1
|
Hannon K, Easley T, Zhang W, Lew D, Sotiras A, Sheline YI, Marquand A, Barch DM, Bijsterbosch JD. Parsing clinical and neurobiological sources of heterogeneity in depression. Biol Psychiatry 2025:S0006-3223(25)01186-2. [PMID: 40348312 DOI: 10.1016/j.biopsych.2025.04.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/28/2025] [Accepted: 04/23/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND Patients with depression vary from one-another in their clinical and neuroimaging presentation, yet the relationship between clinical and neuroimaging sources of variation is poorly understood. Determining sources of heterogeneity in depression is important to gain insights into its diverse and complex neural etiology. This study aims to test if depression heterogeneity is characterized by subgroups that differ both clinically and neurobiologically and/or whether multiple neuroimaging profiles give rise to the same clinical presentation. METHODS This study utilizes population-based data from the UK Biobank over multiple imaging sites. Clinically dissociated groups were selected to isolate clinical characteristics of depression (symptoms of anhedonia, depressed mood, and somatic disturbance; severity indices of lifetime chronicity and acute impairment; and late onset). Residual neuroimaging heterogeneity within each group was assessed using neuroimaging driven clustering. RESULTS The clinically dissociated subgroups had significantly larger neuroimaging normative deviations than a comparison heterogeneous group and had distinct neuroimaging profiles from each other. Imaging driven clustering within each clinically dissociated group identified two stable subtypes within the acute impairment group that differed significantly in cognitive ability, despite identical clinical profiles. CONCLUSIONS The study identified distinct neuroimaging profiles related to particular clinical depression features that may explain inconsistencies in the literature and sub-clusters within the acute impairment group with cognitive differences that were only differentiable by neuroimaging. Our results provide evidence that multiple neuroimaging profiles may give rise to the same clinical presentation, emphasizing the presence of complex interactions between clinical and neuroimaging sources of heterogeneity.
Collapse
Affiliation(s)
- Kayla Hannon
- Department of Radiology, Washington University School of Medicine, 4525 Scott Avenue, Saint Louis, Missouri 63110, USA.
| | - Ty Easley
- Department of Radiology, Washington University School of Medicine, 4525 Scott Avenue, Saint Louis, Missouri 63110, USA
| | - Wei Zhang
- Department of Radiology, Washington University School of Medicine, 4525 Scott Avenue, Saint Louis, Missouri 63110, USA
| | - Daphne Lew
- Institute for Informatics, Data Science, and Biostatistics, Washington University School of Medicine
| | - Aristeidis Sotiras
- Department of Radiology, Washington University School of Medicine, 4525 Scott Avenue, Saint Louis, Missouri 63110, USA; Institute for Informatics, Data Science, and Biostatistics, Washington University School of Medicine
| | | | - Andre Marquand
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre
| | - Deanna M Barch
- Department of Radiology, Washington University School of Medicine, 4525 Scott Avenue, Saint Louis, Missouri 63110, USA; Department of Psychiatry, Washington University School of Medicine; Department of Psychological & Brain Sciences, Washington University
| | - Janine D Bijsterbosch
- Department of Radiology, Washington University School of Medicine, 4525 Scott Avenue, Saint Louis, Missouri 63110, USA.
| |
Collapse
|
2
|
Wen Z, Hammoud MZ, Siegel CE, Laska EM, Abu-Amara D, Etkin A, Milad MR, Marmar CR. Neuroimaging-based variability in subtyping biomarkers for psychiatric heterogeneity. Mol Psychiatry 2025; 30:1966-1975. [PMID: 39511450 PMCID: PMC12015113 DOI: 10.1038/s41380-024-02807-y] [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: 02/28/2024] [Revised: 10/15/2024] [Accepted: 10/18/2024] [Indexed: 11/15/2024]
Abstract
Neuroimaging-based subtyping is increasingly used to explain heterogeneity in psychiatric disorders. However, the clinical utility of these subtyping efforts remains unclear, and replication has been challenging. Here we examined how the choice of neuroimaging measures influences the derivation of neuro-subtypes and the consequences for clinical delineation. On a clinically heterogeneous dataset (total n = 566) that included controls (n = 268) and cases (n = 298) of psychiatric conditions, including individuals diagnosed with post-traumatic stress disorder (PTSD), traumatic brain injury (TBI), and comorbidity of both (PTSD&TBI), we identified neuro-subtypes among the cases using either structural, resting-state, or task-based measures. The neuro-subtypes for each modality had high internal validity but did not significantly differ in their clinical and cognitive profiles. We further show that the choice of neuroimaging measures for subtyping substantially impacts the identification of neuro-subtypes, leading to low concordance across subtyping solutions. Similar variability in neuro-subtyping was found in an independent dataset (n = 1642) comprised of major depression disorder (MDD, n = 848) and controls (n = 794). Our results suggest that the highly anticipated relationships between neuro-subtypes and clinical features may be difficult to discover.
Collapse
Affiliation(s)
- Zhenfu Wen
- Department of Psychiatry, Grossman School of Medicine, New York University, New York, NY, USA
- Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Science Center at Houston, Houston, TX, USA
| | - Mira Z Hammoud
- Department of Psychiatry, Grossman School of Medicine, New York University, New York, NY, USA
- Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Science Center at Houston, Houston, TX, USA
| | - Carole E Siegel
- Department of Psychiatry, Grossman School of Medicine, New York University, New York, NY, USA
| | - Eugene M Laska
- Department of Psychiatry, Grossman School of Medicine, New York University, New York, NY, USA
| | - Duna Abu-Amara
- Department of Psychiatry, Grossman School of Medicine, New York University, New York, NY, USA
| | - Amit Etkin
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Alto Neuroscience, Mountain View, CA, USA
| | - Mohammed R Milad
- Department of Psychiatry, Grossman School of Medicine, New York University, New York, NY, USA.
- Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Science Center at Houston, Houston, TX, USA.
| | - Charles R Marmar
- Department of Psychiatry, Grossman School of Medicine, New York University, New York, NY, USA.
- Neuroscience Institute, New York University, New York, NY, USA.
| |
Collapse
|
3
|
Hannon K, Jarukasemkit S, Balogh L, Ahmad F, Lenzini P, Sotiras A, Bijsterbosch JD. Comparing Data-Driven Subtypes of Depression Informed by Clinical and Neuroimaging Data: A Registered Report. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2025; 5:100473. [PMID: 40236632 PMCID: PMC11999066 DOI: 10.1016/j.bpsgos.2025.100473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 01/21/2025] [Accepted: 02/13/2025] [Indexed: 04/17/2025] Open
Abstract
Background Efforts to elucidate subtypes within depression have yet to establish a consensus. In this study, we aimed to rigorously compare different subtyping approaches in the same participant space to quantitatively test agreement across subtyping approaches and determine whether the different approaches are sensitive to different sources of heterogeneity in depression. Methods We implemented 6 different data-driven subtyping methods developed in previous work using the same UK Biobank participants (n = 2276 participants with depression, n = 1595 healthy control participants). The 6 approaches include 2 symptom-based, 2 structural neuroimaging-based, and 2 functional neuroimaging-based techniques. The resulting subtypes were compared based on participant assignment, stability, and sensitivity to subtype differences in demographics, general health, clinical characteristics, neuroimaging, trauma, cognition, genetics, and inflammation markers. Results We found almost no agreement between the resulting subtypes of the 6 approaches (mean adjusted Rand index [ARI] = 0.006), even within data domains. This finding was largely driven by differences in input feature set (mean ARI = 0.005) rather than clustering algorithm (mean ARI = 0.23). However, each approach had relatively high internal stability across bootstraps (ARI = 0.36-0.89); most approaches performed above null; and most approaches were sensitive to relevant phenotypes within their data domain. Conclusions Despite marginal overlap between approaches, we found the subtyping approaches to be internally consistent. These results explain why previous studies found strong evidence for subtypes within their analysis but with very little convergence between studies. We recommend that in future work, investigators incorporate systematic comparisons between their approach and alternative/previous approaches to facilitate consensus on depression subtypes.
Collapse
Affiliation(s)
- Kayla Hannon
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Setthanan Jarukasemkit
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
- Department of Internal Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Leda Balogh
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
- University of Amsterdam, Amsterdam, the Netherlands
| | - Fyzeen Ahmad
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
- University of Minnesota, Minneapolis, Minnesota
| | - Petra Lenzini
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Aristeidis Sotiras
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
- Institute for Informatics, Data Science, & Biostatistics, Washington University in St. Louis, St. Louis, Missouri
| | | |
Collapse
|
4
|
Bao J, Wen J, Chang C, Mu S, Chen J, Shivakumar M, Cui Y, Erus G, Yang Z, Yang S, Wen Z, Zhao Y, Kim D, Duong-Tran D, Saykin AJ, Zhao B, Davatzikos C, Long Q, Shen L. A genetically informed brain atlas for enhancing brain imaging genomics. Nat Commun 2025; 16:3524. [PMID: 40229250 PMCID: PMC11997130 DOI: 10.1038/s41467-025-57636-6] [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: 11/14/2024] [Accepted: 02/24/2025] [Indexed: 04/16/2025] Open
Abstract
Brain imaging genomics has manifested considerable potential in illuminating the genetic determinants of human brain structure and function. This has propelled us to develop the GIANT (Genetically Informed brAiN aTlas) that accounts for genetic and neuroanatomical variations simultaneously. Integrating voxel-wise heritability and spatial proximity, GIANT clusters brain voxels into genetically informed regions, while retaining fundamental anatomical knowledge. Compared to conventional (non-genetics) brain atlases, GIANT exhibits smaller intra-region variations and larger inter-region variations in terms of voxel-wise heritability. As a result, GIANT yields increased regional SNP heritability, enhanced polygenicity, and its polygenic risk score explains more brain volumetric variation than traditional neuroanatomical brain atlases. We provide extensive validation to GIANT and demonstrate its neuroanatomical validity, confirming its generalizability across populations with diverse genetic ancestries and various brain conditions. Furthermore, we present a comprehensive genetic architecture of the GIANT regions, covering their functional annotation at the molecular levels, their associations with other complex traits/diseases, and the genetic and phenotypic correlations among GIANT-defined imaging endophenotypes. In summary, GIANT constitutes a brain atlas that captures the complexity of genetic and neuroanatomical heterogeneity, thereby enhancing the discovery power and applicability of imaging genomics investigations in biomedical science.
Collapse
Affiliation(s)
- Jingxuan Bao
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Graduate Group in Genomics and Computational Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Junhao Wen
- Laboratory of AI and Biomedical Science (LABS), Columbia University, New York, NY, USA
- Center for Innovation in Imaging Biomarkers and Integrated Diagnostics (CIMBID), Department of Radiology, Columbia University, New York, NY, USA
- New York Genome Center (NYGC), New York, NY, USA
| | - Changgee Chang
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Shizhuo Mu
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jiong Chen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Graduate Group in Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Manu Shivakumar
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Graduate Group in Genomics and Computational Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Yuhan Cui
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Guray Erus
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Zhijian Yang
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Graduate Group in Applied Mathematics and Computational Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Shu Yang
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Zixuan Wen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Graduate Group in Applied Mathematics and Computational Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Yize Zhao
- Department of Biostatistics, Yale University School of Public Health, New Haven, CT, USA
| | - Dokyoon Kim
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Duy Duong-Tran
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Mathematics, United States Naval Academy, Annapolis, MD, USA
| | - Andrew J Saykin
- Department of Radiology and Imaging Sciences, Indiana University, Indianapolis, IN, USA
| | - Bingxin Zhao
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Christos Davatzikos
- Center for AI and Data Science for Integrated Diagnostics (AI2D), Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Qi Long
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
5
|
Su T, Chen B, Liu Q, Chen Y, Yang M, Wang Q, Zhou H, Zhong X, Ning Y. Sex-specific habenular dysconnectivity in patients with late-life depression. Transl Psychiatry 2025; 15:121. [PMID: 40185707 PMCID: PMC11971314 DOI: 10.1038/s41398-025-03329-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 02/23/2025] [Accepted: 03/18/2025] [Indexed: 04/07/2025] Open
Abstract
There are significant sex differences in the prevalence, symptom presentation, treatment response and brain abnormalities of patients with late-life depression (LLD). The functional connectivity of the habenula has been associated with depressive symptoms and cognitive impairments in patients with LLD. However, sex differences in habenular functional connectivity patterns among LLD patients remain unclear. One hundred and fourteen patients with LLD and 75 healthy controls (HCs) were included in the present study. Resting-state functional magnetic resonance imaging was used to analyse the static and dynamic functional connectivity (sFC and dFC) of the habenula. There were significant interactions between diagnosis (LLD vs. HCs) and sex for the dFC of the left habenula with the left insula, precentral gyrus, angular gyrus, and middle frontal gyrus and for the right habenula with the right middle temporal gyrus. Pairwise comparisons revealed a trend of HC males > HC females and LLD males < HC males for the connections between the left habenula and the left precentral gyrus, angular gyrus and middle frontal gyrus. Conversely, a trend of HC males < HC females and LLD males > HC males was found for the connections between the right habenula and right middle temporal pole. Furthermore, there was a significant interaction for the sFC of the right habenula with the right fusiform gyrus, with trends of HC males > HC females, LLD males < HC males, and LLD females > HC females. Regression analysis revealed that left habenular-left insular dFC was associated with long-delay memory in females and working memory in males; right habenular-right middle temporal pole dFC was associated with information processing speed in females. Sex moderated the relationships between cognitive function (global cognition, delay-recalled memory and working memory) and dFC between the left habenula and left insula. In conclusions, this study revealed sex-specific alterations in the functional connectivity patterns of the habenula in LLD patients, and these alterations were associated with various cognitive functions in a sex-specific manner. These findings provide a neurobiological basis for understanding sex differences in LLD patients.
Collapse
Affiliation(s)
- Ting Su
- The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China
| | - Ben Chen
- The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qin Liu
- The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yunheng Chen
- The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China
| | - Mingfeng Yang
- The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qiang Wang
- The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China
| | - Huarong Zhou
- The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xiaomei Zhong
- The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China.
| | - Yuping Ning
- The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China.
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China Guangzhou Medical University, Guangzhou, China.
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, China.
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China.
| |
Collapse
|
6
|
Luo Z, Hu Z, Qiu X, Li W, Wang C, Lan X, Mai S, Chen Y, Liu G, Zhang F, Chen X, You Z, Zeng Y, Liang Y, Chen Y, Lu H, Zhou Y, Ning Y. Resolving heterogeneity of early-onset major depressive disorder through individual differential structural covariance network analysis. J Affect Disord 2025; 374:630-639. [PMID: 39798711 DOI: 10.1016/j.jad.2025.01.025] [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: 05/04/2024] [Revised: 01/01/2025] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
BACKGROUND Early-onset major depressive disorder (EO-MDD) is characterized by its significant heterogeneity, hindering progress in research. Traditional case-control studies, like group-level structural covariance network, struggle to capture individual heterogeneity among EO-MDD patients. METHODS In this study, T1-weighted structural magnetic resonance imaging was obtained from 185 participants, including 103 EO-MDD patients and 82 healthy controls. A subject-level individual differential structural covariance network (IDSCN) was constructed for each patient based on the concept of normative model. Semi-supervised clustering algorithms were then employed to classify EO-MDD subtypes, followed by validation analyses to assess clustering stability. RESULTS Our study identified two neuroanatomical subtypes. The low-covariance subtype is characterized by significant neural maturation gaps across the whole brain and more pronounced anxiety somatization symptoms. Conversely, the high-covariance subtype demonstrates simultaneous mature of brain structures. CONCLUSION Our findings provide valuable insights into the neuroanatomical heterogeneity of EO-MDD patients, highlighting the importance of considering individual symptom profiles in subtype classification. These findings have substantial clinical implications for personalized treatment and precision medicine, offering more effective treatment choices and accurate diagnoses.
Collapse
Affiliation(s)
- Zhanjie Luo
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China; The First School of Clinical Medicine, Southern Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Zhibo Hu
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Xiaowei Qiu
- School of Mental Health, Guangzhou Medical University
| | - Weicheng Li
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China; The First School of Clinical Medicine, Southern Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Chengyu Wang
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Xiaofeng Lan
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Siming Mai
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Yiying Chen
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Guanxi Liu
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China; The First School of Clinical Medicine, Southern Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Fan Zhang
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China; The First School of Clinical Medicine, Southern Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Xiaoyu Chen
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Zerui You
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China; The First School of Clinical Medicine, Southern Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Yexian Zeng
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Yanmei Liang
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Yifang Chen
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China
| | - Hanna Lu
- Department of Psychiatry, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yanling Zhou
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China.
| | - Yuping Ning
- Department of Child and Adolescent Psychiatry, Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, China; The First School of Clinical Medicine, Southern Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China.
| |
Collapse
|
7
|
Pan N, Long Y, Qin K, Pope I, Chen Q, Zhu Z, Cao Y, Li L, Singh MK, McNamara RK, DelBello MP, Chen Y, Fornito A, Gong Q. Mapping ADHD Heterogeneity and Biotypes through Topological Deviations in Morphometric Similarity Networks. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.27.25324802. [PMID: 40196255 PMCID: PMC11974972 DOI: 10.1101/2025.03.27.25324802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is characterized by considerable clinical heterogeneity. This study investigates whether normative modelling of topological properties derived from brain morphometry similarity networks can provide robust stratification markers for ADHD children. Leveraging multisite neurodevelopmental datasets (discovery: 446 ADHD, 708 controls; validation: 554 ADHD, 123 controls), we constructed morphometric similarity networks and developed normative models for three topological metrics: degree centrality, nodal efficiency, and participation coefficient. Through semi-supervised clustering, we delineated putative biotypes and examined their clinical profiles. We further contextualized brain profiles of these biotypes in terms of their neurochemical and functional correlates using large-scale databases, and assessed model generalizability in an independent cohort. ADHD exhibited atypical hub organization across all three topological metrics, with significant case-control differences primarily localized to a covarying multi-metric component in the orbitofrontal cortex. Three biotypes emerged: one characterized by severe overall symptoms and longitudinally persistent emotional dysregulation, accompanied by pronounced topological alterations in the medial prefrontal cortex and pallidum; a second by predominant hyperactivity/impulsivity accompanied by changes in the anterior cingulate cortex and pallidum; and a third by marked inattention with alterations in the superior frontal gyrus. These neural profiles of each biotype showed distinct neurochemical and functional correlates. Critically, the core findings were replicated in an independent validation cohort. Our comprehensive approach reveals three distinct ADHD biotypes with unique clinical-neural patterns, advancing our understanding of ADHD's neurobiological heterogeneity and laying the groundwork for personalized treatment.
Collapse
Affiliation(s)
- Nanfang Pan
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences; Functional & Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu, China
- The Turner Institute for Brain and Mental Health, School of Psychological Sciences and Monash Biomedical Imaging, Monash University, Clayton, Australia
| | - Yajing Long
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences; Functional & Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu, China
| | - Kun Qin
- Department of Radiology, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Isaac Pope
- The Turner Institute for Brain and Mental Health, School of Psychological Sciences and Monash Biomedical Imaging, Monash University, Clayton, Australia
| | - Qiuxing Chen
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences; Functional & Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu, China
| | - Ziyu Zhu
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences; Functional & Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu, China
- Department of Psychiatry, University of Cincinnati, Cincinnati, USA
| | - Ying Cao
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences; Functional & Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu, China
| | - Lei Li
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences; Functional & Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu, China
| | - Manpreet K. Singh
- Department of Psychiatry and Behavioral Sciences, University of California Davis, Sacramento, USA
| | | | | | - Ying Chen
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences; Functional & Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu, China
| | - Alex Fornito
- The Turner Institute for Brain and Mental Health, School of Psychological Sciences and Monash Biomedical Imaging, Monash University, Clayton, Australia
| | - Qiyong Gong
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University; Research Unit of Psychoradiology, Chinese Academy of Medical Sciences; Functional & Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu, China
- Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen, China
| |
Collapse
|
8
|
Wen J, Skampardoni I, Tian YE, Yang Z, Cui Y, Erus G, Hwang G, Varol E, Boquet-Pujadas A, Chand GB, Nasrallah I, Satterthwaite TD, Shou H, Shen L, Toga AW, Zalesky A, Davatzikos C. Neuroimaging-AI endophenotypes reveal underlying mechanisms and genetic factors contributing to progression and development of four brain disorders. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2023.08.16.23294179. [PMID: 37662256 PMCID: PMC10473785 DOI: 10.1101/2023.08.16.23294179] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Recent work leveraging artificial intelligence has offered promise to dissect disease heterogeneity by identifying complex intermediate brain phenotypes, called dimensional neuroimaging endophenotypes (DNEs). We advance the argument that these DNEs capture the degree of expression of respective neuroanatomical patterns measured, offering a dimensional neuroanatomical representation for studying disease heterogeneity and similarities of neurologic and neuropsychiatric diseases. We investigate the presence of nine DNEs derived from independent yet harmonized studies on Alzheimer's disease, autism spectrum disorder, late-life depression, and schizophrenia in the UK Biobank study. Phenome-wide associations align with genome-wide associations, revealing 31 genomic loci (P-value<5×10-8/9) associated with the nine DNEs.The nine DNEs, along with their polygenic risk scores, significantly enhanced the predictive accuracy for 14 systemic disease categories, particularly for conditions related to mental health and the central nervous system, as well as mortality outcomes. These findings underscore the potential of the nine DNEs to capture the expression of disease-related brain phenotypes in individuals of the general population and to relate such measures with genetics, lifestyle factors, and chronic diseases.
Collapse
Affiliation(s)
- Junhao Wen
- Laboratory of AI and Biomedical Science (LABS), Columbia University, New York, NY, USA
- Department of Radiology, Columbia University, New York, NY, USA
- New York Genome Center (NYGC), New York, NY, USA
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Data Science Institute (DSI), Columbia University, New York, NY, USA
- Center for Innovation in Imaging Biomarkers and Integrated Diagnostics (CIMBID), Department of Radiology, Columbia University, New York, NY, USA
- Zuckerman Institute, Columbia University, New York, NY, USA
| | - Ioanna Skampardoni
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Ye Ella Tian
- Systems Lab, Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Melbourne, Victoria, Australia
| | - Zhijian Yang
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Yuhan Cui
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Guray Erus
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Gyujoon Hwang
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Erdem Varol
- Department of Computer Science and Engineering, New York University, New York, USA
| | - Aleix Boquet-Pujadas
- Laboratory of AI and Biomedical Science (LABS), Columbia University, New York, NY, USA
| | - Ganesh B. Chand
- Department of Radiology, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Ilya Nasrallah
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Theodore D. Satterthwaite
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Haochang Shou
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
- Department of Biostatistics, Epidemiology and Informatics University of Pennsylvania Perelman School of Medicine, Philadelphia, USA
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics University of Pennsylvania Perelman School of Medicine, Philadelphia, USA
| | - Arthur W. Toga
- Laboratory of Neuro Imaging (LONI), Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, California, USA
| | - Andrew Zalesky
- Systems Lab, Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Melbourne, Victoria, Australia
| | - Christos Davatzikos
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| |
Collapse
|
9
|
Xu H, Mu S, Bao J, Davatzikos C, Shou H, Shen L. High-dimensional mediation analysis reveals the mediating role of physical activity patterns in genetic pathways leading to AD-like brain atrophy. BioData Min 2025; 18:24. [PMID: 40128806 PMCID: PMC11931790 DOI: 10.1186/s13040-025-00432-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 02/07/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a complex disorder that affects multiple biological systems including cognition, behavior and physical health. Unfortunately, the pathogenic mechanisms behind AD are not yet clear and the treatment options are still limited. Despite the increasing number of studies examining the pairwise relationships between genetic factors, physical activity (PA), and AD, few have successfully integrated all three domains of data, which may help reveal mechanisms and impact of these genomic and phenomic factors on AD. We use high-dimensional mediation analysis as an integrative framework to study the relationships among genetic factors, PA and AD-like brain atrophy quantified by spatial patterns of brain atrophy. RESULTS We integrate data from genetics, PA and neuroimaging measures collected from 13,425 UK Biobank samples to unveil the complex relationship among genetic risk factors, behavior and brain signatures in the contexts of aging and AD. Specifically, we used a composite imaging marker, Spatial Pattern of Abnormality for Recognition of Early AD (SPARE-AD) that characterizes AD-like brain atrophy, as an outcome variable to represent AD risk. Through GWAS, we identified single nucleotide polymorphisms (SNPs) that are significantly associated with SPARE-AD as exposure variables. We employed conventional summary statistics and functional principal component analysis to extract patterns of PA as mediators. After constructing these variables, we utilized a high-dimensional mediation analysis method, Bayesian Mediation Analysis (BAMA), to estimate potential mediating pathways between SNPs, multivariate PA signatures and SPARE-AD. BAMA incorporates Bayesian continuous shrinkage prior to select the active mediators from a large pool of candidates. We identified a total of 22 mediation pathways, indicating how genetic variants can influence SPARE-AD by altering physical activity. By comparing the results with those obtained using univariate mediation analysis, we demonstrate the advantages of high-dimensional mediation analysis methods over univariate mediation analysis. CONCLUSION Through integrative analysis of multi-omics data, we identified several mediation pathways of physical activity between genetic factors and SPARE-AD. These findings contribute to a better understanding of the pathogenic mechanisms of AD. Moreover, our research demonstrates the potential of the high-dimensional mediation analysis method in revealing the mechanisms of disease.
Collapse
Affiliation(s)
- Hanxiang Xu
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Family Medicine and Public Health, University of California, San Diego, CA, 92093, USA
| | - Shizhuo Mu
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jingxuan Bao
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Christos Davatzikos
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Haochang Shou
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
10
|
Boquet-Pujadas A, Zeng J, Tian YE, Yang Z, Shen L, Zalesky A, Davatzikos C, Wen J. MUTATE: a human genetic atlas of multiorgan artificial intelligence endophenotypes using genome-wide association summary statistics. Brief Bioinform 2025; 26:bbaf125. [PMID: 40135505 PMCID: PMC11938998 DOI: 10.1093/bib/bbaf125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 02/09/2025] [Accepted: 03/03/2025] [Indexed: 03/27/2025] Open
Abstract
Artificial intelligence (AI) has been increasingly integrated into imaging genetics to provide intermediate phenotypes (i.e. endophenotypes) that bridge the genetics and clinical manifestations of human disease. However, the genetic architecture of these AI endophenotypes remains largely unexplored in the context of human multiorgan system diseases. Using publicly available genome-wide association study summary statistics from the UK Biobank (UKBB), FinnGen, and the Psychiatric Genomics Consortium, we comprehensively depicted the genetic architecture of 2024 multiorgan AI endophenotypes (MAEs). We comparatively assessed the single-nucleotide polymorphism-based heritability, polygenicity, and natural selection signatures of 2024 MAEs using methods commonly used in the field. Genetic correlation and Mendelian randomization analyses reveal both within-organ relationships and cross-organ interconnections. Bi-directional causal relationships were established between chronic human diseases and MAEs across multiple organ systems, including Alzheimer's disease for the brain, diabetes for the metabolic system, asthma for the pulmonary system, and hypertension for the cardiovascular system. Finally, we derived polygenic risk scores for the 2024 MAEs for individuals not used to calculate MAEs and returned these to the UKBB. Our findings underscore the promise of the MAEs as new instruments to ameliorate overall human health. All results are encapsulated into the MUlTiorgan AI endophenoTypE genetic atlas and are publicly available at https://labs-laboratory.com/mutate.
Collapse
Affiliation(s)
- Aleix Boquet-Pujadas
- Laboratory of AI and Biomedical Science (LABS), Columbia University, 530 W 166th St, New York, NY 10032, United States
| | - Jian Zeng
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD 4072, Australia
| | - Ye Ella Tian
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Alan Gilbert Building, Level 3/161 Barry St, Carlton VIC 3053, Australia
| | - Zhijian Yang
- GE Healthcare, 1040 12th Ave NW, Issaquah, WA 98027, United States
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, 423 N Service Dr, Philadelphia, PA 19104, United States
| | - Andrew Zalesky
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Alan Gilbert Building, Level 3/161 Barry St, Carlton VIC 3053, Australia
| | - Christos Davatzikos
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, 3700 Hamilton Walk Richards Building, 7th Floor Philadelphia, PA 19104, United States
| | | | - Junhao Wen
- Laboratory of AI and Biomedical Science (LABS), Columbia University, 530 W 166th St, New York, NY 10032, United States
- New York Genome Center (NYGC), 101 6th Ave, New York, NY 10013, United States
- Department of Biomedical Engineering, Columbia University, 1210 Amsterdam Ave, New York, NY 10027, United States
- Data Science Institute (DSI), Columbia University, Mudd Building, W 120th St, New York, NY 10027, United States
- Center for Innovation in Imaging Biomarkers and Integrated Diagnostics (CIMBID), Department of Radiology, Columbia University, 530 W 166th St, New York, NY 10032, United States
- Zuckerman Institute, Columbia University, New York, NY, United States
| |
Collapse
|
11
|
Fang K, Wen B, Liu L, Han S, Zhang W. Disrupted intersubject variability architecture in structural and functional brain connectomes in major depressive disorder. Psychol Med 2025; 55:e56. [PMID: 39973062 DOI: 10.1017/s0033291725000078] [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] [Indexed: 02/21/2025]
Abstract
BACKGROUND Major depressive disorder (MDD) is a heterogeneous condition characterized by significant intersubject variability in clinical presentations. Recent neuroimaging studies have indicated that MDD involves altered brain connectivity across widespread regions. However, the variability in abnormal connectivity among MDD patients remains understudied. METHODS Utilizing a large, multi-site dataset comprising 1,276 patients with MDD and 1,104 matched healthy controls, this study aimed to investigate the intersubject variability of structural covariance (IVSC) and functional connectivity (IVFC) in MDD. RESULTS Patients with MDD demonstrated higher IVSC in the precuneus and lingual gyrus, but lower IVSC in the medial frontal gyrus, calcarine, cuneus, and cerebellum anterior lobe. Conversely, they exhibited an overall increase in IVFC across almost the entire brain, including the middle frontal gyrus, anterior cingulate cortex, hippocampus, insula, striatum, and precuneus. Correlation and mediation analyses revealed that abnormal IVSC was positively correlated with gray matter atrophy and mediated the relationship between abnormal IVFC and gray matter atrophy. As the disease progressed, IVFC increased in the left striatum, insula, right lingual gyrus, posterior cingulate, and left calcarine. Pharmacotherapy significantly reduced IVFC in the right insula, superior temporal gyrus, and inferior parietal lobule. Furthermore, we found significant but distinct correlations between abnormal IVSC and IVFC and the distribution of neurotransmitter receptors, suggesting potential molecular underpinnings. Further analysis confirmed that abnormal patterns of IVSC and IVFC were reproducible and MDD specificity. CONCLUSIONS These results elucidate the heterogeneity of abnormal connectivity in MDD, underscoring the importance of addressing this heterogeneity in future research.
Collapse
Affiliation(s)
- Keke Fang
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
- Henan Engineering Research Center for Tumor Precision Medicine and Comprehensive Evaluation, Henan Cancer Hospital
- Henan Provincial Key Laboratory of Anticancer Drug Research, Henan Cancer Hospital
| | - Baohong Wen
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Henan Province, China
| | - Liang Liu
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Henan Province, China
| | - Shaoqiang Han
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Henan Province, China
| | - Wenzhou Zhang
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
- Henan Engineering Research Center for Tumor Precision Medicine and Comprehensive Evaluation, Henan Cancer Hospital
- Henan Provincial Key Laboratory of Anticancer Drug Research, Henan Cancer Hospital
| |
Collapse
|
12
|
Tofani PS, Máximo RDO, Cochar-Soares N, Ramírez PC, Luiz MM, Lima SS, Silva TBPD, Souza TBD, Silveira LC, Guandalini VR, Steptoe A, de Oliveira C, Alexandre TDS. Does the coexistence of pain and depressive symptoms accelerate cognitive decline? Aging Ment Health 2025; 29:334-342. [PMID: 39161104 DOI: 10.1080/13607863.2024.2392737] [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: 01/08/2024] [Accepted: 08/09/2024] [Indexed: 08/21/2024]
Abstract
OBJECTIVES Investigate whether the coexistence of pain and depressive symptoms is a risk factor for cognitive decline in individuals aged 50 or older. METHOD Longitudinal trajectory study involving 4,718 participants from the English Longitudinal Study of Ageing (ELSA). Joint pain was self-reported, and intensity was classified as mild, moderate/intense. Depressive symptoms were investigated using the Centre for Epidemiologic Studies Depression Scale (CES-D-8 ≥ 4). The sample was divided into six groups: no pain and no depression (NP/NDe), mild pain and no depression (MP/NDe), moderate/intense pain and no depression (M-IP/NDe), no pain and depression (NP/De), mild pain and depression (MP/De), and moderate/intense pain and depression (M-IP/De). The outcome of interest was performance in memory, executive function, and global cognition. Generalised linear mixed models were used to analyse performance in the cognitive domains and global cognition score as a function of pain and depressive symptoms during 12 years of follow-up. RESULTS Over time, individuals with M-IP/De had a greater memory decline (-0.038 SD/year, 95%CI: -0.068 to -0.007) and the global cognition score (-0.033 SD/year, 95%CI: -0.063 to -0.002) than those with NP/NDe. CONCLUSION The coexistence of moderate/intense pain and depressive symptoms is a risk factor for the decline of global cognition and memory.
Collapse
Affiliation(s)
- Patrícia Silva Tofani
- Department of Physiotherapy, Federal University of Sergipe, Lagarto, Brazil
- Postgraduate Program in Gerontology, Federal University of São Carlos, São Carlos, Brazil
| | | | - Natália Cochar-Soares
- Postgraduate Program in Gerontology, Federal University of São Carlos, São Carlos, Brazil
| | - Paula Camila Ramírez
- Postgraduate Program in Physical Therapy, Federal University of São Carlos, São Carlos, Brazil
- School of Physical Therapy, Industrial University of Santander, Bucaramanga, Colombia
| | - Mariane Marques Luiz
- Postgraduate Program in Physical Therapy, Federal University of São Carlos, São Carlos, Brazil
| | - Sara Souza Lima
- Postgraduate Program in Gerontology, Federal University of São Carlos, São Carlos, Brazil
| | | | | | - Letícia Coelho Silveira
- Postgraduate Program in Physical Therapy, Federal University of São Carlos, São Carlos, Brazil
| | - Valdete Regina Guandalini
- Postgraduate Program in Gerontology, Federal University of São Carlos, São Carlos, Brazil
- Department of Integrated Education, Health Sciences Centre, Federal University of Espírito Santo, Vitória, Brazil
| | - Andrew Steptoe
- Department of Epidemiology and Public Health, University College London, London, United Kingdom
| | - Cesar de Oliveira
- Department of Epidemiology and Public Health, University College London, London, United Kingdom
| | - Tiago da Silva Alexandre
- Postgraduate Program in Gerontology, Federal University of São Carlos, São Carlos, Brazil
- Postgraduate Program in Physical Therapy, Federal University of São Carlos, São Carlos, Brazil
- Department of Epidemiology and Public Health, University College London, London, United Kingdom
- Department of Gerontology, Federal University of São Carlos, São Carlos, Brazil
| |
Collapse
|
13
|
Xu C, Tao Y, Lin Y, Zhu J, Li Z, Li J, Wang M, Huang T, Shi C. Parsing the heterogeneity of depression: a data-driven subgroup derived from cognitive function. Front Psychiatry 2025; 16:1537331. [PMID: 39950172 PMCID: PMC11821656 DOI: 10.3389/fpsyt.2025.1537331] [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: 11/30/2024] [Accepted: 01/13/2025] [Indexed: 02/16/2025] Open
Abstract
Background Increasing evidences suggests that depression is a heterogeneous clinical syndrome. Cognitive deficits in depression are associated with poor psychosocial functioning and worse response to conventional antidepressants. However, a consistent profile of neurocognitive abnormalities in depression remains unclear. Objective We used data-driven parsing of cognitive performance to reveal subgroups present across depressed individuals and then investigate the change pattern of cognitive subgroups across the course in follow-up. Method We assessed cognition in 163 patients with depression using The Chinese Brief Cognitive Test(C-BCT) and the scores were compared with those of 196 healthy controls (HCs). 58 patients were reassessed after 8 weeks. We used K-means cluster analysis to identify cognitive subgroups, and compared clinical variables among these subgroups. A linear mixed-effects model, incorporating time and group (with interaction term: time × group) as fixed effects, was used to assess cognitive changes over time. Stepwise logistic regression analysis was conducted to identify risk factors associated with these subgroups. Results Two distinct neurocognitive subgroups were identified: (1) a cognitive-impaired subgroup with global impairment across all domains assessed by the C-BCT, and (2) a cognitive-preserved subgroup, exhibited intact cognitive function, with performance well within the healthy range. The cognitive-impaired subgroup presented with more severe baseline symptoms, including depressed mood, guilt, suicidality, and poorer work performance. Significant group × time interactions were observed in the Trail Making Test Part A (TMT-A) and Continuous Performance Test (CPT), but not in Symbol Coding or Digit Span tests. Despite partial improvement in TMT-A and CPT tests, the cognitive-impaired subgroup's scores remained lower than those of the cognitive-preserved subgroup across all tests at the study endpoint. Multiple regression analysis indicated that longer illness duration, lower educational levels, and antipsychotic medication use may be risk factors for cognitive impairment. Conclusion This study identifies distinguishable cognitive subgroups in acute depression, thereby confirming the presence of cognitive heterogeneity. The cognitive-impaired subgroup exhibits distinct symptoms and persistent cognitive deficits even after treatment. Screening for cognitive dysfunction may facilitate more targeted interventions. Clinical Trial Registration https://www.chictr.org, identifier ChiCTR2400092796.
Collapse
Affiliation(s)
- Chenyang Xu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Yanbao Tao
- The First Affiliated Hospital of Xinxiang Medical College, Xinxiang, Henan, China
| | - Yunhan Lin
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Jiahui Zhu
- Beijing Key Laboratory of Mental Disorders, National Clinical Research Center for Mental Disorders and National Center for Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China
| | - Zhuoran Li
- Ontario Institute for Studies in Education, University of Toronto, Toronto, ON, Canada
| | - Jiayi Li
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Mingqia Wang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Tao Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Chuan Shi
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| |
Collapse
|
14
|
Zhang W, Dutt R, Lew D, Barch DM, Bijsterbosch JD. Higher amplitudes of visual networks are associated with trait- but not state-depression. Psychol Med 2025; 54:1-12. [PMID: 39757726 PMCID: PMC11769906 DOI: 10.1017/s0033291724003167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 09/09/2024] [Accepted: 11/07/2024] [Indexed: 01/07/2025]
Abstract
Despite depression being a leading cause of global disability, neuroimaging studies have struggled to identify replicable neural correlates of depression or explain limited variance. This challenge may, in part, stem from the intertwined state (current symptoms; variable) and trait (general propensity; stable) experiences of depression.Here, we sought to disentangle state from trait experiences of depression by leveraging a longitudinal cohort and stratifying individuals into four groups: those in remission ('trait depression group'), those with large longitudinal severity changes in depression symptomatology ('state depression group'), and their respective matched control groups (total analytic n = 1030). We hypothesized that spatial network organization would be linked to trait depression due to its temporal stability, whereas functional connectivity between networks would be more sensitive to state-dependent depression symptoms due to its capacity to fluctuate.We identified 15 large-scale probabilistic functional networks from resting-state fMRI data and performed group comparisons on the amplitude, connectivity, and spatial overlap between these networks, using matched control participants as reference. Our findings revealed higher amplitude in visual networks for the trait depression group at the time of remission, in contrast to controls. This observation may suggest altered visual processing in individuals predisposed to developing depression over time. No significant group differences were observed in any other network measures for the trait-control comparison, nor in any measures for the state-control comparison. These results underscore the overlooked contribution of visual networks to the psychopathology of depression and provide evidence for distinct neural correlates between state and trait experiences of depression.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Rosie Dutt
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Biological Sciences Collegiate Division, University of Chicago, Chicago, IL, USA
| | - Daphne Lew
- Center for Biostatistics and Data Science, Institute for Informatics, Data Science, and Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Deanna M. Barch
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- Department of Psychological and Brain Sciences, Washington University in St. Louis, St. Louis, MO, USA
| | | |
Collapse
|
15
|
Liu D, Qiu L, Han L, Wang Y, Wang F, Liu X, Wu J. Prevalence and influencing factors of medication-related burden among patients with late-life depression in typical city of eastern China: a cross-sectional study. BMC Public Health 2024; 24:3521. [PMID: 39696225 DOI: 10.1186/s12889-024-20939-2] [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: 05/19/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024] Open
Abstract
AIM To evaluate the medication-related burden (MRB) of patients with late-life depression (LLD) and its influencing factors in China using the Living with Medicines Questionnaire-3 (LMQ-3), providing reference for reducing the MRB of those patients. METHOD A cross-sectional study was conducted between September 2023 and January 2024 on 588 patients with LLD. LMQ-3 and MRB factors questionnaire were used for data collection. The distribution of variables was assessed using descriptive analysis, while analyses of Mann-Whitney and Kruskal-Wallis were performed to evaluate inter-group differences. To explore the MRB among patients with LLD and influencing factors, multiple linear regression analysis was performed. RESULTS The median (IQR) LMQ-3 score of 588 participants was 102 (18), indicating a moderate MRB level. Regression analysis revealed a significant trend toward higher perceived burden among patients aged 70-79 years old, living in rural areas, receiving more medical insurance settlements, using all cash, taking more than 5 drugs each time, and taking medicine more than 3 times a day (p < 0.05), which were risk factors for higher MRB. Conversely, patients who lived with their children, had an annual household income (including adult children) more than 50,000 Chinese Yuan, and no adverse drug reactions had lower LMQ-3 scores (p < 0.05), which were protective factors. Patients' concerns about medicine, their lack of autonomy in medicine regimens, and the lack of communication between patients and doctors on treatment regimens were the main causes of the burden. CONCLUSIONS Results of this study provided preliminary evidence of the MRB among patients with LLD. Age, residence, living status, annual household income, type of drug payment, quantity and frequency of medication, and adverse reactions significantly affected the perceived medication burden. It is advisable for health policy makers and health care providers to implement appropriate intervention strategies and burden reduction programs for this vulnerable group.
Collapse
Affiliation(s)
- Dan Liu
- Affiliated Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu, 214151, China
| | - Linghe Qiu
- Affiliated Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu, 214151, China
| | - Lu Han
- Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yajing Wang
- Zhengzhou Ninth People's Hospital, 25 Sha Kou Road, Zhengzhou, Henan, 450008, China
| | - Fei Wang
- Affiliated Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu, 214151, China
| | - Xiaowei Liu
- Affiliated Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu, 214151, China.
| | - Jianhong Wu
- Affiliated Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu, 214151, China.
| |
Collapse
|
16
|
Han S, Tian Y, Zheng R, Wen B, Liu L, Liu H, Wei Y, Chen H, Zhao Z, Xia M, Sun X, Wang X, Wei D, Liu B, Huang CC, Zheng Y, Wu Y, Chen T, Cheng Y, Xu X, Gong Q, Si T, Qiu S, Lin CP, Tang Y, Wang F, Qiu J, Xie P, Li L, He Y, Chen Y, Zhang Y, Cheng J. Shared differential factors underlying individual spontaneous neural activity abnormalities in major depressive disorder. Psychol Med 2024:1-19. [PMID: 39588672 DOI: 10.1017/s0033291724002617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
BACKGROUND In contemporary neuroimaging studies, it has been observed that patients with major depressive disorder (MDD) exhibit aberrant spontaneous neural activity, commonly quantified through the amplitude of low-frequency fluctuations (ALFF). However, the substantial individual heterogeneity among patients poses a challenge to reaching a unified conclusion. METHODS To address this variability, our study adopts a novel framework to parse individualized ALFF abnormalities. We hypothesize that individualized ALFF abnormalities can be portrayed as a unique linear combination of shared differential factors. Our study involved two large multi-center datasets, comprising 2424 patients with MDD and 2183 healthy controls. In patients, individualized ALFF abnormalities were derived through normative modeling and further deconstructed into differential factors using non-negative matrix factorization. RESULTS Two positive and two negative factors were identified. These factors were closely linked to clinical characteristics and explained group-level ALFF abnormalities in the two datasets. Moreover, these factors exhibited distinct associations with the distribution of neurotransmitter receptors/transporters, transcriptional profiles of inflammation-related genes, and connectome-informed epicenters, underscoring their neurobiological relevance. Additionally, factor compositions facilitated the identification of four distinct depressive subtypes, each characterized by unique abnormal ALFF patterns and clinical features. Importantly, these findings were successfully replicated in another dataset with different acquisition equipment, protocols, preprocessing strategies, and medication statuses, validating their robustness and generalizability. CONCLUSIONS This research identifies shared differential factors underlying individual spontaneous neural activity abnormalities in MDD and contributes novel insights into the heterogeneity of spontaneous neural activity abnormalities in MDD.
Collapse
Affiliation(s)
- Shaoqiang Han
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Ya Tian
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Ruiping Zheng
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Baohong Wen
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Liang Liu
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Hao Liu
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yarui Wei
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Huafu Chen
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Zongya Zhao
- School of Medical Engineering, Xinxiang Medical University, Xinxiang, Henan Province, China
| | - Mingrui Xia
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
- Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, China
- IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Xiaoyi Sun
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
- Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, China
- IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
- School of Systems Science, Beijing Normal University, Beijing, China
| | - Xiaoqin Wang
- Key Laboratory of Cognition and Personality (SWU), Ministry of Education, Chongqing, China
- Department of Psychology, Southwest University, Chongqing, China
| | - Dongtao Wei
- Key Laboratory of Cognition and Personality (SWU), Ministry of Education, Chongqing, China
- Department of Psychology, Southwest University, Chongqing, China
| | - Bangshan Liu
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Mental Health Institute of Central South University, China National Technology Institute on Mental Disorders, Hunan Key Laboratory of Psychiatry and Mental Health, Hunan Medical Center for Mental Health, Changsha, Hunan, China
| | - Chu-Chung Huang
- Key Laboratory of Brain Functional Genomics (Ministry of Education), Affiliated Mental Health Center (ECNU), School of Psychology and Cognitive Science, East China Normal University, Shanghai, China
| | - Yanting Zheng
- Department of Radiology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yankun Wu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing, China
| | - Taolin Chen
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuqi Cheng
- Department of Psychiatry, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xiufeng Xu
- Department of Psychiatry, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Qiyong Gong
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, Sichuan, China
| | - Tianmei Si
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing, China
| | - Shijun Qiu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Peking University, Beijing, China
| | - Ching-Po Lin
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Education and Research, Taipei City Hospital, Taipei, Taiwan
| | - Yanqing Tang
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Fei Wang
- Department of Psychiatry, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Jiang Qiu
- Key Laboratory of Cognition and Personality (SWU), Ministry of Education, Chongqing, China
- Department of Psychology, Southwest University, Chongqing, China
| | - Peng Xie
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lingjiang Li
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Mental Health Institute of Central South University, China National Technology Institute on Mental Disorders, Hunan Key Laboratory of Psychiatry and Mental Health, Hunan Medical Center for Mental Health, Changsha, Hunan, China
| | - Yong He
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
- Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, China
- IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
- Chinese Institute for Brain Research, Beijing, China
| | - Yuan Chen
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yong Zhang
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jingliang Cheng
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| |
Collapse
|
17
|
Zhou J, Gong L, Liu X, Chen L, Yang Z. Mendelian randomization in Alzheimer's disease and mild cognitive impairment: Hippocampal volume associations. Neuroscience 2024; 561:30-42. [PMID: 39368607 DOI: 10.1016/j.neuroscience.2024.10.007] [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: 04/11/2024] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 10/07/2024]
Abstract
This study investigates the association between cognitive dysfunction and hippocampal volumes in Alzheimer's Disease (AD) and Mild Cognitive Impairment (MCI) using Mendelian randomization. A meta-analysis of 503 healthy controls, 562 MCI patients, and 389 CE patients revealed significant reductions in hippocampal and subregion volumes in MCI and AD compared to controls. While various subregions showed volume reductions, no causal relationship between hippocampal volume and AD was established through Mendelian randomization analysis. In conclusion, significant volume reductions were observed in MCI and AD patients, highlighting the complexity of the relationship between hippocampal volume and cognitive impairment.
Collapse
Affiliation(s)
- Jianguo Zhou
- Department of Radiology, Lianyungang Affiliated Hospital of Nanjing University of Chinese Medicine, Lianyungang 222004, PR China
| | - Lei Gong
- Department of Radiology, The Fourth People's Hospital of Lianyungang, Affiliated Hospital of Nanjing Medical University Kangda, Lianyungang 222000, PR China
| | - Xiaoli Liu
- Department of Rehabilitation, Lianyungang Affiliated Hospital of Nanjing University of Chinese Medicine, Lianyungang 222004, PR China
| | - Liping Chen
- Department of Rehabilitation, Lianyungang Affiliated Hospital of Nanjing University of Chinese Medicine, Lianyungang 222004, PR China
| | - Zhou Yang
- Department of Rehabilitation, Lianyungang Affiliated Hospital of Nanjing University of Chinese Medicine, Lianyungang 222004, PR China.
| |
Collapse
|
18
|
Hu S, Chen K, Xu Q, Wang F, Na W. Assessing the efficacy and safety of combined buspirone and venlafaxine treatment in late-life depression accompanied by cognitive impairment: A randomized controlled trial. Gen Hosp Psychiatry 2024; 91:1-10. [PMID: 39243483 DOI: 10.1016/j.genhosppsych.2024.09.001] [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: 06/08/2024] [Revised: 09/02/2024] [Accepted: 09/02/2024] [Indexed: 09/09/2024]
Abstract
BACKGROUND Late-life depression, often accompanied by cognitive impairment, poses significant clinical challenges owing to its complex etiology and diverse manifestations. While antidepressants like venlafaxine and anxiolytics such as buspirone are effective for treating depression, their effects on cognitive function remain less well-understood. With the aging population increasingly experiencing geriatric depression, there is an urgent need for innovative treatment approaches that address both depressive symptoms and cognitive impairments. OBJECTIVE This study aimed to evaluate the clinical efficacy and safety of combined buspirone and venlafaxine therapy in elderly patients diagnosed with geriatric depression accompanied by cognitive impairment. METHODS A 12-week, randomized controlled trial was conducted involving 170 elderly patients. Participants were randomized into two groups: one receiving venlafaxine alone (control group) and the other receiving a combination of venlafaxine and buspirone (experimental group). The primary analysis was performed using an Intent-to-Treat (ITT) approach with mixed-effects linear models to assess changes in depressive symptoms, cognitive function, and anxiety levels. A supplementary Per-Protocol (PP) analysis, utilizing repeated measures ANOVA, was also conducted. RESULTS The ITT analysis showed that the combination therapy significantly reduced depressive symptoms, as indicated by the HAMD-17 scores (p = 0.033 at week 12). Cognitive function, as measured by MoCA scores, also improved significantly in the experimental group by week 12 (p = 0.025). However, no statistically significant differences were observed in anxiety reduction between the groups (p = 0.127). The PP analysis confirmed these findings, demonstrating consistent improvements in depressive symptoms and cognitive function, particularly in those who completed the full course of treatment. The incidence of adverse events was comparable between groups, primarily mild and manageable symptoms like dry mouth, dizziness, and fatigue. CONCLUSION The combination of buspirone and venlafaxine was found to be effective in reducing depressive symptoms and enhancing cognitive function in elderly patients with geriatric depression. However, the long-term benefits, especially regarding anxiety reduction, require further investigation. Future studies should consider larger sample sizes, longer follow-up periods, and the inclusion of placebo controls to fully assess the efficacy and safety of this treatment approach.
Collapse
Affiliation(s)
- ShuJia Hu
- Department of Psychiatry, Huzhou Third Municipal Hospital, the Affiliated Hospital of Huzhou University, Huzhou 313000, China.
| | - Ke Chen
- Department of Psychiatry, Huzhou Third Municipal Hospital, the Affiliated Hospital of Huzhou University, Huzhou 313000, China
| | - QiuXia Xu
- Department of Psychiatry, Huzhou Third Municipal Hospital, the Affiliated Hospital of Huzhou University, Huzhou 313000, China
| | - Fei Wang
- Department of Psychiatry, Huzhou Third Municipal Hospital, the Affiliated Hospital of Huzhou University, Huzhou 313000, China
| | - WanQiu Na
- Department of Psychiatry, Huzhou Third Municipal Hospital, the Affiliated Hospital of Huzhou University, Huzhou 313000, China
| |
Collapse
|
19
|
Jahanshad N, Lenzini P, Bijsterbosch J. Current best practices and future opportunities for reproducible findings using large-scale neuroimaging in psychiatry. Neuropsychopharmacology 2024; 50:37-51. [PMID: 39117903 PMCID: PMC11526024 DOI: 10.1038/s41386-024-01938-8] [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: 04/16/2024] [Revised: 06/05/2024] [Accepted: 07/09/2024] [Indexed: 08/10/2024]
Abstract
Research into the brain basis of psychopathology is challenging due to the heterogeneity of psychiatric disorders, extensive comorbidities, underdiagnosis or overdiagnosis, multifaceted interactions with genetics and life experiences, and the highly multivariate nature of neural correlates. Therefore, increasingly larger datasets that measure more variables in larger cohorts are needed to gain insights. In this review, we present current "best practice" approaches for using existing databases, collecting and sharing new repositories for big data analyses, and future directions for big data in neuroimaging and psychiatry with an emphasis on contributing to collaborative efforts and the challenges of multi-study data analysis.
Collapse
Affiliation(s)
- Neda Jahanshad
- Mark and Mary Stevens Neuroimaging & Informatics Institute, Keck School of Medicine of USC, Marina del Rey, CA, 90292, USA.
| | - Petra Lenzini
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Janine Bijsterbosch
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO, 63110, USA.
| |
Collapse
|
20
|
Forbes M, Lotfaliany M, Mohebbi M, Reynolds CF, Woods RL, Orchard S, Chong T, Agustini B, O'Neil A, Ryan J, Berk M. Depressive symptoms and cognitive decline in older adults. Int Psychogeriatr 2024; 36:1039-1050. [PMID: 38623851 DOI: 10.1017/s1041610224000541] [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: 01/20/2024] [Revised: 03/15/2024] [Accepted: 03/23/2024] [Indexed: 04/17/2024]
Abstract
OBJECTIVES Few studies have examined the impact of late-life depression trajectories on specific domains of cognitive function. This study aims to delineate how different depressive symptom trajectories specifically affect cognitive function in older adults. DESIGN Prospective longitudinal cohort study. SETTING Australia and the United States of America. PARTICIPANTS In total, 11,035 community-dwelling older adults with a mean age of 75 years. MEASUREMENTS Depressive trajectories were modelled from depressive symptoms according to annual Centre for Epidemiological Studies Depression Scale 10 (CES-D-10) surveys. Four trajectories of depressive symptoms were identified: low ("nondepressed"), consistently mild ("subthreshold depression"), consistently moderate ("persistent depression"), and initially low but increasing ("emerging depression"). Global cognition (Modified Mini-Mental State Examination [3MS]), verbal fluency (Controlled Oral Word Association Test [COWAT]), processing speed (Symbol Digit Modalities Test [SDMT]), episodic memory (Hopkins Verbal Learning Test - Revised [HVLT-R]), and a composite z-score were assessed over a subsequent median 2 years. RESULTS Subthreshold depression predicted impaired performance on the SDMT (Cohen's d -0.04) and composite score (-0.03); emerging depression predicted impaired performance on the SDMT (-0.13), HVLT-R (-0.09), 3 MS (-0.08) and composite score (-0.09); and persistent depression predicted impaired performance on the SDMT (-0.08), 3 MS (-0.11), and composite score (-0.09). CONCLUSIONS Depressive symptoms are associated with later impaired processing speed. These effects are small. Diverse depression trajectories have different impacts on cognitive function.
Collapse
Affiliation(s)
- Malcolm Forbes
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, VIC, Australia
| | - Mojtaba Lotfaliany
- School of Medicine, Barwon Health, Deakin University, The Institute for Mental and Physical Health and Clinical Translation (IMPACT), Geelong, VC, Australia
| | - Mohammadreza Mohebbi
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, VIC, Australia
| | | | - Robyn L Woods
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - Suzanne Orchard
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - Trevor Chong
- School of Psychological Sciences, Turner Institute for Brain and Mental Health, Monash University, Melbourne, VIC, Australia
| | - Bruno Agustini
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, VIC, Australia
| | - Adrienne O'Neil
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, VIC, Australia
| | - Joanne Ryan
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - Michael Berk
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, VIC, Australia
| |
Collapse
|
21
|
Antoniades M, Srinivasan D, Wen J, Erus G, Abdulkadir A, Mamourian E, Melhem R, Hwang G, Cui Y, Govindarajan ST, Chen AA, Zhou Z, Yang Z, Chen J, Pomponio R, Sotardi S, An Y, Bilgel M, LaMontagne P, Singh A, Benzinger T, Beason-Held L, Marcus DS, Yaffe K, Launer L, Morris JC, Tosun D, Ferrucci L, Bryan RN, Resnick SM, Habes M, Wolk D, Fan Y, Nasrallah IM, Shou H, Davatzikos C. Relationship between MRI brain-age heterogeneity, cognition, genetics and Alzheimer's disease neuropathology. EBioMedicine 2024; 109:105399. [PMID: 39437659 PMCID: PMC11536027 DOI: 10.1016/j.ebiom.2024.105399] [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: 11/13/2023] [Revised: 09/24/2024] [Accepted: 09/30/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Brain ageing is highly heterogeneous, as it is driven by a variety of normal and neuropathological processes. These processes may differentially affect structural and functional brain ageing across individuals, with more pronounced ageing (older brain age) during midlife being indicative of later development of dementia. Here, we examined whether brain-ageing heterogeneity in unimpaired older adults related to neurodegeneration, different cognitive trajectories, genetic and amyloid-beta (Aβ) profiles, and to predicted progression to Alzheimer's disease (AD). METHODS Functional and structural brain age measures were obtained for resting-state functional MRI and structural MRI, respectively, in 3460 cognitively normal individuals across an age range spanning 42-85 years. Participants were categorised into four groups based on the difference between their chronological and predicted age in each modality: advanced age in both (n = 291), resilient in both (n = 260) or advanced in one/resilient in the other (n = 163/153). With the resilient group as the reference, brain-age groups were compared across neuroimaging features of neuropathology (white matter hyperintensity volume, neuronal loss measured with Neurite Orientation Dispersion and Density Imaging, AD-specific atrophy patterns measured with the Spatial Patterns of Abnormality for Recognition of Early Alzheimer's Disease index, amyloid burden using amyloid positron emission tomography (PET), progression to mild cognitive impairment and baseline and longitudinal cognitive measures (trail making task, mini mental state examination, digit symbol substitution task). FINDINGS Individuals with advanced structural and functional brain-ages had more features indicative of neurodegeneration and they had poor cognition. Individuals with a resilient brain-age in both modalities had a genetic variant that has been shown to be associated with age of onset of AD. Mixed brain-age was associated with selective cognitive deficits. INTERPRETATION The advanced group displayed evidence of increased atrophy across all neuroimaging features that was not found in either of the mixed groups. This is in line with biomarkers of preclinical AD and cerebrovascular disease. These findings suggest that the variation in structural and functional brain ageing across individuals reflects the degree of underlying neuropathological processes and may indicate the propensity to develop dementia in later life. FUNDING The National Institute on Aging, the National Institutes of Health, the Swiss National Science Foundation, the Kaiser Foundation Research Institute and the National Heart, Lung, and Blood Institute.
Collapse
Affiliation(s)
- Mathilde Antoniades
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA.
| | - Dhivya Srinivasan
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Junhao Wen
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA; Laboratory of AI and Biomedical Science (LABS), University of Southern California, Los Angeles, CA, USA
| | - Guray Erus
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Ahmed Abdulkadir
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA; Department of Clinical Neuroscience, Center for Research in Neuroscience, Lausanne University Hospital, Lausanne, Switzerland
| | - Elizabeth Mamourian
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Randa Melhem
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Gyujoon Hwang
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA; Department of Psychiatry and Behavioral Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Yuhan Cui
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Sindhuja Tirumalai Govindarajan
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew A Chen
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Zhen Zhou
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhijian Yang
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Jiong Chen
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Raymond Pomponio
- Department of Biostatistics, Colorado School of Public Health, Aurora, CO 80045, USA
| | - Susan Sotardi
- Department of Radiology, Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, USA
| | - Yang An
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Murat Bilgel
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Pamela LaMontagne
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Ashish Singh
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, USA
| | - Tammie Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Lori Beason-Held
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Daniel S Marcus
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Lenore Launer
- Neuroepidemiology Section, Intramural Research Program, National Institute on Aging, Bethesda, MD, USA
| | - John C Morris
- Knight Alzheimer Disease Research Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Duygu Tosun
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Luigi Ferrucci
- National Institute on Aging, National Institute of Health, Baltimore, MD 21224, USA
| | - R Nick Bryan
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Susan M Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Mohamad Habes
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA; Neuroimage Analytics Laboratory (NAL) and the Biggs Institute Neuroimaging Core (BINC), Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - David Wolk
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Yong Fan
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Ilya M Nasrallah
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Haochang Shou
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, USA
| | - Christos Davatzikos
- AI(2)D, Center for AI and Data Science for Integrated Diagnostics, and Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
22
|
Huo S, Rivier CA, Clocchiatti-Tuozzo S, Renedo D, Sunmonu NA, de Havenon A, Sarpong DF, Rosendale N, Sheth KN, Falcone GJ. Brain Health Outcomes in Sexual and Gender Minority Groups: Results From the All of Us Research Program. Neurology 2024; 103:e209863. [PMID: 39321407 DOI: 10.1212/wnl.0000000000209863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Sexual and gender minority (SGM) groups have been historically underrepresented in neurologic research, and their brain health disparities are unknown. We aim to evaluate whether SGM persons are at higher risk of adverse brain health outcomes compared with cisgender straight (non-SGM) individuals. METHODS We conducted a cross-sectional study in the All of Us Research Program, a US population-based study, including all participants with information on gender identity and sexual orientation. We used baseline questionnaires to identify sexual minority (lesbian, gay, bisexual, diverse sexual orientation; nonstraight sexual orientation) and gender minority (gender diverse and transgender; gender identity different from sex assigned at birth) participants. The primary outcome was a composite of stroke, dementia, and late-life depression, assessed using electronic health record data and self-report. Secondarily, we evaluated each disease separately. Furthermore, we evaluated all subgroups of gender and sexual minorities stratified by sex assigned at birth. We used multivariable logistic regression (adjusted for age, sex assigned at birth, race/ethnicity, cardiovascular risk factors, other relevant comorbidities, and neighborhood deprivation index) to assess the relationship between SGM groups and the outcomes. RESULTS Of 413,457 US adults enrolled between May 31, 2017, and June 30, 2022, we included 393,041 participants with available information on sexual orientation and gender identity (mean age 51 [SD 17] years), of whom 39,632 (10%) belonged to SGM groups. Of them, 38,528 (97%) belonged to a sexual minority and 4,431 (11%) to a gender minority. Compared with non-SGM, SGM persons had 15% higher odds of the brain health composite outcome (odds ratio [OR] 1.15, 95% CI 1.08-1.22). In secondary analyses, these results persisted across sexual and gender minorities separately (all 95% CIs > 1). Assessing individual diseases, all SGM groups had higher odds of dementia (SGM vs non-SGM: OR 1.14, 95% CI 1.00-1.29) and late-life depression (SGM vs non-SGM: OR 1.27, 95% CI 1.17-1.38) and transgender women had higher odds of stroke (OR 1.68, 95% CI 1.04-2.70). DISCUSSION In a large US population study, SGM persons had higher odds of adverse brain health outcomes. Further research should explore structural causes of inequity to advance inclusive and diverse neurologic care.
Collapse
Affiliation(s)
- Shufan Huo
- From the Department of Neurology (S.H., C.A.R., S.C.-T., D.R., N.A.S., A.d.H., K.N.S., G.J.F.), Yale Center for Brain and Mind Health (S.H., C.A.R., S.C.-T., D.R., A.d.H., K.N.S., G.J.F.), Department of Internal Medicine (S.C.-T.), Department of Neurosurgery (D.R.), and Office of Health Equity Research (D.F.S.), Yale University School of Medicine, New Haven, CT; and Weill Institute for Neurosciences (N.R.), Department of Neurology, University of California San Francisco
| | - Cyprien A Rivier
- From the Department of Neurology (S.H., C.A.R., S.C.-T., D.R., N.A.S., A.d.H., K.N.S., G.J.F.), Yale Center for Brain and Mind Health (S.H., C.A.R., S.C.-T., D.R., A.d.H., K.N.S., G.J.F.), Department of Internal Medicine (S.C.-T.), Department of Neurosurgery (D.R.), and Office of Health Equity Research (D.F.S.), Yale University School of Medicine, New Haven, CT; and Weill Institute for Neurosciences (N.R.), Department of Neurology, University of California San Francisco
| | - Santiago Clocchiatti-Tuozzo
- From the Department of Neurology (S.H., C.A.R., S.C.-T., D.R., N.A.S., A.d.H., K.N.S., G.J.F.), Yale Center for Brain and Mind Health (S.H., C.A.R., S.C.-T., D.R., A.d.H., K.N.S., G.J.F.), Department of Internal Medicine (S.C.-T.), Department of Neurosurgery (D.R.), and Office of Health Equity Research (D.F.S.), Yale University School of Medicine, New Haven, CT; and Weill Institute for Neurosciences (N.R.), Department of Neurology, University of California San Francisco
| | - Daniela Renedo
- From the Department of Neurology (S.H., C.A.R., S.C.-T., D.R., N.A.S., A.d.H., K.N.S., G.J.F.), Yale Center for Brain and Mind Health (S.H., C.A.R., S.C.-T., D.R., A.d.H., K.N.S., G.J.F.), Department of Internal Medicine (S.C.-T.), Department of Neurosurgery (D.R.), and Office of Health Equity Research (D.F.S.), Yale University School of Medicine, New Haven, CT; and Weill Institute for Neurosciences (N.R.), Department of Neurology, University of California San Francisco
| | - N Abimbola Sunmonu
- From the Department of Neurology (S.H., C.A.R., S.C.-T., D.R., N.A.S., A.d.H., K.N.S., G.J.F.), Yale Center for Brain and Mind Health (S.H., C.A.R., S.C.-T., D.R., A.d.H., K.N.S., G.J.F.), Department of Internal Medicine (S.C.-T.), Department of Neurosurgery (D.R.), and Office of Health Equity Research (D.F.S.), Yale University School of Medicine, New Haven, CT; and Weill Institute for Neurosciences (N.R.), Department of Neurology, University of California San Francisco
| | - Adam de Havenon
- From the Department of Neurology (S.H., C.A.R., S.C.-T., D.R., N.A.S., A.d.H., K.N.S., G.J.F.), Yale Center for Brain and Mind Health (S.H., C.A.R., S.C.-T., D.R., A.d.H., K.N.S., G.J.F.), Department of Internal Medicine (S.C.-T.), Department of Neurosurgery (D.R.), and Office of Health Equity Research (D.F.S.), Yale University School of Medicine, New Haven, CT; and Weill Institute for Neurosciences (N.R.), Department of Neurology, University of California San Francisco
| | - Daniel F Sarpong
- From the Department of Neurology (S.H., C.A.R., S.C.-T., D.R., N.A.S., A.d.H., K.N.S., G.J.F.), Yale Center for Brain and Mind Health (S.H., C.A.R., S.C.-T., D.R., A.d.H., K.N.S., G.J.F.), Department of Internal Medicine (S.C.-T.), Department of Neurosurgery (D.R.), and Office of Health Equity Research (D.F.S.), Yale University School of Medicine, New Haven, CT; and Weill Institute for Neurosciences (N.R.), Department of Neurology, University of California San Francisco
| | - Nicole Rosendale
- From the Department of Neurology (S.H., C.A.R., S.C.-T., D.R., N.A.S., A.d.H., K.N.S., G.J.F.), Yale Center for Brain and Mind Health (S.H., C.A.R., S.C.-T., D.R., A.d.H., K.N.S., G.J.F.), Department of Internal Medicine (S.C.-T.), Department of Neurosurgery (D.R.), and Office of Health Equity Research (D.F.S.), Yale University School of Medicine, New Haven, CT; and Weill Institute for Neurosciences (N.R.), Department of Neurology, University of California San Francisco
| | - Kevin N Sheth
- From the Department of Neurology (S.H., C.A.R., S.C.-T., D.R., N.A.S., A.d.H., K.N.S., G.J.F.), Yale Center for Brain and Mind Health (S.H., C.A.R., S.C.-T., D.R., A.d.H., K.N.S., G.J.F.), Department of Internal Medicine (S.C.-T.), Department of Neurosurgery (D.R.), and Office of Health Equity Research (D.F.S.), Yale University School of Medicine, New Haven, CT; and Weill Institute for Neurosciences (N.R.), Department of Neurology, University of California San Francisco
| | - Guido J Falcone
- From the Department of Neurology (S.H., C.A.R., S.C.-T., D.R., N.A.S., A.d.H., K.N.S., G.J.F.), Yale Center for Brain and Mind Health (S.H., C.A.R., S.C.-T., D.R., A.d.H., K.N.S., G.J.F.), Department of Internal Medicine (S.C.-T.), Department of Neurosurgery (D.R.), and Office of Health Equity Research (D.F.S.), Yale University School of Medicine, New Haven, CT; and Weill Institute for Neurosciences (N.R.), Department of Neurology, University of California San Francisco
| |
Collapse
|
23
|
Kim K, Jang YJ, Shin JH, Park MJ, Kim HS, Seong JK, Jeon HJ. Amyloid deposition and its association with depressive symptoms and cognitive functions in late-life depression: a longitudinal study using amyloid-β PET images and neuropsychological measurements. Alzheimers Res Ther 2024; 16:232. [PMID: 39427221 PMCID: PMC11490031 DOI: 10.1186/s13195-024-01562-0] [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: 06/16/2024] [Accepted: 08/18/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND Although depression is linked to an increased risk of dementia, the association between late-onset depression (LOD) and amyloid burden remains unclear. This study aimed to determine amyloid deposition in patients with LOD compared to healthy controls (HC) using amyloid-beta (Aβ) positron emission tomography (PET) images and neuropsychological assessments. METHODS Forty patients first diagnosed with major depressive disorder after the age of 60 (LOD) and twenty-one healthy volunteers (HC) were enrolled. Depression and anxiety were evaluated using the 17-item Hamilton Depression Scale, Hamilton Anxiety Rating Scale, and Clinical Global Impression Scale. Cognitive function was assessed using the Korean versions of the Mini-Mental Status Examination, Montreal Cognitive Assessment, and Seoul Neuropsychological Screening Battery at baseline and 3-month follow-up. 18F-florbetapir PET images were co-registered with T1-weighted magnetic resonance images. RESULTS There was no significant difference in Aβ deposition between LOD and HC groups. No significant correlation between Aβ burden and depressive symptom severity was found in LOD patients. Higher somatic anxiety was correlated with lower Aβ burden in multiple brain regions, including the left inferior frontal lobe (p = 0.009), right anterior cingulate (p = 0.003), and right superior frontal lobe (p = 0.009). Despite cognitive recovery in areas such as attention (Digit Span Forward, p = 0.026), memory (Auditory Verbal Learning Test Recall Total, p = 0.010; Rey Complex Figure Test Delayed Recall, p = 0.039), and frontal executive function (Contrasting Program, p = 0.033) after three months of antidepressant treatment, cognitive improvement showed no association with amyloid deposition. CONCLUSIONS These findings suggest distinct mechanisms may underlie amyloid deposition in neurodegenerative changes associated with depression. While amyloid burden in specific brain regions negatively correlated with somatic anxiety, it showed no significant correlation with the severity of depression or overall cognitive function.
Collapse
Affiliation(s)
- Kiwon Kim
- Workplace Mental Health Institute, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Yoo Jin Jang
- Department of Psychiatry, Depression Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jeong-Hyeon Shin
- Bio Medical Research Center, Bio Medical and Health Division, Korea Testing Laboratory, Seoul, South Korea
| | - Mi Jin Park
- Department of Psychiatry, Seoul St Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Hyun Soo Kim
- Department of Psychiatry, Dong-A University College of Medicine, Busan, South Korea
| | - Joon-Kyung Seong
- School of Biomedical Engineering, College of Health Science, Korea University, Seoul, South Korea
- Department of Artificial Intelligence, Korea University, Seoul, South Korea
| | - Hong Jin Jeon
- Department of Psychiatry, Depression Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.
- Department of Health Sciences and Technology, Department of Medical Device Management and Research, and Department of Clinical Research Design and Evaluation, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, 81 Irwon-Ro, Gangnam-Gu, Seoul, 06351, South Korea.
| |
Collapse
|
24
|
Wen J, Yang Z, Nasrallah IM, Cui Y, Erus G, Srinivasan D, Abdulkadir A, Mamourian E, Hwang G, Singh A, Bergman M, Bao J, Varol E, Zhou Z, Boquet-Pujadas A, Chen J, Toga AW, Saykin AJ, Hohman TJ, Thompson PM, Villeneuve S, Gollub R, Sotiras A, Wittfeld K, Grabe HJ, Tosun D, Bilgel M, An Y, Marcus DS, LaMontagne P, Benzinger TL, Heckbert SR, Austin TR, Launer LJ, Espeland M, Masters CL, Maruff P, Fripp J, Johnson SC, Morris JC, Albert MS, Bryan RN, Resnick SM, Ferrucci L, Fan Y, Habes M, Wolk D, Shen L, Shou H, Davatzikos C. Genetic and clinical correlates of two neuroanatomical AI dimensions in the Alzheimer's disease continuum. Transl Psychiatry 2024; 14:420. [PMID: 39368996 PMCID: PMC11455841 DOI: 10.1038/s41398-024-03121-5] [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: 06/04/2024] [Revised: 09/18/2024] [Accepted: 09/23/2024] [Indexed: 10/07/2024] Open
Abstract
Alzheimer's disease (AD) is associated with heterogeneous atrophy patterns. We employed a semi-supervised representation learning technique known as Surreal-GAN, through which we identified two latent dimensional representations of brain atrophy in symptomatic mild cognitive impairment (MCI) and AD patients: the "diffuse-AD" (R1) dimension shows widespread brain atrophy, and the "MTL-AD" (R2) dimension displays focal medial temporal lobe (MTL) atrophy. Critically, only R2 was associated with widely known sporadic AD genetic risk factors (e.g., APOE ε4) in MCI and AD patients at baseline. We then independently detected the presence of the two dimensions in the early stages by deploying the trained model in the general population and two cognitively unimpaired cohorts of asymptomatic participants. In the general population, genome-wide association studies found 77 genes unrelated to APOE differentially associated with R1 and R2. Functional analyses revealed that these genes were overrepresented in differentially expressed gene sets in organs beyond the brain (R1 and R2), including the heart (R1) and the pituitary gland, muscle, and kidney (R2). These genes were enriched in biological pathways implicated in dendritic cells (R2), macrophage functions (R1), and cancer (R1 and R2). Several of them were "druggable genes" for cancer (R1), inflammation (R1), cardiovascular diseases (R1), and diseases of the nervous system (R2). The longitudinal progression showed that APOE ε4, amyloid, and tau were associated with R2 at early asymptomatic stages, but this longitudinal association occurs only at late symptomatic stages in R1. Our findings deepen our understanding of the multifaceted pathogenesis of AD beyond the brain. In early asymptomatic stages, the two dimensions are associated with diverse pathological mechanisms, including cardiovascular diseases, inflammation, and hormonal dysfunction-driven by genes different from APOE-which may collectively contribute to the early pathogenesis of AD. All results are publicly available at https://labs-laboratory.com/medicine/ .
Collapse
Affiliation(s)
- Junhao Wen
- Laboratory of AI and Biomedical Science (LABS), University of Southern California, Los Angeles, CA, USA.
| | - Zhijian Yang
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ilya M Nasrallah
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yuhan Cui
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Guray Erus
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dhivya Srinivasan
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ahmed Abdulkadir
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Research Lab in Neuroimaging of the Department of Clinical Neurosciences at Lausanne University Hospital, Lausanne, Switzerland
| | - Elizabeth Mamourian
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gyujoon Hwang
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ashish Singh
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mark Bergman
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jingxuan Bao
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Erdem Varol
- Department of Statistics, Center for Theoretical Neuroscience, Zuckerman Institute, Columbia University, New York, NY, USA
| | - Zhen Zhou
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Aleix Boquet-Pujadas
- Laboratory of AI and Biomedical Science (LABS), University of Southern California, Los Angeles, CA, USA
| | - Jiong Chen
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Arthur W Toga
- Laboratory of NeuroImaging, Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Andrew J Saykin
- Radiology and Imaging Sciences, Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana Alzheimer's Disease Research Center and the Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Timothy J Hohman
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt Genetics Institute, Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Paul M Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Marina del Rey, CA, USA
| | - Sylvia Villeneuve
- Douglas Mental Health University Institute, McGill University, Montréal, QC, Canada
| | - Randy Gollub
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Aristeidis Sotiras
- Department of Radiology and Institute for Informatics, Washington University School of Medicine, St. Louis, MO, USA
| | - Katharina Wittfeld
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Hans J Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Duygu Tosun
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Murat Bilgel
- Laboratory of Behavioral Neuroscience, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Yang An
- Laboratory of Behavioral Neuroscience, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Daniel S Marcus
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Pamela LaMontagne
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tammie L Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Susan R Heckbert
- Cardiovascular Health Research Unit and Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Thomas R Austin
- Cardiovascular Health Research Unit and Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Lenore J Launer
- Neuroepidemiology Section, Intramural Research Program, National Institute on Aging, Bethesda, MD, USA
| | - Mark Espeland
- Sticht Center for Healthy Aging and Alzheimer's Prevention, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Colin L Masters
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Paul Maruff
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Jurgen Fripp
- CSIRO Health and Biosecurity, Australian e-Health Research Centre CSIRO, Brisbane, QLD, Australia
| | - Sterling C Johnson
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - John C Morris
- Knight Alzheimer Disease Research Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Marilyn S Albert
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - R Nick Bryan
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Susan M Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Luigi Ferrucci
- Translational Gerontology Branch, Longitudinal Studies Section, National Institute on Aging, National Institutes of Health, MedStar Harbor Hospital, 3001 S. Hanover Street, Baltimore, MD, 21225, USA
| | - Yong Fan
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mohamad Habes
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - David Wolk
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurology and Penn Memory Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Haochang Shou
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christos Davatzikos
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
25
|
Yang Z, Wen J, Erus G, Govindarajan ST, Melhem R, Mamourian E, Cui Y, Srinivasan D, Abdulkadir A, Parmpi P, Wittfeld K, Grabe HJ, Bülow R, Frenzel S, Tosun D, Bilgel M, An Y, Yi D, Marcus DS, LaMontagne P, Benzinger TLS, Heckbert SR, Austin TR, Waldstein SR, Evans MK, Zonderman AB, Launer LJ, Sotiras A, Espeland MA, Masters CL, Maruff P, Fripp J, Toga AW, O'Bryant S, Chakravarty MM, Villeneuve S, Johnson SC, Morris JC, Albert MS, Yaffe K, Völzke H, Ferrucci L, Nick Bryan R, Shinohara RT, Fan Y, Habes M, Lalousis PA, Koutsouleris N, Wolk DA, Resnick SM, Shou H, Nasrallah IM, Davatzikos C. Brain aging patterns in a large and diverse cohort of 49,482 individuals. Nat Med 2024; 30:3015-3026. [PMID: 39147830 PMCID: PMC11483219 DOI: 10.1038/s41591-024-03144-x] [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: 12/31/2023] [Accepted: 06/20/2024] [Indexed: 08/17/2024]
Abstract
Brain aging process is influenced by various lifestyle, environmental and genetic factors, as well as by age-related and often coexisting pathologies. Magnetic resonance imaging and artificial intelligence methods have been instrumental in understanding neuroanatomical changes that occur during aging. Large, diverse population studies enable identifying comprehensive and representative brain change patterns resulting from distinct but overlapping pathological and biological factors, revealing intersections and heterogeneity in affected brain regions and clinical phenotypes. Herein, we leverage a state-of-the-art deep-representation learning method, Surreal-GAN, and present methodological advances and extensive experimental results elucidating brain aging heterogeneity in a cohort of 49,482 individuals from 11 studies. Five dominant patterns of brain atrophy were identified and quantified for each individual by respective measures, R-indices. Their associations with biomedical, lifestyle and genetic factors provide insights into the etiology of observed variances, suggesting their potential as brain endophenotypes for genetic and lifestyle risks. Furthermore, baseline R-indices predict disease progression and mortality, capturing early changes as supplementary prognostic markers. These R-indices establish a dimensional approach to measuring aging trajectories and related brain changes. They hold promise for precise diagnostics, especially at preclinical stages, facilitating personalized patient management and targeted clinical trial recruitment based on specific brain endophenotypic expression and prognosis.
Collapse
Affiliation(s)
- Zhijian Yang
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Graduate Group in Applied Mathematics and Computational Science, University of Pennsylvania, Philadelphia, PA, USA
- GE Healthcare, Bellevue, WA, USA
| | - Junhao Wen
- Laboratory of AI and Biomedical Science (LABS), Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Guray Erus
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sindhuja T Govindarajan
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Randa Melhem
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth Mamourian
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yuhan Cui
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dhivya Srinivasan
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ahmed Abdulkadir
- Laboratory for Research in Neuroimaging, Department of Clinical Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Paraskevi Parmpi
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Katharina Wittfeld
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Hans J Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- Site Rostock/Greifswald, German Center for Neurodegenerative Diseases (DZNE), Greifswald, Germany
| | - Robin Bülow
- Institute of Diagnostic Radiology and Neuroradiology, University of Greifswald, Greifswald, Germany
| | - Stefan Frenzel
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Duygu Tosun
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Murat Bilgel
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Yang An
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Dahyun Yi
- Institute of Human Behavioral Medicine, Medical Research Center Seoul National University, Seoul, Republic of Korea
| | - Daniel S Marcus
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Pamela LaMontagne
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tammie L S Benzinger
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Susan R Heckbert
- Cardiovascular Health Research Unit and Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Thomas R Austin
- Cardiovascular Health Research Unit and Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Shari R Waldstein
- Department of Psychology, University of Maryland, Baltimore County, Baltimore, MD, USA
| | - Michele K Evans
- Health Disparities Research Section, Laboratory of Epidemiology and Population Sciences, NIA/NIH/IRP, Baltimore, MD, USA
| | - Alan B Zonderman
- Health Disparities Research Section, Laboratory of Epidemiology and Population Sciences, NIA/NIH/IRP, Baltimore, MD, USA
| | - Lenore J Launer
- Neuroepidemiology Section, Intramural Research Program, National Institute on Aging, Bethesda, MD, USA
| | - Aristeidis Sotiras
- Department of Radiology and Institute for Informatics, Data Science & Biostatistics, Washington University in St. Louis, St. Louis, MO, USA
| | - Mark A Espeland
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Colin L Masters
- Florey Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Paul Maruff
- Florey Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Jurgen Fripp
- CSIRO Health and Biosecurity, Australian e-Health Research Centre CSIRO, Brisbane, Queensland, Australia
| | - Arthur W Toga
- Laboratory of Neuro Imaging, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Sid O'Bryant
- Institute for Translational Research University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Mallar M Chakravarty
- Computational Brain Anatomy (CoBrA) Laboratory, Cerebral Imaging Center, Douglas Mental Health University Institute, McGill University, Verdun, Quebec, Canada
| | - Sylvia Villeneuve
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Sterling C Johnson
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - John C Morris
- Knight Alzheimer Disease Research Center, Dept of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Marilyn S Albert
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kristine Yaffe
- Departments of Neurology, Psychiatry and Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Luigi Ferrucci
- Translational Gerontology Branch, Longitudinal Studies Section, National Institute on Aging, National Institutes of Health, MedStar Harbor Hospital, Baltimore, MD, USA
| | - R Nick Bryan
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Russell T Shinohara
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, & Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Yong Fan
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mohamad Habes
- Neuroimage Analytics Laboratory and Biggs Institute Neuroimaging Core, Glenn Biggs Institute for Neurodegenerative Disorders, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Paris Alexandros Lalousis
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Nikolaos Koutsouleris
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- Section for Precision Psychiatry, Department of Psychiatry and Psychotherapy, Ludwig-Maximilian-University Munich, Munich, Germany
| | - David A Wolk
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Susan M Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Haochang Shou
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, & Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Ilya M Nasrallah
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Christos Davatzikos
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
26
|
Wen J, Antoniades M, Yang Z, Hwang G, Skampardoni I, Wang R, Davatzikos C. Dimensional Neuroimaging Endophenotypes: Neurobiological Representations of Disease Heterogeneity Through Machine Learning. Biol Psychiatry 2024; 96:564-584. [PMID: 38718880 PMCID: PMC11374488 DOI: 10.1016/j.biopsych.2024.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/29/2024] [Accepted: 04/22/2024] [Indexed: 05/21/2024]
Abstract
Machine learning has been increasingly used to obtain individualized neuroimaging signatures for disease diagnosis, prognosis, and response to treatment in neuropsychiatric and neurodegenerative disorders. Therefore, it has contributed to a better understanding of disease heterogeneity by identifying disease subtypes with different brain phenotypic measures. In this review, we first present a systematic literature overview of studies using machine learning and multimodal magnetic resonance imaging to unravel disease heterogeneity in various neuropsychiatric and neurodegenerative disorders, including Alzheimer's disease, schizophrenia, major depressive disorder, autism spectrum disorder, and multiple sclerosis, as well as their potential in a transdiagnostic framework, where neuroanatomical and neurobiological commonalities were assessed across diagnostic boundaries. Subsequently, we summarize relevant machine learning methodologies and their clinical interpretability. We discuss the potential clinical implications of the current findings and envision future research avenues. Finally, we discuss an emerging paradigm called dimensional neuroimaging endophenotypes. Dimensional neuroimaging endophenotypes dissects the neurobiological heterogeneity of neuropsychiatric and neurodegenerative disorders into low-dimensional yet informative, quantitative brain phenotypic representations, serving as robust intermediate phenotypes (i.e., endophenotypes), presumably reflecting the interplay of underlying genetic, lifestyle, and environmental processes associated with disease etiology.
Collapse
Affiliation(s)
- Junhao Wen
- Laboratory of AI and Biomedical Science, University of Southern California, Los Angeles, California.
| | - Mathilde Antoniades
- Artificial Intelligence in Biomedical Imaging Laboratory, Center for AI and Data Science for Integrated Diagnostics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Zhijian Yang
- Artificial Intelligence in Biomedical Imaging Laboratory, Center for AI and Data Science for Integrated Diagnostics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Gyujoon Hwang
- Psychiatry and Behavioral Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Ioanna Skampardoni
- Artificial Intelligence in Biomedical Imaging Laboratory, Center for AI and Data Science for Integrated Diagnostics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Rongguang Wang
- Artificial Intelligence in Biomedical Imaging Laboratory, Center for AI and Data Science for Integrated Diagnostics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Christos Davatzikos
- Artificial Intelligence in Biomedical Imaging Laboratory, Center for AI and Data Science for Integrated Diagnostics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
27
|
Wen J, Tian YE, Skampardoni I, Yang Z, Cui Y, Anagnostakis F, Mamourian E, Zhao B, Toga AW, Zalesky A, Davatzikos C. The genetic architecture of biological age in nine human organ systems. NATURE AGING 2024; 4:1290-1307. [PMID: 38942983 PMCID: PMC11446180 DOI: 10.1038/s43587-024-00662-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 05/30/2024] [Indexed: 06/30/2024]
Abstract
Investigating the genetic underpinnings of human aging is essential for unraveling the etiology of and developing actionable therapies for chronic diseases. Here, we characterize the genetic architecture of the biological age gap (BAG; the difference between machine learning-predicted age and chronological age) across nine human organ systems in 377,028 participants of European ancestry from the UK Biobank. The BAGs were computed using cross-validated support vector machines, incorporating imaging, physical traits and physiological measures. We identify 393 genomic loci-BAG pairs (P < 5 × 10-8) linked to the brain, eye, cardiovascular, hepatic, immune, metabolic, musculoskeletal, pulmonary and renal systems. Genetic variants associated with the nine BAGs are predominantly specific to the respective organ system (organ specificity) while exerting pleiotropic links with other organ systems (interorgan cross-talk). We find that genetic correlation between the nine BAGs mirrors their phenotypic correlation. Further, a multiorgan causal network established from two-sample Mendelian randomization and latent causal variance models revealed potential causality between chronic diseases (for example, Alzheimer's disease and diabetes), modifiable lifestyle factors (for example, sleep duration and body weight) and multiple BAGs. Our results illustrate the potential for improving human organ health via a multiorgan network, including lifestyle interventions and drug repurposing strategies.
Collapse
Affiliation(s)
- Junhao Wen
- Laboratory of AI and Biomedical Science (LABS), University of Southern California, Los Angeles, CA, USA.
| | - Ye Ella Tian
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Melbourne, Victoria, Australia
| | - Ioanna Skampardoni
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhijian Yang
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yuhan Cui
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Elizabeth Mamourian
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bingxin Zhao
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Arthur W Toga
- Laboratory of Neuro Imaging (LONI), Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Andrew Zalesky
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Melbourne, Victoria, Australia
| | - Christos Davatzikos
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
28
|
Colombo F, Calesella F, Bravi B, Fortaner-Uyà L, Monopoli C, Tassi E, Carminati M, Zanardi R, Bollettini I, Poletti S, Lorenzi C, Spadini S, Brambilla P, Serretti A, Maggioni E, Fabbri C, Benedetti F, Vai B. Multimodal brain-derived subtypes of Major depressive disorder differentiate patients for anergic symptoms, immune-inflammatory markers, history of childhood trauma and treatment-resistance. Eur Neuropsychopharmacol 2024; 85:45-57. [PMID: 38936143 DOI: 10.1016/j.euroneuro.2024.05.015] [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: 11/21/2023] [Revised: 05/20/2024] [Accepted: 05/27/2024] [Indexed: 06/29/2024]
Abstract
An estimated 30 % of Major Depressive Disorder (MDD) patients exhibit resistance to conventional antidepressant treatments. Identifying reliable biomarkers of treatment-resistant depression (TRD) represents a major goal of precision psychiatry, which is hampered by the clinical and biological heterogeneity. To uncover biologically-driven subtypes of MDD, we applied an unsupervised data-driven framework to stratify 102 MDD patients on their neuroimaging signature, including extracted measures of cortical thickness, grey matter volumes, and white matter fractional anisotropy. Our novel analytical pipeline integrated different machine learning algorithms to harmonize data, perform data dimensionality reduction, and provide a stability-based relative clustering validation. The obtained clusters were characterized for immune-inflammatory peripheral biomarkers, TRD, history of childhood trauma and depressive symptoms. Our results indicated two different clusters of patients, differentiable with 67 % of accuracy: one cluster (n = 59) was associated with a higher proportion of TRD, and higher scores of energy-related depressive symptoms, history of childhood abuse and emotional neglect; this cluster showed a widespread reduction in cortical thickness (d = 0.43-1.80) and volumes (d = 0.45-1.05), along with fractional anisotropy in the fronto-occipital fasciculus, stria terminalis, and corpus callosum (d = 0.46-0.52); the second cluster (n = 43) was associated with cognitive and affective depressive symptoms, thicker cortices and wider volumes. Multivariate analyses revealed distinct brain-inflammation relationships between the two clusters, with increase in pro-inflammatory markers being associated with decreased cortical thickness and volumes. Our stratification of MDD patients based on structural neuroimaging identified clinically-relevant subgroups of MDD with specific symptomatic and immune-inflammatory profiles, which can contribute to the development of tailored personalized interventions for MDD.
Collapse
Affiliation(s)
- Federica Colombo
- University Vita-Salute San Raffaele, Milano, Italy; Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, IRCCS San Raffaele Hospital, Milano, Italy.
| | - Federico Calesella
- Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, IRCCS San Raffaele Hospital, Milano, Italy
| | - Beatrice Bravi
- University Vita-Salute San Raffaele, Milano, Italy; Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, IRCCS San Raffaele Hospital, Milano, Italy
| | - Lidia Fortaner-Uyà
- University Vita-Salute San Raffaele, Milano, Italy; Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, IRCCS San Raffaele Hospital, Milano, Italy
| | - Camilla Monopoli
- Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, IRCCS San Raffaele Hospital, Milano, Italy
| | - Emma Tassi
- Department of Neurosciences and Mental Health, IRCCS Fondazione Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Politecnico di Milano, Department of Electronics, Information and Bioengineering, Milan, Italy
| | | | - Raffaella Zanardi
- University Vita-Salute San Raffaele, Milano, Italy; Mood Disorders Unit, Scientific Institute IRCCS San Raffaele Hospital, Milan, Italy
| | - Irene Bollettini
- Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, IRCCS San Raffaele Hospital, Milano, Italy
| | - Sara Poletti
- University Vita-Salute San Raffaele, Milano, Italy; Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, IRCCS San Raffaele Hospital, Milano, Italy
| | - Cristina Lorenzi
- Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, IRCCS San Raffaele Hospital, Milano, Italy
| | - Sara Spadini
- Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, IRCCS San Raffaele Hospital, Milano, Italy
| | - Paolo Brambilla
- Department of Neurosciences and Mental Health, IRCCS Fondazione Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Eleonora Maggioni
- Politecnico di Milano, Department of Electronics, Information and Bioengineering, Milan, Italy
| | - Chiara Fabbri
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Francesco Benedetti
- University Vita-Salute San Raffaele, Milano, Italy; Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, IRCCS San Raffaele Hospital, Milano, Italy
| | - Benedetta Vai
- University Vita-Salute San Raffaele, Milano, Italy; Psychiatry and Clinical Psychobiology Unit, Division of Neuroscience, IRCCS San Raffaele Hospital, Milano, Italy
| |
Collapse
|
29
|
Chen B, Su T, Yang M, Wang Q, Zhou H, Tan G, Liu S, Wu Z, Zhong X, Ning Y. Static and dynamic functional connectivity of the habenula in late-life depression patient with suicidal ideation. J Affect Disord 2024; 356:499-506. [PMID: 38574869 DOI: 10.1016/j.jad.2024.03.161] [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: 12/27/2023] [Revised: 03/25/2024] [Accepted: 03/26/2024] [Indexed: 04/06/2024]
Abstract
BACKGROUND Suicide is one of the most lethal complications of late-life depression (LLD), and habenular dysfunction may be involved in depression-related suicidality and may serve as a potential target for alleviating suicidal ideation. This study aimed to investigate abnormal functional connectivity of the habenula in LLD patients with suicidal ideation. METHODS One hundred twenty-seven patients with LLD (51 with suicidal ideation (LLD-S) and 76 without suicidal ideation (LLD-NS)) and 75 healthy controls (HCs) were recruited. The static functional connectivity (sFC) and dynamic functional connectivity (dFC) between the habenula and the whole brain were compared among the three groups, and correlation and moderation analyses were applied to investigate whether suicidal ideation moderated the relationships of habenular FC with depressive symptoms and cognitive impairment. RESULTS The dFC between the right habenula and the left orbitofrontal cortex (OFC) increased in the following order: LLD-S > LLD-NS > control. No significant difference in the habenular sFC was found among the LLD-S, LLD-NS and control groups. The dFC between the right habenula and the left OFC was positively associated with global cognitive function and visuospatial skills, and the association between this dFC and visuospatial skills was moderated by suicidal ideation in patients with LLD. CONCLUSION The increased variability in dFC between the right habenula and left OFC was more pronounced in the LLD-S group than in the LLD-NS group, and the association between habenular-OFC dFC and visuospatial skills was moderated by suicidal ideation in patients with LLD.
Collapse
Affiliation(s)
- Ben Chen
- Geriatric Neuroscience Center, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ting Su
- Department of Radiology, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Mingfeng Yang
- Geriatric Neuroscience Center, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qiang Wang
- Geriatric Neuroscience Center, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Huarong Zhou
- Geriatric Neuroscience Center, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Guili Tan
- Department of Rehabilitation, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Siting Liu
- Department of Radiology, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhangying Wu
- Geriatric Neuroscience Center, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaomei Zhong
- Geriatric Neuroscience Center, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Yuping Ning
- Geriatric Neuroscience Center, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China Guangzhou Medical University, Guangzhou, China; The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, Guangzhou, China; Guangdong Engineering Technology Research Center for Translational Medicine of Mental Disorders, Guangzhou, China.
| |
Collapse
|
30
|
Zheng J, Zong X, Tang L, Guo H, Zhao P, Womer FY, Zhang X, Tang Y, Wang F. Characterizing the distinct imaging phenotypes, clinical behavior, and genetic vulnerability of brain maturational subtypes in mood disorders. Psychol Med 2024; 54:2774-2784. [PMID: 38804091 DOI: 10.1017/s0033291724000886] [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: 05/29/2024]
Abstract
BACKGROUND Mood disorders are characterized by great heterogeneity in clinical manifestation. Uncovering such heterogeneity using neuroimaging-based individual biomarkers, clinical behaviors, and genetic risks, might contribute to elucidating the etiology of these diseases and support precision medicine. METHODS We recruited 174 drug-naïve and drug-free patients with major depressive disorder and bipolar disorder, as well as 404 healthy controls. T1 MRI imaging data, clinical symptoms, and neurocognitive assessments, and genetics were obtained and analyzed. We applied regional gray matter volumes (GMV) and quantile normative modeling to create maturation curves, and then calculated individual deviations to identify subtypes within the patients using hierarchical clustering. We compared the between-subtype differences in GMV deviations, clinical behaviors, cell-specific transcriptomic associations, and polygenic risk scores. We also validated the GMV deviations based subtyping analysis in a replication cohort. RESULTS Two subtypes emerged: subtype 1, characterized by increased GMV deviations in the frontal cortex, cognitive impairment, a higher genetic risk for Alzheimer's disease, and transcriptionally associated with Alzheimer's disease pathways, oligodendrocytes, and endothelial cells; and subtype 2, displaying globally decreased GMV deviations, more severe depressive symptoms, increased genetic vulnerability to major depressive disorder and transcriptionally related to microglia and inhibitory neurons. The distinct patterns of GMV deviations in the frontal, cingulate, and primary motor cortices between subtypes were shown to be replicable. CONCLUSIONS Our current results provide vital links between MRI-derived phenotypes, spatial transcriptome, genetic vulnerability, and clinical manifestation, and uncover the heterogeneity of mood disorders in biological and behavioral terms.
Collapse
Affiliation(s)
- Junjie Zheng
- Early Intervention Unit, Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
- Functional Brain Imaging Institute of Nanjing Medical University, Nanjing, China
| | - Xiaofen Zong
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Lili Tang
- Early Intervention Unit, Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
- Functional Brain Imaging Institute of Nanjing Medical University, Nanjing, China
| | - Huiling Guo
- Early Intervention Unit, Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
- Functional Brain Imaging Institute of Nanjing Medical University, Nanjing, China
- School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
| | - Pengfei Zhao
- Early Intervention Unit, Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
- Functional Brain Imaging Institute of Nanjing Medical University, Nanjing, China
| | - Fay Y Womer
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Xizhe Zhang
- School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
| | - Yanqing Tang
- Department of Psychiatry, The First Hospital of China Medical University, Shenyang, China
- Brain Function Research Section, The First Hospital of China Medical University, Shenyang, China
- Department of Gerontology, The First Hospital of China Medical University, Shenyang, China
- Department of Psychiatry, Shengjing Hospital of China Medical University, Shenyang, China
| | - Fei Wang
- Early Intervention Unit, Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
- Functional Brain Imaging Institute of Nanjing Medical University, Nanjing, China
- Department of Mental Health, School of Public Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
31
|
Wen J, Tian YE, Skampardoni I, Yang Z, Cui Y, Anagnostakis F, Mamourian E, Zhao B, Toga AW, Zaleskey A, Davatzikos C. The Genetic Architecture of Biological Age in Nine Human Organ Systems. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.06.08.23291168. [PMID: 37398441 PMCID: PMC10312870 DOI: 10.1101/2023.06.08.23291168] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Understanding the genetic basis of biological aging in multi-organ systems is vital for elucidating age-related disease mechanisms and identifying therapeutic interventions. This study characterized the genetic architecture of the biological age gap (BAG) across nine human organ systems in 377,028 individuals of European ancestry from the UK Biobank. We discovered 393 genomic loci-BAG pairs (P-value<5×10-8) linked to the brain, eye, cardiovascular, hepatic, immune, metabolic, musculoskeletal, pulmonary, and renal systems. We observed BAG-organ specificity and inter-organ connections. Genetic variants associated with the nine BAGs are predominantly specific to the respective organ system while exerting pleiotropic effects on traits linked to multiple organ systems. A gene-drug-disease network confirmed the involvement of the metabolic BAG-associated genes in drugs targeting various metabolic disorders. Genetic correlation analyses supported Cheverud's Conjecture1 - the genetic correlation between BAGs mirrors their phenotypic correlation. A causal network revealed potential causal effects linking chronic diseases (e.g., Alzheimer's disease), body weight, and sleep duration to the BAG of multiple organ systems. Our findings shed light on promising therapeutic interventions to enhance human organ health within a complex multi-organ network, including lifestyle modifications and potential drug repositioning strategies for treating chronic diseases. All results are publicly available at https://labs-laboratory.com/medicine.
Collapse
Affiliation(s)
- Junhao Wen
- Laboratory of AI and Biomedical Science (LABS), Keck School of Medicine of USC, University of Southern California, Los Angeles, California, USA
| | - Ye Ella Tian
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Melbourne, Victoria, Australia
| | - Ioanna Skampardoni
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Zhijian Yang
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Yuhan Cui
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Filippos Anagnostakis
- Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy
| | - Elizabeth Mamourian
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Bingxin Zhao
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Arthur W. Toga
- Laboratory of Neuro Imaging (LONI), Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, California, USA
| | - Andrew Zaleskey
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Melbourne, Victoria, Australia
| | - Christos Davatzikos
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| |
Collapse
|
32
|
Kuai H, Chen J, Tao X, Cai L, Imamura K, Matsumoto H, Liang P, Zhong N. Never-Ending Learning for Explainable Brain Computing. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307647. [PMID: 38602432 PMCID: PMC11200082 DOI: 10.1002/advs.202307647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 03/24/2024] [Indexed: 04/12/2024]
Abstract
Exploring the nature of human intelligence and behavior is a longstanding pursuit in cognitive neuroscience, driven by the accumulation of knowledge, information, and data across various studies. However, achieving a unified and transparent interpretation of findings presents formidable challenges. In response, an explainable brain computing framework is proposed that employs the never-ending learning paradigm, integrating evidence combination and fusion computing within a Knowledge-Information-Data (KID) architecture. The framework supports continuous brain cognition investigation, utilizing joint knowledge-driven forward inference and data-driven reverse inference, bolstered by the pre-trained language modeling techniques and the human-in-the-loop mechanisms. In particular, it incorporates internal evidence learning through multi-task functional neuroimaging analyses and external evidence learning via topic modeling of published neuroimaging studies, all of which involve human interactions at different stages. Based on two case studies, the intricate uncertainty surrounding brain localization in human reasoning is revealed. The present study also highlights the potential of systematization to advance explainable brain computing, offering a finer-grained understanding of brain activity patterns related to human intelligence.
Collapse
Affiliation(s)
- Hongzhi Kuai
- Faculty of EngineeringMaebashi Institute of TechnologyGunma371–0816Japan
- School of Psychology and Beijing Key Laboratory of Learning and CognitionCapital Normal UniversityBeijing100048China
| | - Jianhui Chen
- Faculty of Information TechnologyBeijing University of TechnologyBeijing100124China
- Beijing International Collaboration Base on Brain Informatics and Wisdom ServicesBeijing100124China
| | - Xiaohui Tao
- School of Mathematics, Physics and ComputingUniversity of Southern QueenslandToowoomba4350Australia
| | - Lingyun Cai
- School of Psychology and Beijing Key Laboratory of Learning and CognitionCapital Normal UniversityBeijing100048China
| | - Kazuyuki Imamura
- Faculty of EngineeringMaebashi Institute of TechnologyGunma371–0816Japan
| | - Hiroki Matsumoto
- Faculty of EngineeringMaebashi Institute of TechnologyGunma371–0816Japan
| | - Peipeng Liang
- School of Psychology and Beijing Key Laboratory of Learning and CognitionCapital Normal UniversityBeijing100048China
| | - Ning Zhong
- Faculty of EngineeringMaebashi Institute of TechnologyGunma371–0816Japan
- School of Psychology and Beijing Key Laboratory of Learning and CognitionCapital Normal UniversityBeijing100048China
- Beijing International Collaboration Base on Brain Informatics and Wisdom ServicesBeijing100124China
| |
Collapse
|
33
|
Huang D, Wu Y, Yue J, Wang X. Causal relationship between resting-state networks and depression: a bidirectional two-sample mendelian randomization study. BMC Psychiatry 2024; 24:402. [PMID: 38811927 PMCID: PMC11138044 DOI: 10.1186/s12888-024-05857-2] [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: 11/30/2023] [Accepted: 05/20/2024] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND Cerebral resting-state networks were suggested to be strongly associated with depressive disorders. However, the causal relationship between cerebral networks and depressive disorders remains controversial. In this study, we aimed to investigate the effect of resting-state networks on depressive disorders using a bidirectional Mendelian randomization (MR) design. METHODS Updated summary-level genome-wide association study (GWAS) data correlated with resting-state networks were obtained from a meta-analysis of European-descent GWAS from the Complex Trait Genetics Lab. Depression-related GWAS data were obtained from the FinnGen study involving participants with European ancestry. Resting-state functional magnetic resonance imaging and multiband diffusion imaging of the brain were performed to measure functional and structural connectivity in seven well-known networks. Inverse-variance weighting was used as the primary estimate, whereas the MR-Pleiotropy RESidual Sum and Outliers (PRESSO), MR-Egger, and weighted median were used to detect heterogeneity, sensitivity, and pleiotropy. RESULTS In total, 20,928 functional and 20,573 structural connectivity data as well as depression-related GWAS data from 48,847 patients and 225,483 controls were analyzed. Evidence for a causal effect of the structural limbic network on depressive disorders was found in the inverse variance-weighted limbic network (odds ratio, [Formula: see text]; 95% confidence interval, [Formula: see text]; [Formula: see text]), whereas the causal effect of depressive disorders on SC LN was not found(OR=1.0025; CI,1.0005-1.0046; P=0.012). No significant associations between functional connectivity of the resting-state networks and depressive disorders were found in this MR study. CONCLUSIONS These results suggest that genetically determined structural connectivity of the limbic network has a causal effect on depressive disorders and may play a critical role in its neuropathology.
Collapse
Affiliation(s)
- Dongmiao Huang
- Department of Psychiatry, the Fifth Affiliated Hospital of Sun Yat-sen University, No. 52, East Meihua Road, Zhuhai City, Guangdong Province, 519000, China
| | - Yuelin Wu
- Department of Psychiatry, the Fifth Affiliated Hospital of Sun Yat-sen University, No. 52, East Meihua Road, Zhuhai City, Guangdong Province, 519000, China
| | - Jihui Yue
- Department of Psychiatry, the Fifth Affiliated Hospital of Sun Yat-sen University, No. 52, East Meihua Road, Zhuhai City, Guangdong Province, 519000, China.
| | - Xianglan Wang
- Department of Psychiatry, the Fifth Affiliated Hospital of Sun Yat-sen University, No. 52, East Meihua Road, Zhuhai City, Guangdong Province, 519000, China.
| |
Collapse
|
34
|
Siafarikas N. Personalized medicine in old age psychiatry and Alzheimer's disease. Front Psychiatry 2024; 15:1297798. [PMID: 38751423 PMCID: PMC11094449 DOI: 10.3389/fpsyt.2024.1297798] [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: 09/21/2023] [Accepted: 04/15/2024] [Indexed: 05/18/2024] Open
Abstract
Elderly patients show us unfolded lives with unique individual characteristics. An increasing life span is associated with increasing physical and mental disease burden. Alzheimer's disease (AD) is an increasing challenge in old age. AD cannot be cured but it can be treated. The complexity of old age and AD offer targets for personalized medicine (PM). Targets for stratification of patients, detection of patients at risk for AD or for future targeted therapy are plentiful and can be found in several omic-levels.
Collapse
Affiliation(s)
- Nikias Siafarikas
- Department of Geriatric Psychiatry, Akershus University Hospital, Lørenskog, Norway
| |
Collapse
|
35
|
Skampardoni I, Nasrallah IM, Abdulkadir A, Wen J, Melhem R, Mamourian E, Erus G, Doshi J, Singh A, Yang Z, Cui Y, Hwang G, Ren Z, Pomponio R, Srinivasan D, Govindarajan ST, Parmpi P, Wittfeld K, Grabe HJ, Bülow R, Frenzel S, Tosun D, Bilgel M, An Y, Marcus DS, LaMontagne P, Heckbert SR, Austin TR, Launer LJ, Sotiras A, Espeland MA, Masters CL, Maruff P, Fripp J, Johnson SC, Morris JC, Albert MS, Bryan RN, Yaffe K, Völzke H, Ferrucci L, Benzinger TLS, Ezzati A, Shinohara RT, Fan Y, Resnick SM, Habes M, Wolk D, Shou H, Nikita K, Davatzikos C. Genetic and Clinical Correlates of AI-Based Brain Aging Patterns in Cognitively Unimpaired Individuals. JAMA Psychiatry 2024; 81:456-467. [PMID: 38353984 PMCID: PMC10867779 DOI: 10.1001/jamapsychiatry.2023.5599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 11/29/2023] [Indexed: 02/17/2024]
Abstract
Importance Brain aging elicits complex neuroanatomical changes influenced by multiple age-related pathologies. Understanding the heterogeneity of structural brain changes in aging may provide insights into preclinical stages of neurodegenerative diseases. Objective To derive subgroups with common patterns of variation in participants without diagnosed cognitive impairment (WODCI) in a data-driven manner and relate them to genetics, biomedical measures, and cognitive decline trajectories. Design, Setting, and Participants Data acquisition for this cohort study was performed from 1999 to 2020. Data consolidation and harmonization were conducted from July 2017 to July 2021. Age-specific subgroups of structural brain measures were modeled in 4 decade-long intervals spanning ages 45 to 85 years using a deep learning, semisupervised clustering method leveraging generative adversarial networks. Data were analyzed from July 2021 to February 2023 and were drawn from the Imaging-Based Coordinate System for Aging and Neurodegenerative Diseases (iSTAGING) international consortium. Individuals WODCI at baseline spanning ages 45 to 85 years were included, with greater than 50 000 data time points. Exposures Individuals WODCI at baseline scan. Main Outcomes and Measures Three subgroups, consistent across decades, were identified within the WODCI population. Associations with genetics, cardiovascular risk factors (CVRFs), amyloid β (Aβ), and future cognitive decline were assessed. Results In a sample of 27 402 individuals (mean [SD] age, 63.0 [8.3] years; 15 146 female [55%]) WODCI, 3 subgroups were identified in contrast with the reference group: a typical aging subgroup, A1, with a specific pattern of modest atrophy and white matter hyperintensity (WMH) load, and 2 accelerated aging subgroups, A2 and A3, with characteristics that were more distinct at age 65 years and older. A2 was associated with hypertension, WMH, and vascular disease-related genetic variants and was enriched for Aβ positivity (ages ≥65 years) and apolipoprotein E (APOE) ε4 carriers. A3 showed severe, widespread atrophy, moderate presence of CVRFs, and greater cognitive decline. Genetic variants associated with A1 were protective for WMH (rs7209235: mean [SD] B = -0.07 [0.01]; P value = 2.31 × 10-9) and Alzheimer disease (rs72932727: mean [SD] B = 0.1 [0.02]; P value = 6.49 × 10-9), whereas the converse was observed for A2 (rs7209235: mean [SD] B = 0.1 [0.01]; P value = 1.73 × 10-15 and rs72932727: mean [SD] B = -0.09 [0.02]; P value = 4.05 × 10-7, respectively); variants in A3 were associated with regional atrophy (rs167684: mean [SD] B = 0.08 [0.01]; P value = 7.22 × 10-12) and white matter integrity measures (rs1636250: mean [SD] B = 0.06 [0.01]; P value = 4.90 × 10-7). Conclusions and Relevance The 3 subgroups showed distinct associations with CVRFs, genetics, and subsequent cognitive decline. These subgroups likely reflect multiple underlying neuropathologic processes and affect susceptibility to Alzheimer disease, paving pathways toward patient stratification at early asymptomatic stages and promoting precision medicine in clinical trials and health care.
Collapse
Affiliation(s)
- Ioanna Skampardoni
- Centre for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
- School of Electrical and Computer Engineering, National Technical University of Athens, Greece
| | - Ilya M Nasrallah
- Centre for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
- Department of Radiology, University of Pennsylvania, Philadelphia
| | - Ahmed Abdulkadir
- Centre for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
- Laboratory for Research in Neuroimaging, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Junhao Wen
- Centre for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
- Laboratory of AI and Biomedical Science, Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles
| | - Randa Melhem
- Centre for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
| | - Elizabeth Mamourian
- Centre for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
| | - Guray Erus
- Centre for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
| | - Jimit Doshi
- Centre for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
| | - Ashish Singh
- Centre for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
| | - Zhijian Yang
- Centre for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
| | - Yuhan Cui
- Centre for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
| | - Gyujoon Hwang
- Centre for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
| | - Zheng Ren
- Laboratory of AI and Biomedical Science, Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles
| | - Raymond Pomponio
- Centre for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
| | - Dhivya Srinivasan
- Centre for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
| | | | - Paraskevi Parmpi
- Centre for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
| | - Katharina Wittfeld
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Germany
- German Centre for Neurodegenerative Diseases, Site Greifswald, Greifswald, Germany
| | - Hans J Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Germany
- German Centre for Neurodegenerative Diseases, Site Greifswald, Greifswald, Germany
| | - Robin Bülow
- Institute of Diagnostic Radiology and Neuroradiology, University Medicine Greifswald, Greifswald, Germany
| | - Stefan Frenzel
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Germany
| | - Duygu Tosun
- Department of Radiology and Biomedical Imaging, University of California, San Francisco
| | - Murat Bilgel
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Yang An
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Daniel S Marcus
- Department of Radiology, Washington University School of Medicine, St Louis, Missouri
| | - Pamela LaMontagne
- Department of Radiology, Washington University School of Medicine, St Louis, Missouri
| | - Susan R Heckbert
- Cardiovascular Health Research Unit, University of Washington, Seattle
- Department of Epidemiology, University of Washington, Seattle
| | - Thomas R Austin
- Cardiovascular Health Research Unit, University of Washington, Seattle
- Department of Epidemiology, University of Washington, Seattle
| | - Lenore J Launer
- Neuroepidemiology Section, Intramural Research Program, National Institute on Aging, Bethesda, Maryland
| | - Aristeidis Sotiras
- Department of Radiology and Institute of Informatics, Washington University in St Louis, St Louis, Missouri
| | - Mark A Espeland
- Sticht Centre for Healthy Aging and Alzheimer's Prevention, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Colin L Masters
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Paul Maruff
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Jurgen Fripp
- CSIRO Health and Biosecurity, Australian e-Health Research Centre CSIRO, Brisbane, Queensland, Australia
| | - Sterling C Johnson
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison
| | - John C Morris
- Knight Alzheimer Disease Research Centre, Washington University in St Louis, St Louis, Missouri
| | - Marilyn S Albert
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - R Nick Bryan
- Department of Radiology, University of Pennsylvania, Philadelphia
| | - Kristine Yaffe
- Departments of Neurology, Psychiatry and Epidemiology and Biostatistics, University of California San Francisco, San Francisco
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Tammie L S Benzinger
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St Louis, St Louis, Missouri
| | - Ali Ezzati
- Department of Neurology, University of California, Irvine
| | - Russell T Shinohara
- Centre for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, & Informatics, University of Pennsylvania, Philadelphia
| | - Yong Fan
- Centre for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
| | - Susan M Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Mohamad Habes
- Centre for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
- Neuroimage Analytics Laboratory and the Biggs Institute Neuroimaging Core, Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio
| | - David Wolk
- Department of Neurology, University of Pennsylvania, Philadelphia
| | - Haochang Shou
- Centre for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, & Informatics, University of Pennsylvania, Philadelphia
| | - Konstantina Nikita
- School of Electrical and Computer Engineering, National Technical University of Athens, Greece
| | - Christos Davatzikos
- Centre for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia
| |
Collapse
|
36
|
Wang J, Liang X, Qiu Q, Yan F, Fang Y, Shen C, Wang H, Chen Y, Xiao S, Yue L, Li X. Cognitive trajectories in older adults and the role of depressive symptoms: A 7-year follow-up study. Asian J Psychiatr 2024; 95:104007. [PMID: 38520944 DOI: 10.1016/j.ajp.2024.104007] [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: 09/27/2023] [Revised: 01/06/2024] [Accepted: 03/16/2024] [Indexed: 03/25/2024]
Abstract
OBJECTIVES To examine different trajectories of cognitive changes in elderly adults and explore the mediating role of depressive symptoms. DESIGN A 7-year, community-based, prospective cohort study. SETTING The downtown neighborhood of Shanghai, China. PARTICIPANTS A cohort of 394 older adults, with an average age of 71.8 years, was recruited in 2015 and has been reassessed every two years until 2021. METHODS Latent Class Growth Analysis was used to model aging trajectories and Linear Mixed-Effect Models for Repeated Measures were used to estimate the least squares mean changes of cognition between subjects with depression (DEP+) and without (DEP-) across all visits. RESULTS Three cognitive trajectories were identified: the "successful aging" (SA) trajectory had the best and most consistent performance (n=229, 55.9%); the "normal aging" (NA) trajectory showed lower but stable cognition (n=141, 37.3%); while the "cognitive decline" (CD) trajectory displayed poor and declining cognition (n=24, 6.8%). Depressive symptoms were found to be influential across all trajectories. In the CD trajectory, the MoCA scores of the DEP+ group increased in within-group comparisons and were significantly higher than those of the DEP- group at visits 1 and 3 in between-group comparisons. A similar trend was observed in the NA trajectory, though it did not reach statistical significance. CONCLUSIONS Our research suggests that mild and decreasing depressive symptoms can be a reversible factor that might slow down the irreversible cognitive decline in the elderly. Therefore, we suggest that even mild depressive symptoms in the elderly should be monitored and detected.
Collapse
Affiliation(s)
- Jianjun Wang
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, Shanghai 200030, China; Department of Neurology and Psychology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen 518000, China
| | - Xiao Liang
- Shanghai Xuhui District Mental Health Center, Shanghai 200232, China
| | - Qi Qiu
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Feng Yan
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yuan Fang
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Changyi Shen
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Huijuan Wang
- Shanghai Jingan District Mental Health Center, Shanghai 200040, China
| | - Yuming Chen
- Shanghai Jingan District Mental Health Center, Shanghai 200040, China
| | - Shifu Xiao
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Ling Yue
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, Shanghai 200030, China.
| | - Xia Li
- Department of Geriatric Psychiatry, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; Alzheimer's Disease and Related Disorders Center, Shanghai Jiao Tong University, Shanghai 200030, China.
| |
Collapse
|
37
|
Su T, Chen B, Yang M, Wang Q, Zhou H, Zhang M, Wu Z, Lin G, Wang D, Li Y, Zhong X, Ning Y. Disrupted functional connectivity of the habenula links psychomotor retardation and deficit of verbal fluency and working memory in late-life depression. CNS Neurosci Ther 2024; 30:e14490. [PMID: 37804094 PMCID: PMC11017447 DOI: 10.1111/cns.14490] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/02/2023] [Accepted: 09/23/2023] [Indexed: 10/08/2023] Open
Abstract
BACKGROUND Functional abnormalities of the habenula in patients with depression have been demonstrated in an increasing number of studies, and the habenula is involved in cognitive processing. However, whether patients with late-life depression (LLD) exhibit disrupted habenular functional connectivity (FC) and whether habenular FC mediates the relationship between depressive symptoms and cognitive impairment remain unclear. METHODS Overall, 127 patients with LLD and 75 healthy controls were recruited. The static and dynamic FC between the habenula and the whole brain was compared between LLD patients and healthy controls, and the relationships of habenular FC with depressive symptoms and cognitive impairment were explored by correlation and mediation analyses. RESULTS Compared with the controls, patients with LLD exhibited decreased static FC between the right habenula and bilateral inferior frontal gyrus (IFG); there was no significant difference in dynamic FC of the habenula between the two groups. Additionally, the decreased static FC between the right habenula and IFG was associated with more severe depressive symptoms (especially psychomotor retardation) and cognitive impairment (language, memory, and visuospatial skills). Last, static FC between the right habenula and left IFG partially mediated the relationship between depressive symptoms (especially psychomotor retardation) and cognitive impairment (verbal fluency and working memory). CONCLUSIONS Patients with LLD exhibited decreased static FC between the habenula and IFG but intact dynamic FC of the habenula. This decreased static FC mediated the relationship between depressive symptoms and cognitive impairment.
Collapse
Affiliation(s)
- Ting Su
- Department of RadiologyThe Affiliated Brain Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Ben Chen
- Geriatric Neuroscience CenterThe Affiliated Brain Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Mingfeng Yang
- Geriatric Neuroscience CenterThe Affiliated Brain Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Qiang Wang
- Geriatric Neuroscience CenterThe Affiliated Brain Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Huarong Zhou
- Geriatric Neuroscience CenterThe Affiliated Brain Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Min Zhang
- Geriatric Neuroscience CenterThe Affiliated Brain Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Zhangying Wu
- Geriatric Neuroscience CenterThe Affiliated Brain Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Gaohong Lin
- Geriatric Neuroscience CenterThe Affiliated Brain Hospital of Guangzhou Medical UniversityGuangzhouChina
| | | | - Yue Li
- Guangzhou Medical UniversityGuangzhouChina
| | - Xiaomei Zhong
- Geriatric Neuroscience CenterThe Affiliated Brain Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Yuping Ning
- Geriatric Neuroscience CenterThe Affiliated Brain Hospital of Guangzhou Medical UniversityGuangzhouChina
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China Guangzhou Medical UniversityGuangzhouChina
- The First School of Clinical MedicineSouthern Medical UniversityGuangzhouChina
- Guangdong Engineering Technology Research Center for Translational Medicine of Mental DisordersGuangzhouChina
| |
Collapse
|
38
|
Wen J, Zhao B, Yang Z, Erus G, Skampardoni I, Mamourian E, Cui Y, Hwang G, Bao J, Boquet-Pujadas A, Zhou Z, Veturi Y, Ritchie MD, Shou H, Thompson PM, Shen L, Toga AW, Davatzikos C. The genetic architecture of multimodal human brain age. Nat Commun 2024; 15:2604. [PMID: 38521789 PMCID: PMC10960798 DOI: 10.1038/s41467-024-46796-6] [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: 05/13/2023] [Accepted: 03/06/2024] [Indexed: 03/25/2024] Open
Abstract
The complex biological mechanisms underlying human brain aging remain incompletely understood. This study investigated the genetic architecture of three brain age gaps (BAG) derived from gray matter volume (GM-BAG), white matter microstructure (WM-BAG), and functional connectivity (FC-BAG). We identified sixteen genomic loci that reached genome-wide significance (P-value < 5×10-8). A gene-drug-disease network highlighted genes linked to GM-BAG for treating neurodegenerative and neuropsychiatric disorders and WM-BAG genes for cancer therapy. GM-BAG displayed the most pronounced heritability enrichment in genetic variants within conserved regions. Oligodendrocytes and astrocytes, but not neurons, exhibited notable heritability enrichment in WM and FC-BAG, respectively. Mendelian randomization identified potential causal effects of several chronic diseases on brain aging, such as type 2 diabetes on GM-BAG and AD on WM-BAG. Our results provide insights into the genetics of human brain aging, with clinical implications for potential lifestyle and therapeutic interventions. All results are publicly available at https://labs.loni.usc.edu/medicine .
Collapse
Affiliation(s)
- Junhao Wen
- Laboratory of AI and Biomedical Science (LABS), Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA.
| | - Bingxin Zhao
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhijian Yang
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Guray Erus
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ioanna Skampardoni
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth Mamourian
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yuhan Cui
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gyujoon Hwang
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jingxuan Bao
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | | | - Zhen Zhou
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yogasudha Veturi
- Department of Biobehavioral Health and Statistics, Penn State University, University Park, PA, USA
| | - Marylyn D Ritchie
- Department of Genetics and Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Haochang Shou
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Paul M Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Marina del Rey, CA, USA
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Arthur W Toga
- Laboratory of Neuro Imaging (LONI), Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Christos Davatzikos
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
39
|
Pan C, Cheng S, Liu L, Chen Y, Meng P, Yang X, Li C, Zhang J, Zhang Z, Zhang H, Cheng B, Wen Y, Jia Y, Zhang F. Identification of novel rare variants for anxiety: an exome-wide association study in the UK Biobank. Prog Neuropsychopharmacol Biol Psychiatry 2024; 130:110928. [PMID: 38154517 DOI: 10.1016/j.pnpbp.2023.110928] [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: 07/10/2023] [Revised: 11/19/2023] [Accepted: 12/23/2023] [Indexed: 12/30/2023]
Abstract
BACKGROUND Rare variants are believed to play a substantial role in the genetic architecture of mental disorders, particularly in coding regions. However, limited evidence supports the impact of rare variants on anxiety. METHODS Using whole-exome sequencing data from 200,643 participants in the UK Biobank, we investigated the contribution of rare variants to anxiety. Firstly, we computed genetic risk score (GRS) of anxiety utilizing genotype data and summary data from a genome-wide association study (GWAS) on anxiety disorder. Subsequently, we identified individuals within the lowest 50% GRS, a subgroup more likely to carry pathogenic rare variants. Within this subgroup, we classified individuals with the highest 10% 7-item Generalized Anxiety Disorder scale (GAD-7) score as cases (N = 1869), and those with the lowest 10% GAD-7 score were designated as controls (N = 1869). Finally, we conducted gene-based burden tests and single-variant association analyses to assess the relationship between rare variants and anxiety. RESULTS Totally, 47,800 variants with MAF ≤0.01 were annotated as non-benign coding variants, consisting of 42,698 nonsynonymous SNVs, 489 nonframeshift substitution, 236 frameshift substitution, 617 stop-gain and 40 stop-loss variants. After variation aggregation, 5066 genes were included in gene-based association analysis. Totally, 11 candidate genes were detected in burden test, such as RNF123 (PBonferroni adjusted = 3.40 × 10-6), MOAP1(PBonferroni adjusted = 4.35 × 10-4), CCDC110 (PBonferroni adjusted = 5.83 × 10-4). Single-variant test detected 9 rare variants, such as rs35726701(RNF123)(PBonferroni adjusted = 3.16 × 10-10) and rs16942615(CAMTA2) (PBonferroni adjusted = 4.04 × 10-4). Notably, RNF123, CCDC110, DNAH2, and CSKMT gene were identified in both tests. CONCLUSIONS Our study identified novel candidate genes for anxiety in protein-coding regions, revealing the contribution of rare variants to anxiety.
Collapse
Affiliation(s)
- Chuyu Pan
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, P. R. China
| | - Shiqiang Cheng
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, P. R. China
| | - Li Liu
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, P. R. China
| | - Yujing Chen
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, P. R. China
| | - Peilin Meng
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, P. R. China
| | - Xuena Yang
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, P. R. China
| | - Chun'e Li
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, P. R. China
| | - Jingxi Zhang
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, P. R. China
| | - Zhen Zhang
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, P. R. China
| | - Huijie Zhang
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, P. R. China
| | - Bolun Cheng
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, P. R. China
| | - Yan Wen
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, P. R. China
| | - Yumeng Jia
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, P. R. China
| | - Feng Zhang
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education of China, Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, P. R. China.
| |
Collapse
|
40
|
Chen S, Wang K, Wang H, Gao Y, Nie K, Jiang X, Su H, Tang Y, Lu F, Dong H, Wang Z. The therapeutic effects of saikosaponins on depression through the modulation of neuroplasticity: From molecular mechanisms to potential clinical applications. Pharmacol Res 2024; 201:107090. [PMID: 38309381 DOI: 10.1016/j.phrs.2024.107090] [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: 12/02/2023] [Revised: 01/07/2024] [Accepted: 01/30/2024] [Indexed: 02/05/2024]
Abstract
Depression is a major global health issue that urgently requires innovative and precise treatment options. In this context, saikosaponin has emerged as a promising candidate, offering a variety of therapeutic benefits that may be effective in combating depression. This review delves into the multifaceted potential of saikosaponins in alleviating depressive symptoms. We summarized the effects of saikosaponins on structural and functional neuroplasticity, elaborated the regulatory mechanism of saikosaponins in modulating key factors that affect neuroplasticity, such as inflammation, the hypothalamic-pituitary-adrenal (HPA) axis, oxidative stress, and the brain-gut axis. Moreover, this paper highlights existing gaps in current researches and outlines directions for future studies. A detailed plan is provided for the future clinical application of saikosaponins, advocating for more targeted researches to speed up its transition from preclinical trials to clinical practice.
Collapse
Affiliation(s)
- Shen Chen
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ke Wang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Hongzhan Wang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yang Gao
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Kexin Nie
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xinyue Jiang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Hao Su
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Yueheng Tang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Fuer Lu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Hui Dong
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Zhi Wang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| |
Collapse
|
41
|
Li J, Long Z, Sheng W, Du L, Qiu J, Chen H, Liao W. Transcriptomic Similarity Informs Neuromorphic Deviations in Depression Biotypes. Biol Psychiatry 2024; 95:414-425. [PMID: 37573006 DOI: 10.1016/j.biopsych.2023.08.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/14/2023]
Abstract
BACKGROUND Major depressive disorder (MDD) is complicated by population heterogeneity, motivating the investigation of biotypes through imaging-derived phenotypes. However, neuromorphic heterogeneity in MDD remains unclear, and how the correlated gene expression (CGE) connectome constrains these neuromorphic anomalies in MDD biotypes has not yet been studied. METHODS Here, we related cortical thickness deviations in MDD biotypes to a pattern of CGE connectome. Cortical thickness was estimated from 3-dimensional T1-weighted magnetic resonance images in 2 independent cohorts (discovery cohort: N = 425; replication cohort: N = 217). The transcriptional activity was measured according to Allen Human Brain Atlas. A density peak-based clustering algorithm was used to identify MDD biotypes. RESULTS We found that patients with MDD were clustered into 2 replicated biotypes based on single-patient regional deviations from healthy control participants across 2 datasets. Biotype 1 mainly exhibited cortical thinning across the brain, whereas biotype 2 mainly showed cortical thickening in the brain. Using brainwide gene expression data, we found that deviations of transcriptionally connected neighbors predicted regional deviation for both biotypes. Furthermore, putative CGE-informed epicenters of biotype 1 were concentrated on the cognitive control circuit, whereas biotype 2 epicenters were located in the social perception circuit. The patterns of epicenter likelihood were separately associated with depression- and anxiety-response maps, suggesting that epicenters of MDD biotypes may be associated with clinical efficacies. CONCLUSIONS Our findings linked the CGE connectome and neuromorphic deviations to identify distinct epicenters in MDD biotypes, providing insight into how microscale gene expressions informed MDD biotypes.
Collapse
Affiliation(s)
- Jiao Li
- Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, P.R. China; MOE Key Laboratory for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, P.R. China
| | - Zhiliang Long
- Key Laboratory of Cognition and Personality, Faculty of Psychology, Southwest University, Chongqing, P.R. China
| | - Wei Sheng
- Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, P.R. China; MOE Key Laboratory for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, P.R. China
| | - Lian Du
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Jiang Qiu
- Key Laboratory of Cognition and Personality, Faculty of Psychology, Southwest University, Chongqing, P.R. China
| | - Huafu Chen
- Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, P.R. China; MOE Key Laboratory for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, P.R. China
| | - Wei Liao
- Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, P.R. China; MOE Key Laboratory for Neuroinformation, High-Field Magnetic Resonance Brain Imaging Key Laboratory of Sichuan Province, University of Electronic Science and Technology of China, Chengdu, P.R. China.
| |
Collapse
|
42
|
Young AL, Oxtoby NP, Garbarino S, Fox NC, Barkhof F, Schott JM, Alexander DC. Data-driven modelling of neurodegenerative disease progression: thinking outside the black box. Nat Rev Neurosci 2024; 25:111-130. [PMID: 38191721 DOI: 10.1038/s41583-023-00779-6] [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] [Accepted: 11/30/2023] [Indexed: 01/10/2024]
Abstract
Data-driven disease progression models are an emerging set of computational tools that reconstruct disease timelines for long-term chronic diseases, providing unique insights into disease processes and their underlying mechanisms. Such methods combine a priori human knowledge and assumptions with large-scale data processing and parameter estimation to infer long-term disease trajectories from short-term data. In contrast to 'black box' machine learning tools, data-driven disease progression models typically require fewer data and are inherently interpretable, thereby aiding disease understanding in addition to enabling classification, prediction and stratification. In this Review, we place the current landscape of data-driven disease progression models in a general framework and discuss their enhanced utility for constructing a disease timeline compared with wider machine learning tools that construct static disease profiles. We review the insights they have enabled across multiple neurodegenerative diseases, notably Alzheimer disease, for applications such as determining temporal trajectories of disease biomarkers, testing hypotheses about disease mechanisms and uncovering disease subtypes. We outline key areas for technological development and translation to a broader range of neuroscience and non-neuroscience applications. Finally, we discuss potential pathways and barriers to integrating disease progression models into clinical practice and trial settings.
Collapse
Affiliation(s)
- Alexandra L Young
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK.
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| | - Neil P Oxtoby
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK.
| | - Sara Garbarino
- Life Science Computational Laboratory, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Nick C Fox
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Frederik Barkhof
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK
- Department of Radiology & Nuclear Medicine, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Jonathan M Schott
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Daniel C Alexander
- UCL Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK
| |
Collapse
|
43
|
Chao X, Wang J, Dong Y, Fang Y, Yin D, Wen J, Wang P, Sun W. Neuroimaging of neuropsychological disturbances following ischaemic stroke (CONNECT): a prospective cohort study protocol. BMJ Open 2024; 14:e077799. [PMID: 38286706 PMCID: PMC10826587 DOI: 10.1136/bmjopen-2023-077799] [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: 07/15/2023] [Accepted: 01/09/2024] [Indexed: 01/31/2024] Open
Abstract
INTRODUCTION Neuropsychiatric distubance is a common clinical manifestation in acute ischemic stroke. However, it is frequently overlooked by clinicians. This study aimed to explore the possible aetiology and pathogenesis of neuropsychiatric disturbances following ischaemic stroke (NDIS) from an anatomical and functional perspective with the help of neuroimaging methods. METHOD AND ANALYSIS CONNECT is a prospective cohort study of neuroimaging and its functional outcome in NDIS. We aim to enrol a minimum of 300 individuals with first-ever stroke. The neuropsychological disturbances involved in this study include depression, anxiety disorder, headache, apathy, insomnia, fatigue and cognitive impairment. Using scales that have been shown to be effective in assessing the above symptoms, the NDIS evaluation battery requires at least 2 hours at baseline. Moreover, all patients will be required to complete 2 years of follow-up, during which the NDIS will be re-evaluated at 3 months, 12 months and 24 months by telephone and 6 months by outpatient interview after the index stroke. The primary outcome of our study is the incidence of NDIS at the 6-month mark. Secondary outcomes are related to the severity of NDIS as well as functional rehabilitation of patients. Functional imaging evaluation will be performed at baseline and 6-month follow-up using specific sequences including resting-state functional MRI, diffusion tensor imaging, T1-weighted imaging, T2-weighted imaging, diffusion-weighted imaging, arterial spin labelling, quantitative susceptibility mapping and fluid-attenuated inversion recovery imaging. In addition, we collect haematological information from patients to explore potential biological and genetic markers of NDIS through histological analysis. ETHICS AND DISSEMINATION The CONNECT Study was approved by the Ethics Review Committee of the First Hospital of the University of Science and Technology of China (2021-ky012) and written informed consent will be obtained from all participants. Results will be disseminated via a peer-reviewed journal. TRIAL REGISTRATION NUMBER ChiCTR2100043886.
Collapse
Affiliation(s)
- Xian Chao
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Jinjing Wang
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yiran Dong
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Yirong Fang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Dawei Yin
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Jie Wen
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Peng Wang
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Wen Sun
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| |
Collapse
|
44
|
Hannon K, Bijsterbosch J. Challenges in Identifying Individualized Brain Biomarkers of Late Life Depression. ADVANCES IN GERIATRIC MEDICINE AND RESEARCH 2024; 5:e230010. [PMID: 38348374 PMCID: PMC10861244 DOI: 10.20900/agmr20230010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/15/2024]
Abstract
Research into neuroimaging biomarkers for Late Life Depression (LLD) has identified neural correlates of LLD including increased white matter hyperintensities and reduced hippocampal volume. However, studies into neuroimaging biomarkers for LLD largely fail to converge. This lack of replicability is potentially due to challenges linked to construct variability, etiological heterogeneity, and experimental rigor. We discuss suggestions to help address these challenges, including improved construct standardization, increased sample sizes, multimodal approaches to parse heterogeneity, and the use of individualized analytical models.
Collapse
Affiliation(s)
- Kayla Hannon
- Department of Radiology, Washington University in St Louis, St Louis MO, 63110, USA
| | - Janine Bijsterbosch
- Department of Radiology, Washington University in St Louis, St Louis MO, 63110, USA
| |
Collapse
|
45
|
Wen J, Antoniades M, Yang Z, Hwang G, Skampardoni I, Wang R, Davatzikos C. Dimensional Neuroimaging Endophenotypes: Neurobiological Representations of Disease Heterogeneity Through Machine Learning. ARXIV 2024:arXiv:2401.09517v1. [PMID: 38313197 PMCID: PMC10836087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 02/06/2024]
Abstract
Machine learning has been increasingly used to obtain individualized neuroimaging signatures for disease diagnosis, prognosis, and response to treatment in neuropsychiatric and neurodegenerative disorders. Therefore, it has contributed to a better understanding of disease heterogeneity by identifying disease subtypes that present significant differences in various brain phenotypic measures. In this review, we first present a systematic literature overview of studies using machine learning and multimodal MRI to unravel disease heterogeneity in various neuropsychiatric and neurodegenerative disorders, including Alzheimer's disease, schizophrenia, major depressive disorder, autism spectrum disorder, multiple sclerosis, as well as their potential in transdiagnostic settings. Subsequently, we summarize relevant machine learning methodologies and discuss an emerging paradigm which we call dimensional neuroimaging endophenotype (DNE). DNE dissects the neurobiological heterogeneity of neuropsychiatric and neurodegenerative disorders into a low-dimensional yet informative, quantitative brain phenotypic representation, serving as a robust intermediate phenotype (i.e., endophenotype) largely reflecting underlying genetics and etiology. Finally, we discuss the potential clinical implications of the current findings and envision future research avenues.
Collapse
Affiliation(s)
- Junhao Wen
- Laboratory of AI and Biomedical Science (LABS), Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Mathilde Antoniades
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhijian Yang
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gyujoon Hwang
- Psychiatry and Behavioral Medicine, Medical College of Wisconsin, Watertown Plank Rd, Milwaukee, WI, USA
| | - Ioanna Skampardoni
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rongguang Wang
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christos Davatzikos
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
46
|
Fu CHY, Antoniades M, Erus G, Garcia JA, Fan Y, Arnone D, Arnott SR, Chen T, Choi KS, Fatt CC, Frey BN, Frokjaer VG, Ganz M, Godlewska BR, Hassel S, Ho K, McIntosh AM, Qin K, Rotzinger S, Sacchet MD, Savitz J, Shou H, Singh A, Stolicyn A, Strigo I, Strother SC, Tosun D, Victor TA, Wei D, Wise T, Zahn R, Anderson IM, Craighead WE, Deakin JFW, Dunlop BW, Elliott R, Gong Q, Gotlib IH, Harmer CJ, Kennedy SH, Knudsen GM, Mayberg HS, Paulus MP, Qiu J, Trivedi MH, Whalley HC, Yan CG, Young AH, Davatzikos C. Neuroanatomical dimensions in medication-free individuals with major depressive disorder and treatment response to SSRI antidepressant medications or placebo. NATURE. MENTAL HEALTH 2024; 2:164-176. [PMID: 38948238 PMCID: PMC11211072 DOI: 10.1038/s44220-023-00187-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 11/17/2023] [Indexed: 07/02/2024]
Abstract
Major depressive disorder (MDD) is a heterogeneous clinical syndrome with widespread subtle neuroanatomical correlates. Our objective was to identify the neuroanatomical dimensions that characterize MDD and predict treatment response to selective serotonin reuptake inhibitor (SSRI) antidepressants or placebo. In the COORDINATE-MDD consortium, raw MRI data were shared from international samples (N = 1,384) of medication-free individuals with first-episode and recurrent MDD (N = 685) in a current depressive episode of at least moderate severity, but not treatment-resistant depression, as well as healthy controls (N = 699). Prospective longitudinal data on treatment response were available for a subset of MDD individuals (N = 359). Treatments were either SSRI antidepressant medication (escitalopram, citalopram, sertraline) or placebo. Multi-center MRI data were harmonized, and HYDRA, a semi-supervised machine-learning clustering algorithm, was utilized to identify patterns in regional brain volumes that are associated with disease. MDD was optimally characterized by two neuroanatomical dimensions that exhibited distinct treatment responses to placebo and SSRI antidepressant medications. Dimension 1 was characterized by preserved gray and white matter (N = 290 MDD), whereas Dimension 2 was characterized by widespread subtle reductions in gray and white matter (N = 395 MDD) relative to healthy controls. Although there were no significant differences in age of onset, years of illness, number of episodes, or duration of current episode between dimensions, there was a significant interaction effect between dimensions and treatment response. Dimension 1 showed a significant improvement in depressive symptoms following treatment with SSRI medication (51.1%) but limited changes following placebo (28.6%). By contrast, Dimension 2 showed comparable improvements to either SSRI (46.9%) or placebo (42.2%) (β = -18.3, 95% CI (-34.3 to -2.3), P = 0.03). Findings from this case-control study indicate that neuroimaging-based markers can help identify the disease-based dimensions that constitute MDD and predict treatment response.
Collapse
Affiliation(s)
- Cynthia H. Y. Fu
- School of Psychology, University of East London, London, UK
- Centre for Affective Disorders, Department of Psychological Medicine, Institute of Psychiatry, Psychology, and Neuroscience, King’s College London, London, UK
| | - Mathilde Antoniades
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Guray Erus
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Jose A. Garcia
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Yong Fan
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Danilo Arnone
- Centre for Affective Disorders, Department of Psychological Medicine, Institute of Psychiatry, Psychology, and Neuroscience, King’s College London, London, UK
| | | | - Taolin Chen
- Huaxi MR Research Center, Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, China
| | - Ki Sueng Choi
- Nash Family Center for Advanced Circuit Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Cherise Chin Fatt
- Department of Psychiatry, Center for Depression Research and Clinical Care, University of Texas Southwestern Medical Center, Dallas, TX USA
| | - Benicio N. Frey
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario Canada
- Mood Disorders Treatment and Research Centre and Women’s Health Concerns Clinic, St Joseph’s Healthcare Hamilton, Hamilton, Ontario Canada
| | - Vibe G. Frokjaer
- Neurobiology Research Unit, University Hospital Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Psychiatry, Psychiatric Centre Copenhagen, Copenhagen, Denmark
| | - Melanie Ganz
- Neurobiology Research Unit, University Hospital Rigshospitalet, Copenhagen, Denmark
- Department of Computer Science, University of Copenhagen, Copenhagen, Denmark
| | - Beata R. Godlewska
- Department of Psychiatry, University of Oxford, Oxford, UK
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
| | - Stefanie Hassel
- Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, Alberta Canada
- Department of Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, Alberta Canada
| | - Keith Ho
- Department of Psychiatry, University Health Network, Toronto, Ontario Canada
| | - Andrew M. McIntosh
- Division of Psychiatry, Royal Edinburgh Hospital, University of Edinburgh, Edinburgh, UK
| | - Kun Qin
- Huaxi MR Research Center, Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, China
- Department of Radiology, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Susan Rotzinger
- Department of Psychiatry, University Health Network, Toronto, Ontario Canada
- Centre for Depression and Suicide Studies, Unity Health Toronto, Toronto, Ontario Canada
| | - Matthew D. Sacchet
- Meditation Research Program, Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | | | - Haochang Shou
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
- Penn Statistics in Imaging and Visualization Endeavor (PennSIVE) Center, Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA USA
| | - Ashish Singh
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Aleks Stolicyn
- Division of Psychiatry, Royal Edinburgh Hospital, University of Edinburgh, Edinburgh, UK
| | - Irina Strigo
- Department of Psychiatry, University of California San Francisco, San Francisco, USA
| | - Stephen C. Strother
- Rotman Research Institute, Baycrest Centre, Toronto, Ontario Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario Canada
| | - Duygu Tosun
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA USA
| | | | - Dongtao Wei
- School of Psychology, Southwest University, Chongqing, China
| | - Toby Wise
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Roland Zahn
- Centre for Affective Disorders, Department of Psychological Medicine, Institute of Psychiatry, Psychology, and Neuroscience, King’s College London, London, UK
| | - Ian M. Anderson
- Division of Neuroscience and Experimental Psychology, University of Manchester, Manchester, UK
| | - W. Edward Craighead
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA USA
- Department of Psychology, Emory University, Atlanta, GA USA
| | - J. F. William Deakin
- Division of Neuroscience and Experimental Psychology, University of Manchester, Manchester, UK
| | - Boadie W. Dunlop
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA USA
| | - Rebecca Elliott
- Division of Neuroscience and Experimental Psychology, University of Manchester, Manchester, UK
| | - Qiyong Gong
- Huaxi MR Research Center, Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, China
| | - Ian H. Gotlib
- Department of Psychology, Stanford University, Stanford, CA USA
| | | | - Sidney H. Kennedy
- Department of Psychiatry, University Health Network, Toronto, Ontario Canada
- Centre for Depression and Suicide Studies, Unity Health Toronto, Toronto, Ontario Canada
| | - Gitte M. Knudsen
- Neurobiology Research Unit, University Hospital Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Helen S. Mayberg
- Nash Family Center for Advanced Circuit Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | | | - Jiang Qiu
- School of Psychology, Southwest University, Chongqing, China
| | - Madhukar H. Trivedi
- Department of Psychiatry, Center for Depression Research and Clinical Care, University of Texas Southwestern Medical Center, Dallas, TX USA
| | - Heather C. Whalley
- Division of Psychiatry, Royal Edinburgh Hospital, University of Edinburgh, Edinburgh, UK
| | - Chao-Gan Yan
- Key Laboratory of Behavioral Science, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| | - Allan H. Young
- Centre for Affective Disorders, Department of Psychological Medicine, Institute of Psychiatry, Psychology, and Neuroscience, King’s College London, London, UK
- South London and Maudsley NHS Foundation Trust, Bethlem Royal Hospital, London, UK
| | - Christos Davatzikos
- Center for Biomedical Image Computing and Analytics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| |
Collapse
|
47
|
Yang Z, Wen J, Abdulkadir A, Cui Y, Erus G, Mamourian E, Melhem R, Srinivasan D, Govindarajan ST, Chen J, Habes M, Masters CL, Maruff P, Fripp J, Ferrucci L, Albert MS, Johnson SC, Morris JC, LaMontagne P, Marcus DS, Benzinger TLS, Wolk DA, Shen L, Bao J, Resnick SM, Shou H, Nasrallah IM, Davatzikos C. Gene-SGAN: discovering disease subtypes with imaging and genetic signatures via multi-view weakly-supervised deep clustering. Nat Commun 2024; 15:354. [PMID: 38191573 PMCID: PMC10774282 DOI: 10.1038/s41467-023-44271-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 12/06/2023] [Indexed: 01/10/2024] Open
Abstract
Disease heterogeneity has been a critical challenge for precision diagnosis and treatment, especially in neurologic and neuropsychiatric diseases. Many diseases can display multiple distinct brain phenotypes across individuals, potentially reflecting disease subtypes that can be captured using MRI and machine learning methods. However, biological interpretability and treatment relevance are limited if the derived subtypes are not associated with genetic drivers or susceptibility factors. Herein, we describe Gene-SGAN - a multi-view, weakly-supervised deep clustering method - which dissects disease heterogeneity by jointly considering phenotypic and genetic data, thereby conferring genetic correlations to the disease subtypes and associated endophenotypic signatures. We first validate the generalizability, interpretability, and robustness of Gene-SGAN in semi-synthetic experiments. We then demonstrate its application to real multi-site datasets from 28,858 individuals, deriving subtypes of Alzheimer's disease and brain endophenotypes associated with hypertension, from MRI and single nucleotide polymorphism data. Derived brain phenotypes displayed significant differences in neuroanatomical patterns, genetic determinants, biological and clinical biomarkers, indicating potentially distinct underlying neuropathologic processes, genetic drivers, and susceptibility factors. Overall, Gene-SGAN is broadly applicable to disease subtyping and endophenotype discovery, and is herein tested on disease-related, genetically-associated neuroimaging phenotypes.
Collapse
Affiliation(s)
- Zhijian Yang
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Graduate Group in Applied Mathematics and Computational Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Junhao Wen
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Laboratory of AI and Biomedical Science (LABS), Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Ahmed Abdulkadir
- Laboratory for Research in Neuroimaging, Department of Clinical Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Yuhan Cui
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Guray Erus
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth Mamourian
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Randa Melhem
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dhivya Srinivasan
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sindhuja T Govindarajan
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jiong Chen
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mohamad Habes
- Biggs Alzheimer's Institute, University of Texas San Antonio Health Science Center, San Antonio, TX, USA
| | - Colin L Masters
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Paul Maruff
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Jurgen Fripp
- CSIRO Health and Biosecurity, Australian e-Health Research Centre CSIRO, Brisbane, QLD, Australia
| | - Luigi Ferrucci
- Translational Gerontology Branch, Longitudinal Studies Section, National Institute on Aging, National Institutes of Health, MedStar Harbor Hospital, 3001 S. Hanover Street, Baltimore, MD, USA
| | - Marilyn S Albert
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sterling C Johnson
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - John C Morris
- Knight Alzheimer Disease Research Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Pamela LaMontagne
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Daniel S Marcus
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tammie L S Benzinger
- Knight Alzheimer Disease Research Center, Washington University in St. Louis, St. Louis, MO, USA
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - David A Wolk
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Jingxuan Bao
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Susan M Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA
| | - Haochang Shou
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Ilya M Nasrallah
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Christos Davatzikos
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
48
|
Veitch DP, Weiner MW, Miller M, Aisen PS, Ashford MA, Beckett LA, Green RC, Harvey D, Jack CR, Jagust W, Landau SM, Morris JC, Nho KT, Nosheny R, Okonkwo O, Perrin RJ, Petersen RC, Rivera Mindt M, Saykin A, Shaw LM, Toga AW, Tosun D. The Alzheimer's Disease Neuroimaging Initiative in the era of Alzheimer's disease treatment: A review of ADNI studies from 2021 to 2022. Alzheimers Dement 2024; 20:652-694. [PMID: 37698424 PMCID: PMC10841343 DOI: 10.1002/alz.13449] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/27/2023] [Accepted: 08/01/2023] [Indexed: 09/13/2023]
Abstract
The Alzheimer's Disease Neuroimaging Initiative (ADNI) aims to improve Alzheimer's disease (AD) clinical trials. Since 2006, ADNI has shared clinical, neuroimaging, and cognitive data, and biofluid samples. We used conventional search methods to identify 1459 publications from 2021 to 2022 using ADNI data/samples and reviewed 291 impactful studies. This review details how ADNI studies improved disease progression understanding and clinical trial efficiency. Advances in subject selection, detection of treatment effects, harmonization, and modeling improved clinical trials and plasma biomarkers like phosphorylated tau showed promise for clinical use. Biomarkers of amyloid beta, tau, neurodegeneration, inflammation, and others were prognostic with individualized prediction algorithms available online. Studies supported the amyloid cascade, emphasized the importance of neuroinflammation, and detailed widespread heterogeneity in disease, linked to genetic and vascular risk, co-pathologies, sex, and resilience. Biological subtypes were consistently observed. Generalizability of ADNI results is limited by lack of cohort diversity, an issue ADNI-4 aims to address by enrolling a diverse cohort.
Collapse
Affiliation(s)
- Dallas P. Veitch
- Department of Veterans Affairs Medical CenterNorthern California Institute for Research and Education (NCIRE)San FranciscoCaliforniaUSA
- Department of Veterans Affairs Medical CenterCenter for Imaging of Neurodegenerative DiseasesSan FranciscoCaliforniaUSA
| | - Michael W. Weiner
- Department of Veterans Affairs Medical CenterCenter for Imaging of Neurodegenerative DiseasesSan FranciscoCaliforniaUSA
- Department of Radiology and Biomedical ImagingUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Department of MedicineUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Department of Psychiatry and Behavioral SciencesUniversity of CaliforniaSan FranciscoCaliforniaUSA
- Department of NeurologyUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Melanie Miller
- Department of Veterans Affairs Medical CenterNorthern California Institute for Research and Education (NCIRE)San FranciscoCaliforniaUSA
- Department of Veterans Affairs Medical CenterCenter for Imaging of Neurodegenerative DiseasesSan FranciscoCaliforniaUSA
| | - Paul S. Aisen
- Alzheimer's Therapeutic Research InstituteUniversity of Southern CaliforniaSan DiegoCaliforniaUSA
| | - Miriam A. Ashford
- Department of Veterans Affairs Medical CenterNorthern California Institute for Research and Education (NCIRE)San FranciscoCaliforniaUSA
| | - Laurel A. Beckett
- Division of BiostatisticsDepartment of Public Health SciencesUniversity of CaliforniaDavisCaliforniaUSA
| | - Robert C. Green
- Division of GeneticsDepartment of MedicineBrigham and Women's HospitalBroad Institute Ariadne Labs and Harvard Medical SchoolBostonMassachusettsUSA
| | - Danielle Harvey
- Division of BiostatisticsDepartment of Public Health SciencesUniversity of CaliforniaDavisCaliforniaUSA
| | | | - William Jagust
- Helen Wills Neuroscience InstituteUniversity of California BerkeleyBerkeleyCaliforniaUSA
| | - Susan M. Landau
- Helen Wills Neuroscience InstituteUniversity of California BerkeleyBerkeleyCaliforniaUSA
| | - John C. Morris
- Knight Alzheimer's Disease Research CenterWashington University School of MedicineSaint LouisMissouriUSA
- Department of NeurologyWashington University School of MedicineSaint LouisMissouriUSA
- Department of Pathology and ImmunologyWashington University School of MedicineSaint LouisMissouriUSA
| | - Kwangsik T. Nho
- Department of Radiology and Imaging Sciences and the Indiana Alzheimer's Disease Research CenterIndiana University School of MedicineIndianapolisIndianaUSA
- Center for Computational Biology and BioinformaticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Rachel Nosheny
- Department of Veterans Affairs Medical CenterCenter for Imaging of Neurodegenerative DiseasesSan FranciscoCaliforniaUSA
- Department of Psychiatry and Behavioral SciencesUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Ozioma Okonkwo
- Wisconsin Alzheimer's Disease Research Center and Department of MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Richard J. Perrin
- Knight Alzheimer's Disease Research CenterWashington University School of MedicineSaint LouisMissouriUSA
- Department of NeurologyWashington University School of MedicineSaint LouisMissouriUSA
- Department of Pathology and ImmunologyWashington University School of MedicineSaint LouisMissouriUSA
| | | | - Monica Rivera Mindt
- Department of PsychologyLatin American and Latino Studies InstituteAfrican and African American StudiesFordham UniversityNew YorkNew YorkUSA
- Department of NeurologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Andrew Saykin
- Department of Radiology and Imaging Sciences and the Indiana Alzheimer's Disease Research CenterIndiana University School of MedicineIndianapolisIndianaUSA
- Department of Medical and Molecular GeneticsIndiana University School of MedicineIndianapolisIndianaUSA
| | - Leslie M. Shaw
- Department of Pathology and Laboratory Medicine and the PENN Alzheimer's Disease Research CenterCenter for Neurodegenerative ResearchPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Arthur W. Toga
- Laboratory of Neuro ImagingInstitute of Neuroimaging and InformaticsKeck School of Medicine of University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Duygu Tosun
- Department of Veterans Affairs Medical CenterCenter for Imaging of Neurodegenerative DiseasesSan FranciscoCaliforniaUSA
- Department of Radiology and Biomedical ImagingUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | | |
Collapse
|
49
|
Ishtiak-Ahmed K, Musliner KL, Christensen KS, Mortensen EL, Nierenberg AA, Gasse C. Real-World Evidence on Clinical Outcomes of Commonly Used Antidepressants in Older Adults Initiating Antidepressants for Depression: A Nationwide Cohort Study in Denmark. Am J Psychiatry 2024; 181:47-56. [PMID: 37849303 DOI: 10.1176/appi.ajp.20230356] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2023]
Abstract
OBJECTIVE The authors investigated the clinical outcomes of commonly used antidepressants among older adults who initiated first-time antidepressants for depression by analyzing the 1-year risk of selected clinically relevant outcomes. METHODS This cohort study used nationwide Danish registry data and included all older adults who redeemed a first-time (since 1995) antidepressant prescription with an indication of depression between 2006 and 2017. Only the 10 most frequently redeemed antidepressants were included in the analyses. Outcomes included discontinuation, switching, augmentation, psychiatric hospital contacts, suicide attempt or self-harm, fall-related injuries, cardiovascular events, and all-cause mortality. Incidence rate ratios (IRRs) and 95% confidence intervals were estimated using Poisson regression models, controlling for potential confounders. RESULTS The study sample included 93,883 older adults (mean age, 78.0 years, SD=7.5 years; 56% female). The most frequently prescribed antidepressants were selective serotonin reuptake inhibitors (citalopram, 47.04%; escitalopram, 11.81%; fluoxetine, 0.55%; paroxetine, 0.52%; sertraline, 11.17%), serotonin-norepinephrine reuptake inhibitors (duloxetine, 0.71%; venlafaxine, 1.54%), a tricyclic antidepressant (amitriptyline, 1.86%), and two atypical antidepressants (mianserin, 1.93%; mirtazapine, 22.87%). Compared with users of sertraline (the reference drug in this analysis, as Danish guidelines recommend it as the first-choice treatment for depression), users of most of the other nine antidepressants had a significantly higher risk of discontinuation (e.g., mirtazapine: IRR=1.55, 95% CI=1.50-1.61; venlafaxine: IRR=1.22, 95% CI=1.12-1.32), switching (amitriptyline: IRR=1.45, 95% CI=1.15-1.81; venlafaxine: IRR=1.47, 95% CI=1.20-1.80), augmentation, cardiovascular events, and mortality. Overall, mirtazapine and venlafaxine users had the most adverse outcomes compared with sertraline users. These results remained consistent in analyses stratified by sex and age (≤75 years vs. >75 years). CONCLUSIONS This real-world evidence suggests that clinical outcomes may vary among initiators of commonly used antidepressants in older adults, which may inform benefit-risk evaluation at treatment initiation, and highlights the importance of careful selection of antidepressant treatment.
Collapse
Affiliation(s)
- Kazi Ishtiak-Ahmed
- Department of Affective Disorders (Ishtiak-Ahmed, Musliner, Nierenberg, Gasse) and Psychosis Research Unit (Gasse), Aarhus University Hospital Psychiatry, Aarhus, Denmark; Department of Clinical Medicine (Ishtiak-Ahmed, Musliner, Gasse), Department of Public Health (Christensen), and Research Unit for General Practice (Christensen), Aarhus University, Aarhus, Denmark; Department of Public Health, University of Copenhagen, Copenhagen (Mortensen); Dauten Family Center for Bipolar Treatment Innovation, Massachusetts General Hospital, Boston, and Harvard Medical School, Boston (Nierenberg)
| | - Katherine L Musliner
- Department of Affective Disorders (Ishtiak-Ahmed, Musliner, Nierenberg, Gasse) and Psychosis Research Unit (Gasse), Aarhus University Hospital Psychiatry, Aarhus, Denmark; Department of Clinical Medicine (Ishtiak-Ahmed, Musliner, Gasse), Department of Public Health (Christensen), and Research Unit for General Practice (Christensen), Aarhus University, Aarhus, Denmark; Department of Public Health, University of Copenhagen, Copenhagen (Mortensen); Dauten Family Center for Bipolar Treatment Innovation, Massachusetts General Hospital, Boston, and Harvard Medical School, Boston (Nierenberg)
| | - Kaj Sparle Christensen
- Department of Affective Disorders (Ishtiak-Ahmed, Musliner, Nierenberg, Gasse) and Psychosis Research Unit (Gasse), Aarhus University Hospital Psychiatry, Aarhus, Denmark; Department of Clinical Medicine (Ishtiak-Ahmed, Musliner, Gasse), Department of Public Health (Christensen), and Research Unit for General Practice (Christensen), Aarhus University, Aarhus, Denmark; Department of Public Health, University of Copenhagen, Copenhagen (Mortensen); Dauten Family Center for Bipolar Treatment Innovation, Massachusetts General Hospital, Boston, and Harvard Medical School, Boston (Nierenberg)
| | - Erik Lykke Mortensen
- Department of Affective Disorders (Ishtiak-Ahmed, Musliner, Nierenberg, Gasse) and Psychosis Research Unit (Gasse), Aarhus University Hospital Psychiatry, Aarhus, Denmark; Department of Clinical Medicine (Ishtiak-Ahmed, Musliner, Gasse), Department of Public Health (Christensen), and Research Unit for General Practice (Christensen), Aarhus University, Aarhus, Denmark; Department of Public Health, University of Copenhagen, Copenhagen (Mortensen); Dauten Family Center for Bipolar Treatment Innovation, Massachusetts General Hospital, Boston, and Harvard Medical School, Boston (Nierenberg)
| | - Andrew A Nierenberg
- Department of Affective Disorders (Ishtiak-Ahmed, Musliner, Nierenberg, Gasse) and Psychosis Research Unit (Gasse), Aarhus University Hospital Psychiatry, Aarhus, Denmark; Department of Clinical Medicine (Ishtiak-Ahmed, Musliner, Gasse), Department of Public Health (Christensen), and Research Unit for General Practice (Christensen), Aarhus University, Aarhus, Denmark; Department of Public Health, University of Copenhagen, Copenhagen (Mortensen); Dauten Family Center for Bipolar Treatment Innovation, Massachusetts General Hospital, Boston, and Harvard Medical School, Boston (Nierenberg)
| | - Christiane Gasse
- Department of Affective Disorders (Ishtiak-Ahmed, Musliner, Nierenberg, Gasse) and Psychosis Research Unit (Gasse), Aarhus University Hospital Psychiatry, Aarhus, Denmark; Department of Clinical Medicine (Ishtiak-Ahmed, Musliner, Gasse), Department of Public Health (Christensen), and Research Unit for General Practice (Christensen), Aarhus University, Aarhus, Denmark; Department of Public Health, University of Copenhagen, Copenhagen (Mortensen); Dauten Family Center for Bipolar Treatment Innovation, Massachusetts General Hospital, Boston, and Harvard Medical School, Boston (Nierenberg)
| |
Collapse
|
50
|
Yang Z, Wen J, Erus G, Govindarajan ST, Melhem R, Mamourian E, Cui Y, Srinivasan D, Abdulkadir A, Parmpi P, Wittfeld K, Grabe HJ, Bülow R, Frenzel S, Tosun D, Bilgel M, An Y, Yi D, Marcus DS, LaMontagne P, Benzinger TL, Heckbert SR, Austin TR, Waldstein SR, Evans MK, Zonderman AB, Launer LJ, Sotiras A, Espeland MA, Masters CL, Maruff P, Fripp J, Toga A, O’Bryant S, Chakravarty MM, Villeneuve S, Johnson SC, Morris JC, Albert MS, Yaffe K, Völzke H, Ferrucci L, Bryan NR, Shinohara RT, Fan Y, Habes M, Lalousis PA, Koutsouleris N, Wolk DA, Resnick SM, Shou H, Nasrallah IM, Davatzikos C. Five dominant dimensions of brain aging are identified via deep learning: associations with clinical, lifestyle, and genetic measures. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.12.29.23300642. [PMID: 38234857 PMCID: PMC10793523 DOI: 10.1101/2023.12.29.23300642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Brain aging is a complex process influenced by various lifestyle, environmental, and genetic factors, as well as by age-related and often co-existing pathologies. MRI and, more recently, AI methods have been instrumental in understanding the neuroanatomical changes that occur during aging in large and diverse populations. However, the multiplicity and mutual overlap of both pathologic processes and affected brain regions make it difficult to precisely characterize the underlying neurodegenerative profile of an individual from an MRI scan. Herein, we leverage a state-of-the art deep representation learning method, Surreal-GAN, and present both methodological advances and extensive experimental results that allow us to elucidate the heterogeneity of brain aging in a large and diverse cohort of 49,482 individuals from 11 studies. Five dominant patterns of neurodegeneration were identified and quantified for each individual by their respective (herein referred to as) R-indices. Significant associations between R-indices and distinct biomedical, lifestyle, and genetic factors provide insights into the etiology of observed variances. Furthermore, baseline R-indices showed predictive value for disease progression and mortality. These five R-indices contribute to MRI-based precision diagnostics, prognostication, and may inform stratification into clinical trials.
Collapse
Affiliation(s)
- Zhijian Yang
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Graduate Group in Applied Mathematics and Computational Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Junhao Wen
- Laboratory of AI and Biomedical Science (LABS), Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, California, USA
| | - Guray Erus
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sindhuja T. Govindarajan
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Randa Melhem
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth Mamourian
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yuhan Cui
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dhivya Srinivasan
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ahmed Abdulkadir
- Laboratory for Research in Neuroimaging, Department of Clinical Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Paraskevi Parmpi
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Katharina Wittfeld
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Germany
| | - Hans J. Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Germany
- German Center for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, Germany
| | - Robin Bülow
- Institute of Diagnostic Radiology and Neuroradiology, University of Greifswald, Germany
| | - Stefan Frenzel
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Germany
| | - Duygu Tosun
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Murat Bilgel
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Yang An
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Dahyun Yi
- Institute of Human Behavioral Medicine, Medical Research Center Seoul National University, Seoul, Republic of Korea
| | - Daniel S. Marcus
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Pamela LaMontagne
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tammie L.S. Benzinger
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Susan R. Heckbert
- Cardiovascular Health Research Unit and Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Thomas R. Austin
- Cardiovascular Health Research Unit and Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Shari R. Waldstein
- Department of Psychology, University of Maryland, Baltimore County, Catonsville, MD, USA
| | - Michele K. Evans
- Health Disparities Research Section, Laboratory of Epidemiology and Population Sciences, NIA/NIH/IRP, Baltimore, MD, USA
| | - Alan B. Zonderman
- Health Disparities Research Section, Laboratory of Epidemiology and Population Sciences, NIA/NIH/IRP, Baltimore, MD, USA
| | - Lenore J. Launer
- Neuroepidemiology Section, Intramural Research Program, National Institute on Aging, Bethesda, Maryland, USA
| | - Aristeidis Sotiras
- Department of Radiology and Institute of Informatics, Washington University in St. Luis, St. Luis, MO63110, USA
| | - Mark A. Espeland
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Colin L. Masters
- Florey Institute, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Paul Maruff
- Florey Institute, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Jurgen Fripp
- CSIRO Health and Biosecurity, Australian e-Health Research Centre CSIRO, Brisbane, Queensland, Australia
| | - Arthur Toga
- Laboratory of Neuro Imaging, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, California, USA
| | - Sid O’Bryant
- Institute for Translational Research University of North Texas Health Science Center Fort Worth Texas USA
| | - Mallar M. Chakravarty
- Computational Brain Anatomy (CoBrA) Laboratory, Cerebral Imaging Center, Douglas Mental Health University Institute, McGill University, Verdun, Quebec, Canada
| | - Sylvia Villeneuve
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Sterling C. Johnson
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - John C. Morris
- Knight Alzheimer Disease Research Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Marilyn S. Albert
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kristine Yaffe
- Departments of Neurology, Psychiatry and Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Luigi Ferrucci
- Translational Gerontology Branch, Longitudinal Studies Section, National Institute on Aging, National Institutes of Health, MedStar Harbor Hospital, 3001 S. Hanover Street, Baltimore, MD, USA
| | - Nick R. Bryan
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Russell T. Shinohara
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, & Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Yong Fan
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mohamad Habes
- Biggs Alzheimer’s Institute, University of Texas San Antonio Health Science Center, USA
| | - Paris Alexandros Lalousis
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
| | - Nikolaos Koutsouleris
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
- Section for Precision Psychiatry, Department of Psychiatry and Psychotherapy, Ludwig-Maximilian-University Munich, Munich, Germany
| | - David A. Wolk
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Susan M. Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Haochang Shou
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Statistics in Imaging and Visualization Center, Department of Biostatistics, Epidemiology, & Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Ilya M. Nasrallah
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Christos Davatzikos
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for and Data Science for Integrated Diagnostics (AID), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|