Collapse
Affiliation(s)
- Ming-Jun Ge
- College of Life Science, Anhui Normal University, No. 1 Beijing East Road, Wuhu 241000, China
| | - Geng Chen
- College of Life Science, Anhui Normal University, No. 1 Beijing East Road, Wuhu 241000, China
| | - Zhen-Qiang Zhang
- College of Life Science, Anhui Normal University, No. 1 Beijing East Road, Wuhu 241000, China
| | - Zong-Hao Yu
- College of Life Science, Anhui Normal University, No. 1 Beijing East Road, Wuhu 241000, China
| | - Jun-Xian Shen
- College of Life Science, Anhui Normal University, No. 1 Beijing East Road, Wuhu 241000, China
| | - Chuan Pan
- College of Life Science, Anhui Normal University, No. 1 Beijing East Road, Wuhu 241000, China
| | - Fei Han
- College of Life Science, Anhui Normal University, No. 1 Beijing East Road, Wuhu 241000, China
| | - Hui Xu
- College of Life Science, Anhui Normal University, No. 1 Beijing East Road, Wuhu 241000, China; Anhui College of Traditional Chinese Medicine, No. 18 Wuxiashan West Road, Wuhu 241002, China
| | - Xiu-Ling Zhu
- College of Life Science, Anhui Normal University, No. 1 Beijing East Road, Wuhu 241000, China; Department of Anatomy, Wannan Medical College, No. 22 Wenchang West Road, Wuhu 241002, China
| | - Ya-Ping Lu
- College of Life Science, Anhui Normal University, No. 1 Beijing East Road, Wuhu 241000, China.
| |
Collapse
|
14
|
Jiang Y, Xu L, Cao Y, Meng F, Jiang S, Yang M, Zheng Z, Zhang Y, Yang L, Wang M, Sun G, Liu J, Li C, Cui M. Effects of Interleukin-19 overexpression in the medial prefrontal cortex on anxiety-related behaviors, BDNF expression and p38/JNK/ERK pathways. Brain Res Bull 2024; 212:110952. [PMID: 38636611 DOI: 10.1016/j.brainresbull.2024.110952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/27/2024] [Accepted: 04/14/2024] [Indexed: 04/20/2024]
Abstract
Anxiety is a prevalent mental illness known for its high incidence, comorbidity, and tendency to recur, posing significant societal and individual burdens. Studies have highlighted Interleukin-19 (IL-19) as having potential relevance in neuropsychiatric disorders. Our previous research revealed that IL-19 overexpression in colonies exacerbated anxiety-related behaviors induced by dextran sodium sulfate/stress. However, the precise role and molecular mechanisms of IL-19 in anxiety regulation remain uncertain. In this study, we initiated an acute restraint stress (ARS)-induced anxious mouse model and identified heightened expression of IL-19 and IL-20Rα in the medial prefrontal cortex (mPFC) of ARS mice. Notably, IL-19 and IL-20Rα were predominantly present in the excitatory pyramidal neurons of the mPFC under both basal and ARS conditions. Utilizing the adeno-associated virus (AAV) strategy, we demonstrated that IL-19 overexpression in the mPFC induced anxiety-related behaviors and elevated stress susceptibility. Additionally, we observed decreased protein levels of brain-derived neurotrophic factor (BDNF) and postsynaptic density protein 95 (PSD95) in the mPFC of IL-19 overexpression mice, accompanied by reduced phosphorylation of in the p38, JNK, and Erk signaling pathways. These findings emphasize the role of IL-19 in modulating anxiety-related behaviors within the mPFC and suggest its potential as a pathological gene and therapeutic target for anxiety.
Collapse
Affiliation(s)
- Yuting Jiang
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Lihong Xu
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Yifan Cao
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Fantao Meng
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Shujun Jiang
- Department of Physiology, Binzhou Medical University, Shandong, China
| | - Mengyu Yang
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Ziteng Zheng
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Yi Zhang
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Lu Yang
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Meiqin Wang
- Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China; Department of Physiology, Binzhou Medical University, Shandong, China
| | - Guizhi Sun
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Jing Liu
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| | - Chen Li
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| | - Minghu Cui
- Department of Psychology, Binzhou Medical University Hospital, Binzhou, Shandong, China; Medical Research Center, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| |
Collapse
|
15
|
Han Y, Yan H, Shan X, Li H, Liu F, Xie G, Li P, Guo W. Enhanced interhemispheric resting-state functional connectivity of the visual network is an early treatment response of paroxetine in patients with panic disorder. Eur Arch Psychiatry Clin Neurosci 2024; 274:497-506. [PMID: 37253876 PMCID: PMC10228425 DOI: 10.1007/s00406-023-01627-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 05/22/2023] [Indexed: 06/01/2023]
Abstract
This study aimed to detect alterations in interhemispheric interactions in patients with panic disorder (PD), determine whether such alterations could serve as biomarkers for the diagnosis and prediction of therapeutic outcomes, and map dynamic changes in interhemispheric interactions in patients with PD after treatment. Fifty-four patients with PD and 54 healthy controls (HCs) were enrolled in this study. All participants underwent clinical assessment and a resting-state functional magnetic resonance imaging scan at (i) baseline and (ii) after paroxetine treatment for 4 weeks. A voxel-mirrored homotopic connectivity (VMHC) indicator, support vector machine (SVM), and support vector regression (SVR) were used in this study. Patients with PD showed reduced VMHC in the fusiform, middle temporal/occipital, and postcentral/precentral gyri, relative to those of HCs. After treatment, the patients exhibited enhanced VMHC in the lingual gyrus, relative to the baseline data. The VMHC of the fusiform and postcentral/precentral gyri contributed most to the classification (accuracy = 87.04%). The predicted changes were accessed from the SVR using the aberrant VMHC as features. Positive correlations (p < 0.001) were indicated between the actual and predicted changes in the severity of anxiety. These findings suggest that impaired interhemispheric coordination in the cognitive-sensory network characterized PD and that VMHC can serve as biomarkers and predictors of the efficiency of PD treatment. Enhanced VMHC in the lingual gyrus of patients with PD after treatment implied that pharmacotherapy recruited the visual network in the early stages.
Collapse
Affiliation(s)
- Yiding Han
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Haohao Yan
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Xiaoxiao Shan
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Huabing Li
- Department of Radiology, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Feng Liu
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Guojun Xie
- Department of Psychiatry, The Third People's Hospital of Foshan, Foshan, 528000, Guangdong, China
| | - Ping Li
- Department of Psychiatry, Qiqihar Medical University, Qiqihar, 161006, Heilongjiang, China
| | - Wenbin Guo
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
16
|
Liu H, Hao Z, Qiu S, Wang Q, Zhan L, Huang L, Shao Y, Wang Q, Su C, Cao Y, Sun J, Wang C, Lv Y, Li M, Shen W, Li H, Jia X. Grey matter structural alterations in anxiety disorders: a voxel-based meta-analysis. Brain Imaging Behav 2024; 18:456-474. [PMID: 38150133 DOI: 10.1007/s11682-023-00842-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/07/2023] [Indexed: 12/28/2023]
Abstract
Anxiety disorders (ADs) are a group of prevalent and destructive mental illnesses, but the current understanding of their underlying neuropathology is still unclear. Employing voxel-based morphometry (VBM), previous studies have demonstrated several common brain regions showing grey matter volume (GMV) abnormalities. However, contradictory results have been reported among these studies. Considering that different subtypes of ADs exhibit common core symptoms despite different diagnostic criteria, and previous meta-analyses have found common core GMV-altered brain regions in ADs, the present research aimed to combine the results of individual studies to identify common GMV abnormalities in ADs. Therefore, we first performed a systematic search in PubMed, Embase, and Web of Science on studies investigating GMV differences between patients with ADs and healthy controls (HCs). Then, the anisotropic effect-size signed differential mapping (AES-SDM) was applied in this meta-analysis. A total of 24 studies (including 25 data sets) were included in the current study, and 906 patients with ADs and 1003 HCs were included. Compared with the HCs, the patients with ADs showed increased GMV in the left superior parietal gyrus, right angular gyrus, left precentral gyrus, and right lingual gyrus, and decreased GMV in the bilateral insula, bilateral thalamus, left caudate, and right putamen. In conclusion, the current study has identified some abnormal GMV brain regions that are related to the pathological mechanisms of anxiety disorders. These findings could contribute to a better understanding of the underlying neuropathology of ADs.
Collapse
Affiliation(s)
- Han Liu
- School of Psychology, Zhejiang Normal University, Jinhua, China
- Intelligent Laboratory of Zhejiang Province in Mental Health and Crisis Intervention for Children and Adolescents, Zhejiang Normal University, Jinhua, China
| | - Zeqi Hao
- School of Psychology, Zhejiang Normal University, Jinhua, China
- Intelligent Laboratory of Zhejiang Province in Mental Health and Crisis Intervention for Children and Adolescents, Zhejiang Normal University, Jinhua, China
| | - Shasha Qiu
- School of Psychology, Zhejiang Normal University, Jinhua, China
- Intelligent Laboratory of Zhejiang Province in Mental Health and Crisis Intervention for Children and Adolescents, Zhejiang Normal University, Jinhua, China
| | - Qianqian Wang
- School of Psychology, Zhejiang Normal University, Jinhua, China
- Intelligent Laboratory of Zhejiang Province in Mental Health and Crisis Intervention for Children and Adolescents, Zhejiang Normal University, Jinhua, China
| | - Linlin Zhan
- School of Western Languages, Heilongjiang University, Heilongjiang, China
| | - Lina Huang
- Department of Radiology, Changshu No.2 People's Hospital, The Affiliated Changshu Hospital of Xuzhou Medical University, Changshu, Jiangsu, China
| | - Youbin Shao
- School of Psychology, Zhejiang Normal University, Jinhua, China
- Intelligent Laboratory of Zhejiang Province in Mental Health and Crisis Intervention for Children and Adolescents, Zhejiang Normal University, Jinhua, China
| | - Qing Wang
- Research Center of Brain and Cognitive Neuroscience, Liaoning Normal University, Dalian, China
| | - Chang Su
- School of Psychology, Zhejiang Normal University, Jinhua, China
- Intelligent Laboratory of Zhejiang Province in Mental Health and Crisis Intervention for Children and Adolescents, Zhejiang Normal University, Jinhua, China
| | - Yikang Cao
- School of Information and Electronics Technology, Jiamusi University, Jiamusi, China
| | - Jiawei Sun
- School of Information and Electronics Technology, Jiamusi University, Jiamusi, China
| | - Chunjie Wang
- Institute of Brain Science, Department of Psychology, School of Education, Hangzhou Normal University, Hangzhou, China
- Center for Cognition and Brain Disorders, the Affiliated Hospital, Hangzhou Normal University, Hangzhou, China
| | - Yating Lv
- Center for Cognition and Brain Disorders, the Affiliated Hospital, Hangzhou Normal University, Hangzhou, China
| | - Mengting Li
- School of Psychology, Zhejiang Normal University, Jinhua, China
- Intelligent Laboratory of Zhejiang Province in Mental Health and Crisis Intervention for Children and Adolescents, Zhejiang Normal University, Jinhua, China
| | - Wenbin Shen
- Department of Radiology, Changshu No.2 People's Hospital, The Affiliated Changshu Hospital of Xuzhou Medical University, Changshu, Jiangsu, China
| | - Huayun Li
- School of Psychology, Zhejiang Normal University, Jinhua, China.
- Intelligent Laboratory of Zhejiang Province in Mental Health and Crisis Intervention for Children and Adolescents, Zhejiang Normal University, Jinhua, China.
| | - Xize Jia
- School of Psychology, Zhejiang Normal University, Jinhua, China.
- Intelligent Laboratory of Zhejiang Province in Mental Health and Crisis Intervention for Children and Adolescents, Zhejiang Normal University, Jinhua, China.
| |
Collapse
|
17
|
Broeders TAA, Linsen F, Louter TS, Nawijn L, Penninx BWJH, van Tol MJ, van der Wee NJA, Veltman DJ, van der Werf YD, Schoonheim MM, Vinkers CH. Dynamic reconfigurations of brain networks in depressive and anxiety disorders: The influence of antidepressants. Psychiatry Res 2024; 334:115774. [PMID: 38341928 DOI: 10.1016/j.psychres.2024.115774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 01/30/2024] [Accepted: 02/04/2024] [Indexed: 02/13/2024]
Abstract
Major Depressive Disorder (MDD) and anxiety disorders are highly comorbid recurrent psychiatric disorders. Reduced dynamic reconfiguration of brain regions across subnetworks may play a critical role underlying these deficits, with indications of normalization after treatment with antidepressants. This study investigated dynamic reconfigurations in controls and individuals with a current MDD and/or anxiety disorder including antidepressant users and non-users in a large sample (N = 207) of adults. We quantified the number of subnetworks a region switched to (promiscuity) as well as the total number of switches (flexibility). Average whole-brain (i.e., global) values and subnetwork-specific values were compared between diagnosis and antidepressant groups. No differences in reconfiguration dynamics were found between individuals with a current MDD (N = 49), anxiety disorder (N = 46), comorbid MDD and anxiety disorder (N = 55), or controls (N = 57). Global and sensorimotor network (SMN) promiscuity and flexibility were higher in antidepressant users (N = 49, regardless of diagnosis) compared to non-users (N = 101) and controls. Dynamic reconfigurations were considerably higher in antidepressant users relative to non-users and controls, but not significantly altered in individuals with a MDD and/or anxiety disorder. The increase in antidepressant users was apparent across the whole brain and in the SMN when investigating subnetworks. These findings help disentangle how antidepressants improve symptoms.
Collapse
Affiliation(s)
- T A A Broeders
- Department of Anatomy & Neurosciences, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
| | - F Linsen
- Department of Anatomy & Neurosciences, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands; Department of Psychiatry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - T S Louter
- Department of Anatomy & Neurosciences, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands; Department of Psychiatry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - L Nawijn
- Department of Psychiatry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - B W J H Penninx
- Department of Psychiatry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - M J van Tol
- Department of Neuroscience, University Medical Center Groningen, Groningen, The Netherlands
| | - N J A van der Wee
- Department of Psychiatry, Leiden University Medical Center, Leiden, The Netherlands
| | - D J Veltman
- Department of Anatomy & Neurosciences, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Y D van der Werf
- Department of Psychiatry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - M M Schoonheim
- Department of Anatomy & Neurosciences, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - C H Vinkers
- Department of Anatomy & Neurosciences, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands; Department of Psychiatry, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands; Amsterdam Public Health, Mental Health program, Amsterdam, The Netherlands; GGZ inGeest Mental Health Care, Amsterdam, The Netherlands
| |
Collapse
|
18
|
Sacca V, Chai-Zhang TC, Hodges S, Amores J, Guler S, Todorova N, McDonald CM, Ge T, Kong J. Morphological changes of the limbic system associated with acute and chronic low-back pain: A UK biobank imaging study. Eur J Pain 2024; 28:608-619. [PMID: 38009393 PMCID: PMC10947961 DOI: 10.1002/ejp.2206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 09/29/2023] [Accepted: 11/01/2023] [Indexed: 11/28/2023]
Abstract
BACKGROUND Low back pain (LBP) is a major public health issue that influences physical and emotional factors integral to the limbic system. This study aims to investigate the association between LBP and brain morphometry alterations as the duration of LBP increases (acute vs. chronic). METHODS We used the UK Biobank data to investigate the morphological features of the limbic system in acute LBP (N = 115), chronic LBP (N = 243) and controls (N = 358), and tried to replicate our findings with an independent dataset composed of 45 acute LBP participants evaluated at different timepoints throughout 1 year from the OpenPain database. RESULTS We found that in comparison with chronic LBP and pain-free controls, acute LBP was associated with increased volumes of the nucleus accumbens, amygdala, hippocampus, and thalamus, and increased grey matter volumes in the hippocampus and posterior cingulate gyrus. In the replication cohort, we found non-significantly larger hippocampus and thalamus volumes in the 3-month visit (acute LBP) compared to the 1-year visit (chronic LBP), with similar effect sizes as the UK Biobank dataset. CONCLUSIONS Our results suggest that acute LBP is associated with dramatic morphometric increases in the limbic system and mesolimbic pathway, which may reflect an active brain response and self-regulation in the early stage of LBP. SIGNIFICANCE Our study suggests that LBP in the acute phase is associated with the brain morphometric changes (increase) in some limbic areas, indicating that the acute phase of LBP may represent a crucial stage of self-regulation and active response to the disease's onset.
Collapse
Affiliation(s)
- Valeria Sacca
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, United States
| | - Thalia Celeste Chai-Zhang
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, United States
| | - Sierra Hodges
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, United States
| | - Judith Amores
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, United States
| | - Seyhmus Guler
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, United States
| | - Nevyana Todorova
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, United States
| | - Caroline Merritt McDonald
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, United States
| | - Tian Ge
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, United States
| | - Jian Kong
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, United States
| |
Collapse
|
19
|
Huang M, Ma H, Zou Y, Fan W, Tu L, Zhao J, Ma G, Diao N, Li X, Han P, Zhu L, Shi H. Structural alterations of brain in different disease states of Crohn's disease: Results of a cross-sectional study in a Chinese hospital. Heliyon 2024; 10:e27446. [PMID: 38510022 PMCID: PMC10951496 DOI: 10.1016/j.heliyon.2024.e27446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 02/28/2024] [Indexed: 03/22/2024] Open
Abstract
Rationale and objectives To investigate alterations in the brain structure in patients with Crohn's disease in activity (CD-A) and in remission (CD-R) compared to healthy controls (HCs) and explore the relationship between gray matter volume (GMV) and psychological disorders. Materials and methods A total of 127 CD patients (62 CD-A, 65 CD-R) and 92 healthy controls (HCs) were enrolled and analyzed in this study. The Crohn's disease activity index (CDAI) was used as the grouping criteria. Voxel-based morphometry (VBM) was applied to investigate gray matter volume (GMV), white matter volume (WMV) and global cerebrospinal fluid (CSF) volume alterations. Pearson correlation analysis was used to evaluate the relationships. Results The CSF volume was negatively correlated with the disease duration in CD-R. Increased GMV of CD was observed in the parahippocampal gyrus, precentral gyrus, precuneous cortex, and subcallosal cortex, decreased was located in the occipital pole, precentral gyrus, inferior temporal gyrus, middle frontal gyrus, angular gyrus, frontal pole, lateral occipital cortex, and lingual gyrus. The GMV in the right temporal pole, left precuneous cortex, and left cingulate gyrus had a positive correlation with erythrocyte and hemoglobin in CD groups. The GMV in the right frontal pole, right postcentral gyrus, and left cingulate gyrus had a negative correlation with somatization in the CD groups. The GMV in the right temporal pole had a negative correlation with psychoticism and other in the CD groups. The GMV in the left cingulate gyrus was positive with bowel symptoms and systemic symptoms in the CD groups. Conclusion Alterations of GMV in CD-A and CD-R and associated correlation with psychological disorders may provide evidence for possible neuro-mechanisms of CD with psychological disorders.
Collapse
Affiliation(s)
- Mengting Huang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Hui Ma
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yan Zou
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Wenliang Fan
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Lei Tu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Zhao
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Guina Ma
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Nan Diao
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Xin Li
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Ping Han
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Liangru Zhu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Heshui Shi
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| |
Collapse
|
20
|
Han Y, Yan H, Shan X, Li H, Liu F, Li P, Zhao J, Guo W. Shared and distinctive neural substrates of generalized anxiety disorder with or without depressive symptoms and their roles in prognostic prediction. J Affect Disord 2024; 348:207-217. [PMID: 38160885 DOI: 10.1016/j.jad.2023.12.067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 11/05/2023] [Accepted: 12/24/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND The neurophysiological mechanisms underlying generalized anxiety disorder (GAD) with or without depressive symptoms are obscure. This study aimed to uncover them and assess their predictive value for treatment response. METHODS We enrolled 98 GAD patients [58 (age: 33.22 ± 10.23 years old, males/females: 25/33) with and 40 (age: 33.65 ± 10.49 years old, males/females: 14/26) without depressive symptoms] and 54 healthy controls (HCs, age: 32.28 ± 10.56 years old, males/females: 21/33). Patients underwent clinical assessments and resting-state functional MRI (rs-fMRI) at baseline and after 4-week treatment with paroxetine, while HCs underwent rs-fMRI at baseline only. Regional homogeneity (ReHo) was employed to measure intrinsic brain activity. We compared ReHo in patients to HCs and examined changes in ReHo within the patient groups after treatment. Support vector regression (SVR) analyses were conducted separately for each patient group to predict the patients' treatment response. RESULTS Both patient groups exhibited higher ReHo in the middle/superior frontal gyrus decreased ReHo in different brain regions compared to HCs. Furthermore, differences in ReHo were detected between the two patient groups. After treatment, the patient groups displayed distinct ReHo change patterns. By utilizing SVR based on baseline abnormal ReHo, we effectively predicted treatment response of patients (p-value for correlation < 0.05). LIMITATIONS The dropout rate was relatively high. CONCLUSIONS This study identified shared and unique neural substrates in GAD patients with or without depressive symptoms, potentially serving as biomarkers for treatment response prediction. Comorbid depressive symptoms were associated with differences in disease manifestation and treatment response compared to pure GAD cases.
Collapse
Affiliation(s)
- Yiding Han
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Haohao Yan
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Xiaoxiao Shan
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Huabing Li
- Department of Radiology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Feng Liu
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Ping Li
- Department of Psychiatry, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, China
| | - Jingping Zhao
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Wenbin Guo
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China.
| |
Collapse
|
21
|
Edmiston EK, Chase HW, Jones N, Nhan TJ, Phillips ML, Fournier JC. Differential role of fusiform gyrus coupling in depressive and anxiety symptoms during emotion perception. Soc Cogn Affect Neurosci 2024; 19:nsae009. [PMID: 38334745 PMCID: PMC10908550 DOI: 10.1093/scan/nsae009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 12/06/2023] [Accepted: 02/01/2024] [Indexed: 02/10/2024] Open
Abstract
Anxiety and depression co-occur; the neural substrates of shared and unique components of these symptoms are not understood. Given emotional alterations in internalizing disorders, we hypothesized that function of regions associated with emotion processing/regulation, including the anterior cingulate cortex (ACC), amygdala and fusiform gyrus (FG), would differentiate these symptoms. Forty-three adults with depression completed an emotional functional magnetic resonance imaging task and the Hamilton Depression and Anxiety Scales. We transformed these scales to examine two orthogonal components, one representing internalizing symptom severity and the other the type of internalizing symptoms (anxiety vs depression). We extracted blood oxygen level dependent signal from FG subregions, ACC, and amygdala and performed generalized psychophysiological interaction analyses to assess relationships between symptoms and brain function. Type of internalizing symptoms was associated with FG3-FG1 coupling (F = 8.14, P = 0.007). More coupling was associated with a higher concentration of depression, demonstrating that intra-fusiform coupling is differentially associated with internalizing symptom type (anxiety vs depression). We found an interaction between task condition and internalizing symptoms and dorsal (F = 4.51, P = 0.014) and rostral ACC activity (F = 4.27, P = 0.012). Post hoc comparisons revealed that less activity was associated with greater symptom severity during emotional regulation. Functional coupling differences during emotional processing are associated with depressive relative to anxiety symptoms and internalizing symptom severity. These findings could inform future treatments for depression.
Collapse
Affiliation(s)
- Elliot Kale Edmiston
- Department of Psychiatry, University of Massachusetts Chan Medical School, Worcester, MA 01605, United States
| | - Henry W Chase
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, United States
| | - Neil Jones
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, United States
| | - Tiffany J Nhan
- Department of Psychiatry, University of Massachusetts Chan Medical School, Worcester, MA 01605, United States
| | - Mary L Phillips
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, United States
| | - Jay C Fournier
- Department of Psychiatry and Behavioral Health, The Ohio State University College of Medicine, Columbus, OH 43210, United States
| |
Collapse
|
22
|
Yan H, Han Y, Shan X, Li H, Liu F, Zhao J, Li P, Guo W. Shared and distinctive dysconnectivity patterns underlying pure generalized anxiety disorder (GAD) and comorbid GAD and depressive symptoms. J Psychiatr Res 2024; 170:225-236. [PMID: 38159347 DOI: 10.1016/j.jpsychires.2023.12.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 12/14/2023] [Accepted: 12/18/2023] [Indexed: 01/03/2024]
Abstract
The resting-state connectivity features underlying pure generalized anxiety disorder (GAD, G1) and comorbid GAD and depressive symptoms (G2) have not been directly compared. Furthermore, it is unclear whether these features might serve as potential prognostic biomarkers and change with treatment. Degree centrality (DC) in G1 (40 subjects), G2 (58 subjects), and healthy controls (HCs, 54 subjects) was compared before treatment, and the DC of G1 or G2 at baseline was compared with that after 4 weeks of paroxetine treatment. Using support vector regression (SVR), voxel-wise DC across the entire brain and abnormal DC at baseline were employed to predict treatment response. At baseline, G1 and G2 exhibited lower DC in the left mid-cingulate cortex and vermis IV/V compared to HCs. Additionally, compared to HCs, G1 had lower DC in the left middle temporal gyrus, while G2 showed higher DC in the right inferior temporal/fusiform gyrus. However, there was no significant difference in DC between G1 and G2. The SVR based on abnormal DC at baseline could successfully predict treatment response in responders in G2 or in G1 and G2. Notably, the predictive performance based on abnormal DC at baseline surpassed that based on DC across the entire brain. After treatment, G2 responders showed lower DC in the right medial orbital frontal gyrus, while no change in DC was identified in G1 responders. The G1 and G2 showed common and distinct dysconnectivity patterns and they could potentially serve as prognostic biomarkers. Furthermore, DC in patients with GAD could change with treatment.
Collapse
Affiliation(s)
- Haohao Yan
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Yiding Han
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Xiaoxiao Shan
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Huabing Li
- Department of Radiology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Feng Liu
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jingping Zhao
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Ping Li
- Department of Psychiatry, Qiqihar Medical University, Qiqihar, Heilongjiang, 161006, China
| | - Wenbin Guo
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
23
|
Matar D, Serhan A, El Bilani S, Faraj RA, Hadi BA, Fakhoury M. Psychopharmacological Approaches for Neural Plasticity and Neurogenesis in Major Depressive Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1456:27-48. [PMID: 39261422 DOI: 10.1007/978-981-97-4402-2_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
Major depressive disorder (MDD) is a mental health disorder associated with cognitive impairment, dysregulated appetite, fatigue, insomnia or hypersomnia, and severe mood changes that significantly impact the ability of the affected individual to perform day-to-day tasks, leading to suicide in the worst-case scenario. As MDD is becoming more prevalent, affecting roughly 300 million individuals worldwide, its treatment has become a major point of interest. Antidepressants acting as selective serotonin reuptake inhibitors (SSRIs) are currently used as the first line of treatment for MDD. Other antidepressants currently used for the treatment of MDD include the serotonin and norepinephrine reuptake inhibitors (SNRIs), tricyclic antidepressants (TCAs), and monoamine oxidase inhibitors (MAOIs). However, although effective in alleviating symptoms of MDD, most antidepressants require weeks or even months of regular administration prior to eliciting a rational clinical effect. Owing to the strong evidence showing a relationship between neural plasticity, neurogenesis, and MDD, researchers have also looked at the possibility of using treatment modalities that target these processes in an attempt to improve clinical outcome. The overarching aim of this chapter is to highlight the role of neural plasticity and neurogenesis in the pathophysiology of MDD and discuss the most recently studied treatment strategies that target these processes by presenting supporting evidence from both animal and human studies.
Collapse
Affiliation(s)
- Dina Matar
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| | - Aya Serhan
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| | - Sabah El Bilani
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| | - Rashel Abi Faraj
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon
| | - Bayan Ali Hadi
- School of Pharmacy, Lebanese American University, Beirut, Lebanon
| | - Marc Fakhoury
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut, Lebanon.
| |
Collapse
|
24
|
Efendioğlu MK, Orhan EK, Şen C, Sönmez S, Orhan KS, Baykan B. Olfactory function assessment of migraine patients by using the Sniffin' sticks test: A clinical study. Am J Otolaryngol 2024; 45:104076. [PMID: 37832332 DOI: 10.1016/j.amjoto.2023.104076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 09/29/2023] [Indexed: 10/15/2023]
Abstract
OBJECTIVE This as a cross-sectional controlled clinical study. We hypothesis that the olfactory functions in migraine patients may differ from the healthy controls. In this study, we evaluated the olfactory functions by using a Sniffin' Sticks test battery, which is a reliable and semi quantitative test to evaluate for olfactory dysfunction. METHODS Patients above 18 years of age who had migraine received a definitive diagnosis of migraine from experienced headache specialists based on the criteria of The International Classification of Headache Disorders-3 were included. Odor threshold, discrimination, and identification parameters were assessed using the "Sniffin' Sticks" test. RESULTS One-hundred and one migraine patients (age [mean ± SD], 36.9 ± 10.4 years; range, 18-60 years) and sixty healthy volunteers (age 34.5 ± 13.2 years, range 18-65 years) participated in our study. The median odor threshold score [percentiles 25th-75th] was 8.3 [6.5-9.8] for the migraine group during attack free period and 4.5[3.6-6.0] for the control group. It was found that the migraine group had a median odor discrimination score of 10.0 [10.0-13.0] and the control group 12.0 [11.0-13.0]. These differences were statistically significant (p < 0.001 and p = 0.032 respectively). The median odor discrimination and identification scores were statistically significant higher for the participants with higher educational level group than in those of lower educational group (p < 0.0001). The median odor discrimination and identification scores of those without allodynia (12.0 [10.0-14.0] and 13.0 [10.0-13.0] respectively) were higher than that of those with allodynia (11.0 [9.0-12.0] and 11.0 [10.0-13.0] respectively) (p = 0.037 and p = 0.034 respectively). CONCLUSIONS We found that the odor thresholds, discrimination and identification scores of the migraine group demonstrate differences from those of the healthy group and in relation to allodynia.
Collapse
Affiliation(s)
- Merih Karbay Efendioğlu
- Istanbul University, Istanbul Faculty of Medicine, Department of Neurology, Istanbul, Turkey
| | - Elif Kocasoy Orhan
- Istanbul University, Istanbul Faculty of Medicine, Department of Neurology, Istanbul, Turkey
| | - Cömert Şen
- Istanbul University, Istanbul Faculty of Medicine, Department of Otorhinolaryngology, Istanbul, Turkey
| | - Said Sönmez
- Istanbul University, Istanbul Faculty of Medicine, Department of Otorhinolaryngology, Istanbul, Turkey.
| | - Kadir Serkan Orhan
- Istanbul University, Istanbul Faculty of Medicine, Department of Otorhinolaryngology, Istanbul, Turkey
| | - Betül Baykan
- Istanbul University, Istanbul Faculty of Medicine, Department of Neurology, Istanbul, Turkey
| |
Collapse
|
25
|
He Z, Zheng Y, Ni J, Huang J, Pang Q, Chen T, Muhlert N, Elliott R. Loneliness is related to smaller gray matter volumes in ACC and right VLPFC in people with major depression: a UK biobank study. Cereb Cortex 2023; 33:11656-11667. [PMID: 37874025 DOI: 10.1093/cercor/bhad399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/08/2023] [Accepted: 10/09/2023] [Indexed: 10/25/2023] Open
Abstract
The anterior cingulate cortex (ACC) and right ventrolateral prefrontal cortex (VLPFC) are thought to have important roles in loneliness (feeling of social isolation/exclusion) experience or regulation and in the pathophysiology of their disturbance in major depressive disorder (MDD). However, the structural abnormalities of these regions and the correlates with loneliness in MDD across the healthy population have not fully been clarified. The study analyzed the link between loneliness and gray matter volumes (GMVs) in the ACC and right VLPFC among 1,005 patients with MDD and 7,247 healthy controls (HCs) using UK Biobank data. Significant reductions in GMV in the right VLPFC were found in MDD males compared to HCs. MDD males also showed a higher association between loneliness and reduced GMVs in the right VLPFC and bilateral ACC than HCs. No such associations were found in MDD females. The findings suggest that loneliness may influence brain structures crucial for emotion experience and regulation, particularly in middle-older aged men with MDD. This highlights the potential adverse effects of loneliness on brain structure in MDD and suggests that social engagement could have a positive impact.
Collapse
Affiliation(s)
- Zhenhong He
- School of Psychology, Shenzhen University, Shenzhen 518060, China
- Division of Neuroscience and Experimental Psychology, School of Biological Science, University of Manchester, Manchester M13 9PL, United Kingdom
| | - Youcun Zheng
- School of Science and Engineering, Chinese University of Hong Kong, Shenzhen 518172, China
| | - Jingxuan Ni
- School of Psychology, Shenzhen University, Shenzhen 518060, China
| | - Jin Huang
- School of Mathematical Sciences, Shenzhen University, Shenzhen 518060, China
| | - Qingqing Pang
- School of Psychology, Shenzhen University, Shenzhen 518060, China
| | - Tongtong Chen
- School of Humanities, Shenzhen University, Shenzhen 518060, China
| | - Nils Muhlert
- Division of Neuroscience and Experimental Psychology, School of Biological Science, University of Manchester, Manchester M13 9PL, United Kingdom
| | - Rebecca Elliott
- Division of Neuroscience and Experimental Psychology, School of Biological Science, University of Manchester, Manchester M13 9PL, United Kingdom
| |
Collapse
|
26
|
Liang J, Yu Q, Liu Y, Qiu Y, Tang R, Yan L, Zhou P. Gray matter abnormalities in patients with major depressive disorder and social anxiety disorder: a voxel-based meta-analysis. Brain Imaging Behav 2023; 17:749-763. [PMID: 37725323 PMCID: PMC10733224 DOI: 10.1007/s11682-023-00797-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2023] [Indexed: 09/21/2023]
Abstract
BACKGROUND Major depressive and social anxiety disorders have a high comorbidity rate and similar cognitive patterns. However, their unique and shared neuroanatomical characteristics have not been fully identified. METHODS Voxel-based morphometric studies comparing gray matter volume between patients with major depressive disorder/social anxiety disorder and healthy controls were searched using 4 electronic databases from the inception to March 2022. Stereotactic data were extracted and subsequently tested for convergence and differences using activation likelihood estimation. In addition, based on the result of the meta-analysis, behavioral analysis was performed to assess the functional roles of the regions affected by major depressive disorder and/or social anxiety disorder. RESULTS In total, 34 studies on major depressive disorder with 2873 participants, and 10 studies on social anxiety disorder with 1004 subjects were included. Gray matter volume conjunction analysis showed that the right parahippocampal gyrus region, especially the amygdala, was smaller in patients compared to healthy controls. The contrast analysis of major depressive disorder and social anxiety disorder revealed lower gray matter volume in the right lentiform nucleus and medial frontal gyrus in social anxiety disorder and lower gray matter volume in the left parahippocampal gyrus in major depressive disorder. Behavioral analysis showed that regions with lower gray matter volume in social anxiety disorder are strongly associated with negative emotional processes. CONCLUSIONS The shared and unique patterns of gray matter volume abnormalities in patients with major depressive and social anxiety disorder may be linked to the underlying neuropathogenesis of these mental illnesses and provide potential biomarkers. PROSPERO registration number: CRD42021277546.
Collapse
Affiliation(s)
- Junquan Liang
- Shenzhen Bao'an Chinese Medicine Hospital, The Seventh Clinical Medical School of Guangzhou University of Chinese Medicine, Shenzhen, 518101, Guangdong, China
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences (CAS), Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, Guangdong, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qiaoyun Yu
- Jingzhou Traditional Chinese Medicine Hospital, Jingzhou, Hubei, China
| | - Yuchen Liu
- Shenzhen Luohu District Hospital of TCM, Shenzhen, Guangdong, China
| | - Yidan Qiu
- Centre for the Study of Applied Psychology, Guangdong Key Laboratory of Mental Health and Cognitive Science, School of Psychology, Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, Guangdong, China
| | - Rundong Tang
- Shenzhen Bao'an Chinese Medicine Hospital, The Seventh Clinical Medical School of Guangzhou University of Chinese Medicine, Shenzhen, 518101, Guangdong, China
| | - Luda Yan
- Shenzhen Bao'an Chinese Medicine Hospital, The Seventh Clinical Medical School of Guangzhou University of Chinese Medicine, Shenzhen, 518101, Guangdong, China
| | - Peng Zhou
- Shenzhen Bao'an Chinese Medicine Hospital, The Seventh Clinical Medical School of Guangzhou University of Chinese Medicine, Shenzhen, 518101, Guangdong, China.
| |
Collapse
|
27
|
van Kleef RS, Müller A, van Velzen LS, Marie Bas-Hoogendam J, van der Wee NJA, Schmaal L, Veltman DJ, Rive MM, Ruhé HG, Marsman JBC, van Tol MJ. Functional MRI correlates of emotion regulation in major depressive disorder related to depressive disease load measured over nine years. Neuroimage Clin 2023; 40:103535. [PMID: 37984226 PMCID: PMC10696117 DOI: 10.1016/j.nicl.2023.103535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 10/26/2023] [Accepted: 10/30/2023] [Indexed: 11/22/2023]
Abstract
Major Depressive Disorder (MDD) often is a recurrent and chronic disorder. We investigated the neurocognitive underpinnings of the incremental risk for poor disease course by exploring relations between enduring depression and brain functioning during regulation of negative and positive emotions using cognitive reappraisal. We used fMRI-data from the longitudinal Netherlands Study of Depression and Anxiety acquired during an emotion regulation task in 77 individuals with MDD. Task-related brain activity was related to disease load, calculated from presence and severity of depression in the preceding nine years. Additionally, we explored task related brain-connectivity. Brain functioning in individuals with MDD was further compared to 35 controls to explore overlap between load-effects and general effects related to MDD history/presence. Disease load was not associated with changes in affect or with brain activity, but with connectivity between areas essential for processing, integrating and regulating emotional information during downregulation of negative emotions. Results did not overlap with general MDD-effects. Instead, MDD was generally associated with lower parietal activity during downregulation of negative emotions. During upregulation of positive emotions, disease load was related to connectivity between limbic regions (although driven by symptomatic state), and connectivity between frontal, insular and thalamic regions was lower in MDD (vs controls). Results suggest that previous depressive load relates to brain connectivity in relevant networks during downregulation of negative emotions. These abnormalities do not overlap with disease-general abnormalities and could foster an incremental vulnerability to recurrence or chronicity of MDD. Therefore, optimizing emotion regulation is a promising therapeutic target for improving long-term MDD course.
Collapse
Affiliation(s)
- Rozemarijn S van Kleef
- Department of Biomedical Sciences of Cells and Systems, Cognitive Neuroscience Center, University Medical Center Groningen, Groningen, the Netherlands.
| | - Amke Müller
- Department of Psychology, Helmut Schmidt University / University of the Federal Armed Forces Hamburg, Hamburg, Germany
| | - Laura S van Velzen
- Orygen Parkville, VIC, Centre for Youth Mental Health, University of Melbourne, Melbourne, VIC, Australia
| | - Janna Marie Bas-Hoogendam
- Developmental and Educational Psychology, Institute of Psychology, Leiden University, Leiden, the Netherlands; Department of Psychiatry, Leiden University Medical Center, Leiden, the Netherlands; Leiden Institute for Brain and Cognition, Leiden University Medical Center, the Netherlands
| | - Nic J A van der Wee
- Department of Psychiatry, Leiden University Medical Center, Leiden, the Netherlands; Leiden Institute for Brain and Cognition, Leiden University Medical Center, the Netherlands
| | - Lianne Schmaal
- Orygen Parkville, VIC, Centre for Youth Mental Health, University of Melbourne, Melbourne, VIC, Australia
| | - Dick J Veltman
- Department of Psychiatry, Amsterdam UMC location VUMC & Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Maria M Rive
- Department of Psychiatry, Amsterdam UMC location AMC, Amsterdam, the Netherlands; Triversum, Department of Child and Adolescent Psychiatry, GGZ Noord-Holland Noord, Hoorn, the Netherlands
| | - Henricus G Ruhé
- Department of Psychiatry, Radboudumc, Nijmegen, the Netherlands; Donders Institute for Brain, Cognition and Behavior, Radboud University, Nijmegen, the Netherlands
| | - Jan-Bernard C Marsman
- Department of Biomedical Sciences of Cells and Systems, Cognitive Neuroscience Center, University Medical Center Groningen, Groningen, the Netherlands
| | - Marie-José van Tol
- Department of Biomedical Sciences of Cells and Systems, Cognitive Neuroscience Center, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
28
|
Cabreira V, McWhirter L, Carson A. Functional Cognitive Disorder: Diagnosis, Treatment, and Differentiation from Secondary Causes of Cognitive Difficulties. Neurol Clin 2023; 41:619-633. [PMID: 37775194 DOI: 10.1016/j.ncl.2023.02.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
Abstract
Functional cognitive disorder is an increasingly common cause of referral to the memory clinic. As a substantial source of disability, clinicians involved in the management of patients with cognitive complaints need to familiarize themselves with this important differential diagnosis. Our approach focuses on the identification of positive features of internal inconsistency (historical and clinical clues alongside patterns of performance) instead of an exclusionary approach. Although effective treatments are desperately needed, promising therapies include metacognitive retraining and cognitive-behavioral therapy modalities. Future research should focus on a better understanding of disease trajectories and outcomes as well as the development of evidence-based interventions.
Collapse
Affiliation(s)
- Verónica Cabreira
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Laura McWhirter
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.
| | - Alan Carson
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
29
|
Mohammadi S, Seyedmirzaei H, Salehi MA, Jahanshahi A, Zakavi SS, Dehghani Firouzabadi F, Yousem DM. Brain-based Sex Differences in Depression: A Systematic Review of Neuroimaging Studies. Brain Imaging Behav 2023; 17:541-569. [PMID: 37058182 PMCID: PMC10102695 DOI: 10.1007/s11682-023-00772-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2023] [Indexed: 04/15/2023]
Abstract
Major depressive disorder (MDD) is a common psychiatric illness with a wide range of symptoms such as mood decline, loss of interest, and feelings of guilt and worthlessness. Women develop depression more often than men, and the diagnostic criteria for depression mainly rely on female patients' symptoms. By contrast, male depression usually manifests as anger attacks, aggression, substance use, and risk-taking behaviors. Various studies have focused on the neuroimaging findings in psychiatric disorders for a better understanding of their underlying mechanisms. With this review, we aimed to summarize the existing literature on the neuroimaging findings in depression, separated by male and female subjects. A search was conducted on PubMed and Scopus for magnetic resonance imaging (MRI), functional MRI (fMRI), and diffusion tensor imaging (DTI) studies of depression. After screening the search results, 15 MRI, 12 fMRI, and 4 DTI studies were included. Sex differences were mainly reflected in the following regions: 1) total brain, hippocampus, amygdala, habenula, anterior cingulate cortex, and corpus callosum volumes, 2) frontal and temporal gyri functions, along with functions of the caudate nucleus and prefrontal cortex, and 3) frontal fasciculi and frontal projections of corpus callosum microstructural alterations. Our review faces limitations such as small sample sizes and heterogeneity in populations and modalities. But in conclusion, it reflects the possible roles of sex-based hormonal and social factors in the depression pathophysiology.
Collapse
Affiliation(s)
- Soheil Mohammadi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Homa Seyedmirzaei
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Interdisciplinary Neuroscience Research Program (INRP), Tehran University of Medical Sciences, Tehran, Iran
| | | | - Ali Jahanshahi
- School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Seyed Sina Zakavi
- School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - David M Yousem
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institution, Baltimore, MD, USA.
| |
Collapse
|
30
|
Zhang E, Hauson AO, Pollard AA, Meis B, Lackey NS, Carson B, Khayat S, Fortea L, Radua J. Lateralized grey matter volume changes in adolescents versus adults with major depression: SDM-PSI meta-analysis. Psychiatry Res Neuroimaging 2023; 335:111691. [PMID: 37837793 DOI: 10.1016/j.pscychresns.2023.111691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 05/22/2023] [Accepted: 07/19/2023] [Indexed: 10/16/2023]
Abstract
The current study is the first meta-analysis to examine grey matter volume (GMV) changes in adolescents and across the lifespan in major depressive disorder (MDD). Seed-based d mapping-with permutation of subject images (SDM-PSI) has advantages over previous coordinate-based meta-analytical methods (CBMA), such as reducing bias (via the MetaNSUE algorithm) and including non-statistically significant unreported effects. SDM-PSI was used to analyze 105 whole-brain GMV voxel-based morphometry (VBM) studies comparing 6,530 individuals with MDD versus 6,821 age-matched healthy controls (HC). A laterality effect was observed in which adults with MDD showed lower GMV than adult HC in left fronto-temporo-parietal structures (superior temporal gyrus, insula, Rolandic operculum, and inferior frontal gyrus). However, these abnormalities were not statistically significant for adolescent MDD versus adolescent HC. Instead, adolescent MDD showed lower GMV than adult MDD in right temporo-parietal structures (angular gyrus and middle temporal gyrus). These regional differences may be used as potential biomarkers to predict and monitor treatment outcomes as well as to choose the most effective treatments in adolescents versus adults. Finally, due to the paucity of youth, older adult, and longitudinal studies, future studies should attempt to replicate these GMV findings and examine whether they correlate with treatment response and illness severity.
Collapse
Affiliation(s)
- Emily Zhang
- California School of Professional Psychology, Clinical Psychology Ph.D. Program, San Diego, CA, United States of America; Institute of Brain Research and Integrated Neuropsychological Services (iBRAINs.org), San Diego, CA, United States of America
| | - Alexander O Hauson
- California School of Professional Psychology, Clinical Psychology Ph.D. Program, San Diego, CA, United States of America; Institute of Brain Research and Integrated Neuropsychological Services (iBRAINs.org), San Diego, CA, United States of America; Department of Psychiatry, University of California San Diego, La Jolla, CA, United States of America.
| | - Anna A Pollard
- California School of Professional Psychology, Clinical Psychology Ph.D. Program, San Diego, CA, United States of America; Institute of Brain Research and Integrated Neuropsychological Services (iBRAINs.org), San Diego, CA, United States of America
| | - Benjamin Meis
- California School of Professional Psychology, Clinical Psychology Ph.D. Program, San Diego, CA, United States of America; Institute of Brain Research and Integrated Neuropsychological Services (iBRAINs.org), San Diego, CA, United States of America
| | - Nicholas S Lackey
- California School of Professional Psychology, Clinical Psychology Ph.D. Program, San Diego, CA, United States of America; Institute of Brain Research and Integrated Neuropsychological Services (iBRAINs.org), San Diego, CA, United States of America
| | - Bryce Carson
- California School of Professional Psychology, Clinical Psychology Ph.D. Program, San Diego, CA, United States of America; Institute of Brain Research and Integrated Neuropsychological Services (iBRAINs.org), San Diego, CA, United States of America
| | - Sarah Khayat
- Institute of Brain Research and Integrated Neuropsychological Services (iBRAINs.org), San Diego, CA, United States of America
| | - Lydia Fortea
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain; Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Joaquim Radua
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain; Department of Medicine, University of Barcelona, Barcelona, Spain; Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet, Stockholm, Sweden; Department of Psychosis Studies, Institute of Psychology, Psychiatry, and Neuroscience, King's College London, London, United Kingdom
| |
Collapse
|
31
|
Qin L, Liang X, Qi Y, Luo Y, Xiao Q, Huang D, Zhou C, Jiang L, Zhou M, Zhou Y, Tang J, Tang Y. MPFC PV + interneurons are involved in the antidepressant effects of running exercise but not fluoxetine therapy. Neuropharmacology 2023:109669. [PMID: 37473999 DOI: 10.1016/j.neuropharm.2023.109669] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 07/22/2023]
Abstract
Depression is a complex psychiatric disorder. Previous studies have shown that running exercise reverses depression-like behavior faster and more effectively than fluoxetine therapy. GABAergic interneurons, including the PV+ interneuron subtype, in the medial prefrontal cortex (MPFC) are involved in pathological changes of depression. It was unknown whether running exercise and fluoxetine therapy reverse depression-like behavior via GABAergic interneurons or the PV+ interneurons subtype in MPFC. To address this issue, we subjected mice with chronic unpredictable stress (CUS) to a 4-week running exercise or fluoxetine therapy. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis showed that running exercise enriched GABAergic synaptic pathways in the MPFC of CUS-exposed mice. However, the number of PV+ interneurons but not the total number of GABAergic interneurons in the MPFC of mice exposed to CUS reversed by running exercise, not fluoxetine therapy. Running exercise increased the relative gene expression levels of the PV gene in the MPFC of CUS-exposed mice without altering other subtypes of GABAergic interneurons. Moreover, running exercise and fluoxetine therapy both significantly improved the length, area and volume of dendrites and the spine morphology of PV+ interneurons in the MPFC of mice exposed to CUS. However, running exercise but not fluoxetine therapy improved the dendritic complexity level of PV+ interneurons in the MPFC of mice exposed to CUS. In summary, the number and dendritic complexity level of PV+ interneurons may be important therapeutic targets for the mechanism by which running exercise reverses depression-like behavior faster and more effectively than fluoxetine therapy.
Collapse
Affiliation(s)
- Lu Qin
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China; Laboratory of Stem Cells and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China
| | - Xin Liang
- Laboratory of Stem Cells and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China; Department of Pathology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China
| | - Yingqiang Qi
- Institute of Life Science, Chongqing Medical University, Chongqing, 400016, PR China
| | - Yanmin Luo
- Laboratory of Stem Cells and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China; Department of Physiology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China
| | - Qian Xiao
- Laboratory of Stem Cells and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China; Department of Radioactive Medicine, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China
| | - Dujuan Huang
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China; Laboratory of Stem Cells and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China
| | - Chunni Zhou
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China; Laboratory of Stem Cells and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China
| | - Lin Jiang
- Laboratory of Stem Cells and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China; Lab Teaching & Management Center, Chongqing Medical University, Chongqing, 400016, PR China
| | - Mei Zhou
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China; Laboratory of Stem Cells and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China
| | - Yuning Zhou
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China; Laboratory of Stem Cells and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China
| | - Jing Tang
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China; Laboratory of Stem Cells and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China.
| | - Yong Tang
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China; Laboratory of Stem Cells and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, PR China.
| |
Collapse
|
32
|
Olayinka JN, Akawa OB, Ogbu EK, Eduviere AT, Ozolua RI, Soliman M. Apigenin attenuates depressive-like behavior via modulating monoamine oxidase A enzyme activity in chronically stressed mice. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2023; 5:100161. [PMID: 37501771 PMCID: PMC10368777 DOI: 10.1016/j.crphar.2023.100161] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 06/30/2023] [Accepted: 07/07/2023] [Indexed: 07/29/2023] Open
Abstract
Chronic stress is a risk factor for depression and is characterized by elevated levels of brain monoamine oxidase A (MAOA). Mounting evidence has shown that MAOA is a biochemical link between stress and depression. Apigenin (API), a natural flavonoid, as demonstrated in vitro inhibitory effect on MAOA, is suggestive of antidepressant-like activity. However, the in vivo inhibitory effect of API on MAOA and how it affects depression still remain unclear. Here, we report the probable mechanisms of action of API in chronic unpredictable mild stress (CUMS)-induced depression in mice. Treatment with API reversed anhedonia, and reduced anxiety and immobility time in behavioral studies. API reduced brain corticosterone and malondialdehyde (MDA) levels but increased brain levels of glutathione and superoxide dismutase. Furthermore, interleukin-6 and tumor necrosis factor-α were attenuated by API. It also restored cell loss and inhibited the activity of MAOA in the hippocampal brain regions and prefrontal cortex. Comparative binding affinity of API for MAOA (-7.7 kcal/mol) through molecular docking studies was greater than that of reference compound, clorgyline (-6.8 kcal/mol). Favorable hydrophobic interactions important to API binding at MAOA binding cavity was revealed to include conventional hydrogen bond (Cys323 and Tyr444), π-Sulfur (Cys323), π-π Stacked (Tyr407), π-π T-shaped (Phe208), π-lone pair and π-alkyl (Ile335, Ile180) interactions. These results suggest that API is a potent, selective, reversible inhibitor of MAOA with capability of attenuating CUMS-induced depression via inhibiting MAOA enzyme activity and altering other pathomechanisms.
Collapse
Affiliation(s)
- Juliet N. Olayinka
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, Afe Babalola University, P.M.B. 5454, Ado-Ekiti, Ekiti State, Nigeria
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Benin, Benin City, 300001, Nigeria
| | - Oluwole B. Akawa
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, Afe Babalola University, P.M.B. 5454, Ado-Ekiti, Ekiti State, Nigeria
- Molecular Bio-computation and Drug Design Laboratory, Discipline of Pharmaceutical Chemistry, University of Kwazulu-Natal, South Africa
| | - Emmanuela K. Ogbu
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, Afe Babalola University, P.M.B. 5454, Ado-Ekiti, Ekiti State, Nigeria
| | - Anthony T. Eduviere
- Department of Pharmacology, Faculty of Basic Medical Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Raymond I. Ozolua
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Benin, Benin City, 300001, Nigeria
| | - Mahmoud Soliman
- Molecular Bio-computation and Drug Design Laboratory, Discipline of Pharmaceutical Chemistry, University of Kwazulu-Natal, South Africa
| |
Collapse
|
33
|
Hadipour M, Meftahi GH, Jahromi GP. Date palm spathe extract reverses chronic stress-induced changes in dendritic arborization in the amygdala and impairment of hippocampal long-term potentiation. Synapse 2023:e22278. [PMID: 37315214 DOI: 10.1002/syn.22278] [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: 02/25/2023] [Revised: 05/30/2023] [Accepted: 05/31/2023] [Indexed: 06/16/2023]
Abstract
Chronic restraint stress induces anxiety-like behaviors and emotional abnormalities via an alteration of synaptic remodeling in the amygdala and the hippocampus. Given that the date palm spathe has been shown to have neuroprotective effects on different experimental models, this study aimed to address whether the date palm spathe extract (hydroalcoholic extract of date palm spathe [HEDPP]) can reduce chronic restraint stress-induced behavioral, electrophysiological, and morphological changes in the rat model. Thirty-two male Wistar rats (weight 200-220 g) were randomly divided into control, stress, HEDPP, and stress + HEDPP for 14 days. Animals were submitted to restraint stress for 2 h per day for 14 consecutive days. The animals of the HEDPP and stress + HEDPP groups were supplemented with HEDPP (125 mg/kg) during these 14 days, 30 min before being placed in the restraint stress tube. We used passive avoidance, open-field test, and field potential recording to assess emotional memory, anxiety-like behavioral and long-term potentiation in the CA1 region of the hippocampus, respectively. Moreover, Golgi-Cox staining was used to investigate the amygdala neuron dendritic arborization. Results showed that stress induction was associated with behavioral changes (anxiety-like behavioral and emotional memory impairment), and the administration of HEDPP effectively normalized these deficits. HEDPP remarkably amplified the slope and amplitude of mean-field excitatory postsynaptic potentials (fEPSPs) in the CA1 area of the hippocampus in stressed rats. Chronic restraint stress significantly decreased the dendritic arborization in the central and basolateral nucleus of the amygdala neuron. HEDPP suppressed this stress effect in the central nucleus of the amygdala. Our findings indicated that HEDPP administration improves stress-induced learning impairment and memory and anxiety-like behaviors by preventing adverse effects on synaptic plasticity in the hippocampus and amygdala.
Collapse
Affiliation(s)
| | | | - Gila Pirzad Jahromi
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
34
|
Tian YE, Di Biase MA, Mosley PE, Lupton MK, Xia Y, Fripp J, Breakspear M, Cropley V, Zalesky A. Evaluation of Brain-Body Health in Individuals With Common Neuropsychiatric Disorders. JAMA Psychiatry 2023; 80:567-576. [PMID: 37099313 PMCID: PMC10134046 DOI: 10.1001/jamapsychiatry.2023.0791] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 02/15/2023] [Indexed: 04/27/2023]
Abstract
Importance Physical health and chronic medical comorbidities are underestimated, inadequately treated, and often overlooked in psychiatry. A multiorgan, systemwide characterization of brain and body health in neuropsychiatric disorders may enable systematic evaluation of brain-body health status in patients and potentially identify new therapeutic targets. Objective To evaluate the health status of the brain and 7 body systems across common neuropsychiatric disorders. Design, Setting, and Participants Brain imaging phenotypes, physiological measures, and blood- and urine-based markers were harmonized across multiple population-based neuroimaging biobanks in the US, UK, and Australia, including UK Biobank; Australian Schizophrenia Research Bank; Australian Imaging, Biomarkers, and Lifestyle Flagship Study of Ageing; Alzheimer's Disease Neuroimaging Initiative; Prospective Imaging Study of Ageing; Human Connectome Project-Young Adult; and Human Connectome Project-Aging. Cross-sectional data acquired between March 2006 and December 2020 were used to study organ health. Data were analyzed from October 18, 2021, to July 21, 2022. Adults aged 18 to 95 years with a lifetime diagnosis of 1 or more common neuropsychiatric disorders, including schizophrenia, bipolar disorder, depression, generalized anxiety disorder, and a healthy comparison group were included. Main Outcomes and Measures Deviations from normative reference ranges for composite health scores indexing the health and function of the brain and 7 body systems. Secondary outcomes included accuracy of classifying diagnoses (disease vs control) and differentiating between diagnoses (disease vs disease), measured using the area under the receiver operating characteristic curve (AUC). Results There were 85 748 participants with preselected neuropsychiatric disorders (36 324 male) and 87 420 healthy control individuals (40 560 male) included in this study. Body health, especially scores indexing metabolic, hepatic, and immune health, deviated from normative reference ranges for all 4 neuropsychiatric disorders studied. Poor body health was a more pronounced illness manifestation compared to brain changes in schizophrenia (AUC for body = 0.81 [95% CI, 0.79-0.82]; AUC for brain = 0.79 [95% CI, 0.79-0.79]), bipolar disorder (AUC for body = 0.67 [95% CI, 0.67-0.68]; AUC for brain = 0.58 [95% CI, 0.57-0.58]), depression (AUC for body = 0.67 [95% CI, 0.67-0.68]; AUC for brain = 0.58 [95% CI, 0.58-0.58]), and anxiety (AUC for body = 0.63 [95% CI, 0.63-0.63]; AUC for brain = 0.57 [95% CI, 0.57-0.58]). However, brain health enabled more accurate differentiation between distinct neuropsychiatric diagnoses than body health (schizophrenia-other: mean AUC for body = 0.70 [95% CI, 0.70-0.71] and mean AUC for brain = 0.79 [95% CI, 0.79-0.80]; bipolar disorder-other: mean AUC for body = 0.60 [95% CI, 0.59-0.60] and mean AUC for brain = 0.65 [95% CI, 0.65-0.65]; depression-other: mean AUC for body = 0.61 [95% CI, 0.60-0.63] and mean AUC for brain = 0.65 [95% CI, 0.65-0.66]; anxiety-other: mean AUC for body = 0.63 [95% CI, 0.62-0.63] and mean AUC for brain = 0.66 [95% CI, 0.65-0.66). Conclusions and Relevance In this cross-sectional study, neuropsychiatric disorders shared a substantial and largely overlapping imprint of poor body health. Routinely monitoring body health and integrated physical and mental health care may help reduce the adverse effect of physical comorbidity in people with mental illness.
Collapse
Affiliation(s)
- Ye Ella Tian
- Department of Psychiatry, Melbourne Neuropsychiatry Centre, Melbourne Medical School, the University of Melbourne, Melbourne, Victoria, Australia
| | - Maria A. Di Biase
- Department of Psychiatry, Melbourne Neuropsychiatry Centre, Melbourne Medical School, the University of Melbourne, Melbourne, Victoria, Australia
| | - Philip E. Mosley
- Clinical Brain Networks Group, Queensland Institute of Medical Research Berghofer Medical Institute, Brisbane, Queensland, Australia
- Queensland Brain Institute, Brisbane, Queensland, Australia
- Australian e-Health Research Centre, Commonwealth Scientific and Industrial Research Organisation Health and Biosecurity, Brisbane, Queensland, Australia
| | - Michelle K. Lupton
- Queensland Institute of Medical Research Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Ying Xia
- Australian e-Health Research Centre, Commonwealth Scientific and Industrial Research Organisation Health and Biosecurity, Brisbane, Queensland, Australia
| | - Jurgen Fripp
- Australian e-Health Research Centre, Commonwealth Scientific and Industrial Research Organisation Health and Biosecurity, Brisbane, Queensland, Australia
| | - Michael Breakspear
- Discipline of Psychiatry, College of Health, Medicine and Wellbeing, the University of Newcastle, Newcastle, New South Wales, Australia
- School of Psychological Sciences, College of Engineering, Science and Environment, the University of Newcastle, Newcastle, New South Wales, Australia
| | - Vanessa Cropley
- Department of Psychiatry, Melbourne Neuropsychiatry Centre, Melbourne Medical School, the University of Melbourne, Melbourne, Victoria, Australia
| | - Andrew Zalesky
- Department of Psychiatry, Melbourne Neuropsychiatry Centre, Melbourne Medical School, the University of Melbourne, Melbourne, Victoria, Australia
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, the University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
35
|
Bansal R, Hellerstein DJ, Sawardekar S, Chen Y, Peterson BS. A randomized controlled trial of desvenlafaxine-induced structural brain changes in the treatment of persistent depressive disorder. Psychiatry Res Neuroimaging 2023; 331:111634. [PMID: 36996664 DOI: 10.1016/j.pscychresns.2023.111634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 02/27/2023] [Accepted: 03/15/2023] [Indexed: 04/01/2023]
Abstract
The anatomical changes that antidepressant medications induce in the brain and through which they exert their therapeutic effects remain largely unknown. We randomized 61 patients with Persistent Depressive Disorder (PDD) to receive either desvenlafaxine or placebo in a 12-week trial and acquired anatomical MRI scans in 42 of those patients at baseline before randomization and immediately at the end of the trial. We also acquired MRIs once in 39 age- and sex-matched healthy controls. We assessed whether the serotonin-norepinephrine reuptake inhibitor, desvenlafaxine, differentially changed cortical thickness during the trial compared with placebo. Patients relative to controls at baseline had thinner cortices across the brain. Although baseline thickness was not associated with symptom severity, thicker baseline cortices predicted greater reduction in symptom severity in those treated with desvenlafaxine but not placebo. We did not detect significant treatment-by-time effects on cortical thickness. These findings suggest that baseline thickness may serve as predictive biomarkers for treatment response to desvenlafaxine. The absence of treatment-by-time effects may be attributable either to use of insufficient desvenlafaxine dosing, a lack of desvenlafaxine efficacy in treating PDD, or the short trial duration.
Collapse
Affiliation(s)
- Ravi Bansal
- Institute for the Developing Mind, Children's Hospital Los Angeles, CA 90027, USA; Department of Pediatrics, Keck School of Medicine at the University of Southern California, Los Angeles, CA 90033, USA.
| | - David J Hellerstein
- Depression Evaluation Service, Division of Clinical Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA; Department of Psychiatry, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA; New York State Psychiatric Institute, New York, NY 10032, USA
| | - Siddhant Sawardekar
- Institute for the Developing Mind, Children's Hospital Los Angeles, CA 90027, USA
| | - Ying Chen
- Depression Evaluation Service, Division of Clinical Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA; New York State Psychiatric Institute, New York, NY 10032, USA
| | - Bradley S Peterson
- Institute for the Developing Mind, Children's Hospital Los Angeles, CA 90027, USA; Department of Psychiatry, Keck School of Medicine at the University of Southern California, Los Angeles, CA, USA 90033, USA
| |
Collapse
|
36
|
Shokouh Alaei H, Ghoshuni M, Vosough I. Directed brain network analysis in anxious and non-anxious depression based on EEG source reconstruction and graph theory. Biomed Signal Process Control 2023. [DOI: 10.1016/j.bspc.2023.104666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
|
37
|
Yan H, Han Y, Shan X, Li H, Liu F, Xie G, Li P, Guo W. Common and exclusive spontaneous neural activity patterns underlying pure generalized anxiety disorder and comorbid generalized anxiety disorder and depression. J Affect Disord 2023; 331:82-91. [PMID: 36958484 DOI: 10.1016/j.jad.2023.03.047] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/05/2023] [Accepted: 03/16/2023] [Indexed: 03/25/2023]
Abstract
BACKGROUND This study aimed to identify common and exclusive neural substrates underlying pure generalized anxiety disorder (GAD, G0) and comorbid GAD and depression (G1), assess whether they could assist in diagnosis and prediction of treatment response, and determine whether comorbid depression in GAD patients would change their neural plasticity. METHODS A longitudinal study was conducted, involving 98 patients (40 in the G0 group and 58 in the G1 group) and 54 healthy controls (HCs). The fractional amplitude of low-frequency fluctuations (fALFF), support vector machine, and support vector regression were employed. RESULTS The shared neural underpinnings across the two subtypes of GAD were hyperactivity in the right cerebellar Crus II and inferior temporal gyrus and hypoactivity in the right postcentral gyrus. The G1 group showed hypoactivity in the frontal gyrus, compared with HCs, and hyperactivity in the middle temporal gyrus, compared with the G0 group or HCs. These alterations could aid in diagnosis and the prediction of treatment response with high accuracy. After treatment, both the G1 and G0 groups showed higher fALFF than those before treatment but were located in different brain regions. LIMITATIONS The study was performed in a single center and subjects showed a fairly homogeneous ethnicity. CONCLUSIONS Common and exclusive neural substrates underlying the two subtypes of GAD were identified, which could assist in diagnosis and the prediction of treatment response. Pharmacotherapy for the two subtypes of GAD recruited different pathways, suggesting that comorbid depression in GAD patients would change their neural plasticity.
Collapse
Affiliation(s)
- Haohao Yan
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Yiding Han
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Xiaoxiao Shan
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Huabing Li
- Department of Radiology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Feng Liu
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Guojun Xie
- Department of Psychiatry, The Third People's Hospital of Foshan, Foshan 528000, Guangdong, China
| | - Ping Li
- Department of Psychiatry, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, China
| | - Wenbin Guo
- Department of Psychiatry, and National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China.
| |
Collapse
|
38
|
Jatupornpoonsub T, Thimachai P, Supasyndh O, Wongsawat Y. QEEG characteristics associated with malnutrition-inflammation complex syndrome. Front Hum Neurosci 2023; 17:944988. [PMID: 36825130 PMCID: PMC9941172 DOI: 10.3389/fnhum.2023.944988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 01/19/2023] [Indexed: 02/09/2023] Open
Abstract
End-stage renal disease (ESRD) has been linked to cerebral complications due to the comorbidity of malnutrition and inflammation, which is referred to as malnutrition-inflammation complex syndrome (MICS). The severity of this condition is clinically assessed with the malnutrition-inflammation score (MIS), and a cutoff of five is used to optimally distinguish patients with and without MICS. However, this tool is still invasive and inconvenient, because it combines medical records, physical examination, and laboratory results. These steps require clinicians and limit MIS usage on a regular basis. Cerebral diseases in ESRD patients can be evaluated reliably and conveniently by using quantitative electroencephalogram (QEEG), which possibly reflects the severity of MICS likewise. Given the links between kidney and brain abnormalities, we hypothesized that some QEEG patterns might be associated with the severity of MICS and could be used to distinguish ESRD patients with and without MICS. Hence, we recruited 62 ESRD participants and divided them into two subgroups: ESRD with MICS (17 women (59%), age 60.31 ± 7.79 years, MIS < 5) and ESRD without MICS (20 women (61%), age 62.03 ± 9.29 years, MIS ≥ 5). These participants willingly participated in MIS and QEEG assessments. We found that MICS-related factors may alter QEEG characteristics, including the absolute power of the delta, theta, and beta 1 bands, the relative power of the theta and beta 3 subbands, the coherence of the delta and theta bands, and the amplitude asymmetry of the beta 1 band, in certain brain regions. Although most of these QEEG patterns are significantly correlated with MIS, the delta absolute power, beta 1 amplitude asymmetry, and theta coherence are the optimal inputs for the logistic regression model, which can accurately classify ESRD patients with and without MICS (90.0 ± 5.7% area under the receiver operating characteristic curve). We suggest that these QEEG features can be used not only to evaluate the severity of cerebral disorders in ESRD patients but also to noninvasively monitor MICS in clinical practice.
Collapse
Affiliation(s)
- Tirapoot Jatupornpoonsub
- Brain-Computer Interface Laboratory, Department of Biomedical Engineering, Faculty of Engineering, Mahidol University, Nakhon Pathom, Thailand
| | - Paramat Thimachai
- Division of Nephrology, Department of Medicine, Phramongkutklao Hospital, Bangkok, Thailand
| | - Ouppatham Supasyndh
- Division of Nephrology, Department of Medicine, Phramongkutklao Hospital, Bangkok, Thailand
| | - Yodchanan Wongsawat
- Brain-Computer Interface Laboratory, Department of Biomedical Engineering, Faculty of Engineering, Mahidol University, Nakhon Pathom, Thailand,*Correspondence: Yodchanan Wongsawat ✉
| |
Collapse
|
39
|
Baggio T, Grecucci A, Meconi F, Messina I. Anxious Brains: A Combined Data Fusion Machine Learning Approach to Predict Trait Anxiety from Morphometric Features. SENSORS (BASEL, SWITZERLAND) 2023; 23:610. [PMID: 36679404 PMCID: PMC9863274 DOI: 10.3390/s23020610] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 12/30/2022] [Accepted: 01/01/2023] [Indexed: 06/17/2023]
Abstract
Trait anxiety relates to the steady propensity to experience and report negative emotions and thoughts such as fear and worries across different situations, along with a stable perception of the environment as characterized by threatening stimuli. Previous studies have tried to investigate neuroanatomical features related to anxiety mostly using univariate analyses and thus giving rise to contrasting results. The aim of this study is to build a predictive model of individual differences in trait anxiety from brain morphometric features, by taking advantage of a combined data fusion machine learning approach to allow generalization to new cases. Additionally, we aimed to perform a network analysis to test the hypothesis that anxiety-related networks have a central role in modulating other networks not strictly associated with anxiety. Finally, we wanted to test the hypothesis that trait anxiety was associated with specific cognitive emotion regulation strategies, and whether anxiety may decrease with ageing. Structural brain images of 158 participants were first decomposed into independent covarying gray and white matter networks with a data fusion unsupervised machine learning approach (Parallel ICA). Then, supervised machine learning (decision tree) and backward regression were used to extract and test the generalizability of a predictive model of trait anxiety. Two covarying gray and white matter independent networks successfully predicted trait anxiety. The first network included mainly parietal and temporal regions such as the postcentral gyrus, the precuneus, and the middle and superior temporal gyrus, while the second network included frontal and parietal regions such as the superior and middle temporal gyrus, the anterior cingulate, and the precuneus. We also found that trait anxiety was positively associated with catastrophizing, rumination, other- and self-blame, and negatively associated with positive refocusing and reappraisal. Moreover, trait anxiety was negatively associated with age. This paper provides new insights regarding the prediction of individual differences in trait anxiety from brain and psychological features and can pave the way for future diagnostic predictive models of anxiety.
Collapse
Affiliation(s)
- Teresa Baggio
- Clinical and Affective Neuroscience Lab (CLI.A.N. Lab), Department of Psychology and Cognitive Sciences (DiPSCo), University of Trento, 38068 Rovereto, Italy
| | - Alessandro Grecucci
- Clinical and Affective Neuroscience Lab (CLI.A.N. Lab), Department of Psychology and Cognitive Sciences (DiPSCo), University of Trento, 38068 Rovereto, Italy
- Centre for Medical Sciences, CISMed, University of Trento, 38122 Trento, Italy
| | - Federica Meconi
- Clinical and Affective Neuroscience Lab (CLI.A.N. Lab), Department of Psychology and Cognitive Sciences (DiPSCo), University of Trento, 38068 Rovereto, Italy
| | - Irene Messina
- Clinical and Affective Neuroscience Lab (CLI.A.N. Lab), Department of Psychology and Cognitive Sciences (DiPSCo), University of Trento, 38068 Rovereto, Italy
- Department of Economics, Universitas Mercatorum, 00186 Rome, Italy
| |
Collapse
|
40
|
Meng P, Cheng B, Pan C, Liu L, Cheng S, Yang X, Chen Y, Li C, Zhang H, Zhang Z, Zhang J, He D, Shi S, Chu X, Cai Q, Zhang N, Qin X, Zhao Y, Wei W, Jia Y, Wen Y, Zhang F. Evaluating the role of anxiety on the association between irritable bowel syndrome and brain volumes: a mediation analysis in the UK Biobank cohort. Brain Commun 2023; 5:fcad116. [PMID: 37091589 PMCID: PMC10116581 DOI: 10.1093/braincomms/fcad116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 01/24/2023] [Accepted: 04/07/2023] [Indexed: 04/25/2023] Open
Abstract
There is a strong link between irritable bowel syndrome and brain volumes, yet, to date, research examining the mediators of this association has been little. Based on the phenotypic data of 15 248 participants from the UK Biobank, a two-stage mediation analysis was performed to assess the association among brain volumes, anxiety, and irritable bowel syndrome. In the first stage, we identified the candidate mediating role of anxiety for irritable bowel syndrome associated with brain volumes using regression models. Then, we quantified the magnitude of the mediation effects by evaluating the average causal-mediated effect and proportion of mediation through performing mediation analyses in the R package in the second stage. In the first stage, we identified the partly mediating role of anxiety in the association between irritable bowel syndrome and the volume of thalamus (P left = 1.16 × 10-4, P right = 2.41 × 10-4), and grey matter (P left = 3.22 × 10-2, P right = 1.18 × 10-2) in the VIIIa cerebellum. In the second stage, we observed that the proportion of the total effect of irritable bowel syndrome on volume of thalamus mediated by anxiety was 14.3% for the left region (β Average causal-mediated effect = -0.008, P Average causal-mediated effect = 0.004) and 14.6% for the right region (β Average causal-mediated effect = -0.007, P Average causal-mediated effect = 0.006). Anxiety mediated 30.8% for the left region (β Average causal-mediated effect = -0.013, P Average causal-mediated effect = 0.002) and 21.6% for the right region (β Average causal-mediated effect = -0.010, P Average causal-mediated effect x= 0.018) of the total effect of irritable bowel syndrome on the volume of grey matter in the VIIIa cerebellum. Our study revealed the indirect mediating role of anxiety in the association between irritable bowel syndrome and brain volumes, promoting our understanding of the functional mechanisms of irritable bowel syndrome and its related psychosocial factors.
Collapse
Affiliation(s)
| | | | - Chuyu Pan
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China
| | - Li Liu
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China
| | - Shiqiang Cheng
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China
| | - Xuena Yang
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China
| | - Yujing Chen
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China
| | - Chun’e Li
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China
| | - Huijie Zhang
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China
| | - Zhen Zhang
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China
| | - Jingxi Zhang
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China
| | - Dan He
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China
| | - Sirong Shi
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China
| | - Xiaoge Chu
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China
| | - Qingqing Cai
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China
| | - Na Zhang
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China
| | - Xiaoyue Qin
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China
| | - Yijing Zhao
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China
| | - Wenming Wei
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China
| | - Yumeng Jia
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China
| | - Yan Wen
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China
| | - Feng Zhang
- Correspondence to: Feng Zhang Key Laboratory of Trace Elements and Endemic Diseases National Health Commission of the People’s Republic of China School of Public Health, Health Science Center Xi’an Jiaotong University, No. 76 Yan Ta West Road, Xi’an 710061, China E-mail:
| |
Collapse
|
41
|
Halaris A, Cook J. The Glutamatergic System in Treatment-Resistant Depression and Comparative Effectiveness of Ketamine and Esketamine: Role of Inflammation? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1411:487-512. [PMID: 36949323 DOI: 10.1007/978-981-19-7376-5_21] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
The glutamatergic system is the primary excitatory pathway within the CNS and is responsible for cognition, memory, learning, emotion, and mood. Because of its significant importance in widespread nervous system function, it is tightly regulated through multiple mechanisms, such as glutamate recycling, microglial interactions, and inflammatory pathways. Imbalance within the glutamatergic system has been implicated in a wide range of pathological conditions including neurodegenerative conditions, neuromuscular conditions, and mood disorders including depression. Major depressive disorder (MDD) is the most common mood disorder worldwide, has a high prevalence rate, and afflicts approximately 280 million people. While there are numerous treatments for the disease, 30-40% of patients are unresponsive to treatment and deemed treatment resistant; approximately another third experience only partial improvement (World Health Organization, Depression fact sheet [Internet], 2020). Esketamine, the S-enantiomer of ketamine, was approved by the Food and Drug Administration for treatment-resistant depression (TRD) in 2019 and has offered new hope to patients. It is the first treatment targeting the glutamatergic system through a complex mechanism. Numerous studies have implicated imbalance in the glutamatergic system in depression and treatment resistance. Esketamine and ketamine principally work through inhibition of the NMDA receptor, though more recent studies have implicated numerous other mechanisms mediating the antidepressant efficacy of these agents. These mechanisms include increase in brain-derived neurotrophic factor (BDNF), activation of mammalian target of the rapamycin complex (mTORC), and reduction in inflammation. Esketamine and ketamine have been shown to decrease inflammation in numerous ways principally through reducing pro-inflammatory cytokines (e.g., TNF-α, IL-6) (Loix et al., Acta Anaesthesiol Belg 62(1):47-58, 2011; Chen et al., Psychiatry Res 269:207-11, 2018; Kopra et al., J Psychopharmacol 35(8):934-45, 2021). This anti-inflammatory effect has also been shown to be involved in the antidepressive properties of both ketamine and esketamine (Chen et al., Psychiatry Res 269:207-11, 2018; Kopra et al., J Psychopharmacol 35(8):934-45, 2021).
Collapse
Affiliation(s)
- Angelos Halaris
- Department of Psychiatry, Loyola University Stritch School of Medicine, Maywood, IL, USA.
| | - John Cook
- Department of Psychiatry, Loyola University Stritch School of Medicine, Maywood, IL, USA
| |
Collapse
|
42
|
Schönthaler EMD, Dalkner N, Schwalsberger K, Reininghaus EZ, Reininghaus B. Psychopathic personality traits stress immunity and social potency moderate the relationship between emotional competence and cognitive functions in depression. Front Psychiatry 2023; 14:1061642. [PMID: 37051168 PMCID: PMC10084668 DOI: 10.3389/fpsyt.2023.1061642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 03/03/2023] [Indexed: 04/14/2023] Open
Abstract
Background Psychopathic personality traits (PPT) and depression have both been shown to worsen emotional and cognitive functions. Moreover, PPT and depression share similar underlying neuronal circuits tapping into the emotional and cognitive domains. However, little is known about the influence of PPT on emotion and cognition in individuals with depression. Objectives This study aimed to examine the correlative relationships and moderating role of PPT in the association between emotional competence and cognitive functions in individuals with depression. Methods Data from 373 individuals diagnosed with depression (158 males, 215 females) were examined within a cohort study. Subjects filled out validated questionnaires surveying PPT and emotional competences. Furthermore, a comprehensive neuropsychological test battery was administered. Results Correlation analyses revealed a significant positive association between emotional competence and cognitive functions. Further, negative associations between emotional competence and the PPT "Blame Externalisation" and "Careless Nonplanfulness," as well as positive associations with psychopathic "Social Potency" and "Stress Immunity" were found. Moderation analyses indicated a significant positive influence of psychopathic "Stress Immunity" and "Social Influence" on the relationship between emotional competence and cognitive functions. Conclusion The findings highlight the importance of integrating PPT in depression research. Considering PPT in depression treatment could also facilitate the therapeutic process by identifying individual traits as resilience-strengthening or potentially harmful factors for depressive symptomatology. This study represents a stepping stone for further research regarding the role of personality traits in psychiatric disorders and their treatment.
Collapse
|
43
|
Dandan T, Jingjing S, Ruolin Z, Peng L, Xiaojing G, Qinglin Z, Jiang Q. Right inferior frontal gyrus gray matter density mediates the effect of tolerance of ambiguity on scientific problem finding. CURRENT PSYCHOLOGY 2022. [DOI: 10.1007/s12144-022-04007-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
44
|
Espinoza Oyarce DA, Burns R, Butterworth P, Cherbuin N. A New Measure of the Revised Reinforcement Sensitivity Theory. SOCIAL PSYCHOLOGY 2022. [DOI: 10.1027/1864-9335/a000502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Abstract. The reinforcement sensitivity theory (RST) proposes that neurobiological systems mediate behavior and their functioning can be associated with personality. The functions and associations of RST systems were revised into fight–flight–freeze system (FFFS), behavioral approach/activation system (rBAS), and behavioral inhibition system (rBIS); however, there is limited study of the revised systems due to lack of validated measures. We investigated scale structure, sex invariance, and psychometric properties of the revised RST questionnaire (rRST-Q). The rRST-Q showed good fit as a 5-factor structure with free interfactor correlations and was sex invariant, and associations with personality and mental health measures were consistent with theory and literature. The rRST-Q is a reliable measure, and its use will help understand the link between brain, personality, and behavior.
Collapse
Affiliation(s)
- Daniela A. Espinoza Oyarce
- Centre for Research on Ageing, Health and Wellbeing, National Centre for Epidemiology and Population Health, The Australian National University, Canberra, Australia
| | - Richard Burns
- Centre for Research on Ageing, Health and Wellbeing, National Centre for Epidemiology and Population Health, The Australian National University, Canberra, Australia
| | - Peter Butterworth
- Centre for Research on Ageing, Health and Wellbeing, National Centre for Epidemiology and Population Health, The Australian National University, Canberra, Australia
| | - Nicolas Cherbuin
- Centre for Research on Ageing, Health and Wellbeing, National Centre for Epidemiology and Population Health, The Australian National University, Canberra, Australia
| |
Collapse
|
45
|
Cheng B, Wang X, Roberts N, Zhou Y, Wang S, Deng P, Meng Y, Deng W, Wang J. Abnormal dynamics of resting-state functional activity and couplings in postpartum depression with and without anxiety. Cereb Cortex 2022; 32:5597-5608. [PMID: 35174863 DOI: 10.1093/cercor/bhac038] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 02/05/2023] Open
Abstract
Postpartum depression (PPD) and PPD comorbid with anxiety (PPD-A) are highly prevalent and severe mental health problems in postnatal women. PPD and PPD-A share similar pathopsychological features, leading to ongoing debates regarding the diagnostic and neurobiological uniqueness. This paper aims to delineate common and disorder-specific neural underpinnings and potential treatment targets for PPD and PPD-A by characterizing functional dynamics with resting-state functional magnetic resonance imaging in 138 participants (45 first-episode, treatment-naïve PPD; 31 PDD-A patients; and 62 healthy postnatal women [HPW]). PPD-A group showed specifically increased dynamic amplitude of low-frequency fluctuation in the subgenual anterior cingulate cortex (sgACC) and increased dynamic functional connectivity (dFC) between the sgACC and superior temporal sulcus. PPD group exhibited specifically increased static FC (sFC) between the sgACC and ventral anterior insula. Common disrupted sFC between the sgACC and middle temporal gyrus was found in both PPD and PPD-A patients. Interestingly, dynamic changes in dFC between the sgACC and superior temporal gyrus could differentiate PPD, PPD-A, and HPW. Our study presents initial evidence on specifically abnormal functional dynamics of limbic, emotion regulation, and social cognition systems in patients with PDD and PPD-A, which may facilitate understanding neurophysiological mechanisms, diagnosis, and treatment for PPD and PPD-A.
Collapse
Affiliation(s)
- Bochao Cheng
- Department of Radiology, West China Second University Hospital of Sichuan University, Chengdu 610041, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China
| | - Xiuli Wang
- Department of Psychiatry, The Fourth People's Hospital of Chengdu, University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Neil Roberts
- Edinburgh Imaging facility, The Queen's Medical Research Institute (QMRI), School of Clinical Sciences, University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| | - Yushan Zhou
- Department of Nuclear Medicine, West China Hospital of Sichuan University, Chengdu 610041, China.,Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Song Wang
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Pengcheng Deng
- Department of Radiology, West China Second University Hospital of Sichuan University, Chengdu 610041, China
| | - Yajing Meng
- Department of Psychiatry, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Wei Deng
- Department of Psychiatry, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Jiaojian Wang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming 650500, China
| |
Collapse
|
46
|
Wu H, Li T, Peng C, Yang C, Bian Y, Li X, Xiao Q, Wang P, Zhang Z, Zhang Y. The right prefrontal cortex (PFC) can distinguish anxious depression from non-anxious depression: A promising functional near infrared spectroscopy study (fNIRS). J Affect Disord 2022; 317:319-328. [PMID: 36007594 DOI: 10.1016/j.jad.2022.08.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 07/11/2022] [Accepted: 08/10/2022] [Indexed: 10/31/2022]
Abstract
BACKGROUND Anxious depression is a serious mental disorder characterized by comorbidity of anxiety and depression, and its symptoms are similar to those of non-anxious depression. This study aimed to use functional near-infrared spectroscopy (fNIRS) as a tool to distinguish between patients with anxious and non-anxious depression based on differences in hemodynamic changes in the right prefrontal cortex during the verbal fluency task. It is helpful to improve the diagnostic accuracy of the two disorders to further promote their therapeutic effect and prognosis. METHODS A total of 105 subjects, comprising 39 patients with anxious depression, 32 patients with non-anxious depression, and 32 healthy controls, were evaluated using 53-channel fNIRS and the Depression and Anxiety Clinical Scale. RESULTS Hemodynamic activation was significantly enhanced in the right dorsolateral prefrontal cortex (DLPFC) and right frontopole cortex (FPC) in the anxious depressed group compared with the non-anxious depressed and healthy groups. LIMITATIONS First, Hospital Anxiety and Depression Scale (HADS) was used to evaluate the scores of anxiety and depression among the three groups in our study. Different scales may result in different research results. Therefore, other scales (HAM, the Montgomery Asberg Depression Rating Scale, or the Beck Depression Inventory) should be used for further verification. Second, although all the samples we have chosen were patients with the diagnosis of anxious depression or no-anxious depression, we did not distinguish between different severity of anxious depression or no-anxious depression. Third, pure anxiety was not included as the control condition in our study. CONCLUSIONS There are significant differences in activation patterns of the right DLPFC and right FPC areas between patients with and without anxious depression. Moreover, the right FPC area is promising as a brain region to assess the severity of anxious depression. fNIRS may be a potential tool to improve diagnostic accuracy for both disorders.
Collapse
Affiliation(s)
- Huifen Wu
- School of Education, Huazhong University of Science and Technology, Wuhan, China; School of Education, Hubei Engineering University, Xiaogan, China
| | - Taiping Li
- School of Education, Huazhong University of Science and Technology, Wuhan, China
| | - Cong Peng
- School of Education, Huazhong University of Science and Technology, Wuhan, China
| | - Caihong Yang
- School of Psychology, Central China Normal University, Wuhan, China
| | - Yueran Bian
- School of Education, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoqin Li
- School of Education, Huazhong University of Science and Technology, Wuhan, China
| | - Qiang Xiao
- Department of Psychiatry, Huazhong University of Science and Technology Hospital, Wuhan, China
| | - Pu Wang
- Department of Rehabilitation Medicine in The Seventh Affiliated Hospital (Shenzhen), Sun Yat-Sen University, Shenzhen, China
| | - Zhe Zhang
- Department of Humanities and Social Sciences, Huazhong University of Science and Technology, Wuhan, China.
| | - Yan Zhang
- School of Education, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
47
|
The neuroanatomy of social trust predicts depression vulnerability. Sci Rep 2022; 12:16724. [PMID: 36202831 PMCID: PMC9537537 DOI: 10.1038/s41598-022-20443-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 09/13/2022] [Indexed: 12/01/2022] Open
Abstract
Trust attitude is a social personality trait linked with the estimation of others’ trustworthiness. Trusting others, however, can have substantial negative effects on mental health, such as the development of depression. Despite significant progress in understanding the neurobiology of trust, whether the neuroanatomy of trust is linked with depression vulnerability remains unknown. To investigate a link between the neuroanatomy of trust and depression vulnerability, we assessed trust and depressive symptoms and employed neuroimaging to acquire brain structure data of healthy participants. A high depressive symptom score was used as an indicator of depression vulnerability. The neuroanatomical results observed with the healthy sample were validated in a sample of clinically diagnosed depressive patients. We found significantly higher depressive symptoms among low trusters than among high trusters. Neuroanatomically, low trusters and depressive patients showed similar volume reduction in brain regions implicated in social cognition, including the dorsolateral prefrontal cortex (DLPFC), dorsomedial PFC, posterior cingulate, precuneus, and angular gyrus. Furthermore, the reduced volume of the DLPFC and precuneus mediated the relationship between trust and depressive symptoms. These findings contribute to understanding social- and neural-markers of depression vulnerability and may inform the development of social interventions to prevent pathological depression.
Collapse
|
48
|
Jones JS, Goldstein SJ, Wang J, Gardus J, Yang J, Parsey RV, DeLorenzo C. Evaluation of brain structure and metabolism in currently depressed adults with a history of childhood trauma. Transl Psychiatry 2022; 12:392. [PMID: 36115855 PMCID: PMC9482635 DOI: 10.1038/s41398-022-02153-z] [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: 10/18/2021] [Revised: 08/26/2022] [Accepted: 09/05/2022] [Indexed: 11/22/2022] Open
Abstract
Structural differences in the dorsolateral prefrontal cortex (DLPFC), anterior cingulate cortex (ACC), hippocampus, and amygdala were reported in adults who experienced childhood trauma; however, it is unknown whether metabolic differences accompany these structural differences. This multimodal imaging study examined structural and metabolic correlates of childhood trauma in adults with major depressive disorder (MDD). Participants with MDD completed the Childhood Trauma Questionnaire (CTQ, n = 83, n = 54 female (65.1%), age: 30.4 ± 14.1) and simultaneous positron emission tomography (PET)/magnetic resonance imaging (MRI). Structure (volume, n = 80, and cortical thickness, n = 81) was quantified from MRI using Freesurfer. Metabolism (metabolic rate of glucose uptake) was quantified from dynamic 18F-fluorodeoxyglucose (FDG)-PET images (n = 70) using Patlak graphical analysis. A linear mixed model was utilized to examine the association between structural/metabolic variables and continuous childhood trauma measures while controlling for confounding factors. Bonferroni correction was applied. Amygdala volumes were significantly inversely correlated with continuous CTQ scores. Specifically, volumes were lower by 7.44 mm3 (95% confidence interval [CI]: -12.19, -2.68) per point increase in CTQ. No significant relationship was found between thickness/metabolism and CTQ score. While longitudinal studies are required to establish causation, this study provides insight into potential consequences of, and therefore potential therapeutic targets for, childhood trauma in the prevention of MDD. This work aims to reduce heterogeneity in MDD studies by quantifying neurobiological correlates of trauma within MDD. It further provides biological targets for future interventions aimed at preventing MDD following trauma. To our knowledge, this is the first simultaneous positron emission tomography (PET) and magnetic resonance imaging (MRI) study to assess both structure and metabolism associated with childhood trauma in adults with MDD.
Collapse
Affiliation(s)
- Joshua S. Jones
- grid.16416.340000 0004 1936 9174University of Rochester, Rochester, NY USA
| | - Samantha J. Goldstein
- grid.36425.360000 0001 2216 9681Department of Psychiatry and Behavioral Science, Stony Brook University, New York, NY USA
| | - Junying Wang
- grid.36425.360000 0001 2216 9681Department of Applied Mathematics and Statistics, Stony Brook University, New York, NY USA
| | - John Gardus
- grid.36425.360000 0001 2216 9681Department of Psychiatry and Behavioral Science, Stony Brook University, New York, NY USA
| | - Jie Yang
- grid.36425.360000 0001 2216 9681Department of Family, Population & Preventive Medicine, Stony Brook University, New York, NY USA
| | - Ramin V. Parsey
- grid.36425.360000 0001 2216 9681Department of Psychiatry and Behavioral Science, Stony Brook University, New York, NY USA
| | - Christine DeLorenzo
- grid.36425.360000 0001 2216 9681Department of Psychiatry and Behavioral Science, Stony Brook University, New York, NY USA ,grid.36425.360000 0001 2216 9681Department of Biomedical Engineering, Stony Brook University, New York, NY USA
| |
Collapse
|
49
|
Nawijn L, Dinga R, Aghajani M, van Tol M, van der Wee NJA, Wunder A, Veltman DJ, Penninx BWHJ. Neural correlates of anxious distress in depression: A neuroimaging study of reactivity to emotional faces and resting-state functional connectivity. Depress Anxiety 2022; 39:573-585. [PMID: 35536093 PMCID: PMC9543619 DOI: 10.1002/da.23264] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 04/12/2022] [Accepted: 04/16/2022] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND Comorbid anxiety disorders and anxious distress are highly prevalent in major depressive disorder (MDD). The presence of the DSM-5 anxious distress specifier (ADS) has been associated with worse treatment outcomes and chronic disease course. However, little is known about the neurobiological correlates of anxious distress in MDD. METHODS We probed the relation between the DSM-5 ADS and task-related reactivity to emotional faces, as well as resting-state functional connectivity patterns of intrinsic salience and basal ganglia networks in unmedicated MDD patients with (MDD/ADS+, N = 24) and without ADS (MDD/ADS-, N = 48) and healthy controls (HC, N = 59). Both categorical and dimensional measures of ADS were investigated. RESULTS MDD/ADS+ patients had higher left amygdala responses to emotional faces compared to MDD/ADS- patients (p = .015)-part of a larger striato-limbic cluster. MDD/ADS+ did not differ from MDD/ADS- or controls in resting-state functional connectivity of the salience or basal ganglia networks. CONCLUSIONS Current findings suggest that amygdala and striato-limbic hyperactivity to emotional faces may be a neurobiological hallmark specific to MDD with anxious distress, relative to MDD without anxious distress. This may provide preliminary indications of the underlying mechanisms of anxious distress in depression, and underline the importance to account for heterogeneity in depression research.
Collapse
Affiliation(s)
- Laura Nawijn
- Department of Psychiatry, Amsterdam Neuroscience, Amsterdam UMC, Location VUmcVrije Universiteit AmsterdamAmsterdamThe Netherlands,Department of Psychiatry, Amsterdam Neuroscience, Amsterdam UMC, Location Academic Medical CenterUniversity of AmsterdamAmsterdamThe Netherlands
| | - Richard Dinga
- Department of Psychiatry, Amsterdam Neuroscience, Amsterdam UMC, Location VUmcVrije Universiteit AmsterdamAmsterdamThe Netherlands,Donders Institute for Brain, Cognition and BehaviourRadboud UniversityNijmegenThe Netherlands
| | - Moji Aghajani
- Department of Psychiatry, Amsterdam Neuroscience, Amsterdam UMC, Location VUmcVrije Universiteit AmsterdamAmsterdamThe Netherlands,Section Forensic Family & Youth Care, Institute of Education and Child StudiesLeiden UniversityLeidenThe Netherlands
| | - Marie‐José van Tol
- Department of Biomedical Sciences of Cells and SystemsUniversity Medical Center Groningen, Cognitive Neuroscience CenterGroningenThe Netherlands
| | | | - Andreas Wunder
- Boehringer Ingelheim Pharma GmbH & Co. KG, Translational Medicine and Clinical PharmacologyBiberach an der RissGermany
| | - Dick J. Veltman
- Department of Psychiatry, Amsterdam Neuroscience, Amsterdam UMC, Location VUmcVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Brenda W. H. J. Penninx
- Department of Psychiatry, Amsterdam Neuroscience, Amsterdam UMC, Location VUmcVrije Universiteit AmsterdamAmsterdamThe Netherlands
| |
Collapse
|
50
|
Zhukovsky P, Wainberg M, Milic M, Tripathy SJ, Mulsant BH, Felsky D, Voineskos AN. Multiscale neural signatures of major depressive, anxiety, and stress-related disorders. Proc Natl Acad Sci U S A 2022; 119:e2204433119. [PMID: 35648832 PMCID: PMC9191681 DOI: 10.1073/pnas.2204433119] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 04/15/2022] [Indexed: 11/21/2022] Open
Abstract
The extent of shared and distinct neural mechanisms underlying major depressive disorder (MDD), anxiety, and stress-related disorders is still unclear. We compared the neural signatures of these disorders in 5,405 UK Biobank patients and 21,727 healthy controls. We found the greatest case–control differences in resting-state functional connectivity and cortical thickness in MDD, followed by anxiety and stress-related disorders. Neural signatures for MDD and anxiety disorders were highly concordant, whereas stress-related disorders showed a distinct pattern. Controlling for cross-disorder genetic risk somewhat decreased the similarity between functional neural signatures of stress-related disorders and both MDD and anxiety disorders. Among cases and healthy controls, reduced within-network and increased between-network frontoparietal and default mode connectivity were associated with poorer cognitive performance (processing speed, attention, associative learning, and fluid intelligence). These results provide evidence for distinct neural circuit function impairments in MDD and anxiety disorders compared to stress disorders, yet cognitive impairment appears unrelated to diagnosis and varies with circuit function.
Collapse
Affiliation(s)
- Peter Zhukovsky
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5T 1R8, Canada
| | - Michael Wainberg
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada
| | - Milos Milic
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5T 1R8, Canada
| | - Shreejoy J. Tripathy
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5T 1R8, Canada
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Benoit H. Mulsant
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5T 1R8, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Daniel Felsky
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5T 1R8, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Aristotle N. Voineskos
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada
- Department of Psychiatry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5T 1R8, Canada
| |
Collapse
|