1
|
Sahoo SS, Turner MD, Wang L, Ambite JL, Appaji A, Rajasekar A, Lander HM, Wang Y, Turner JA. NeuroBridge ontology: computable provenance metadata to give the long tail of neuroimaging data a FAIR chance for secondary use. Front Neuroinform 2023; 17:1216443. [PMID: 37554248 PMCID: PMC10406126 DOI: 10.3389/fninf.2023.1216443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 07/05/2023] [Indexed: 08/10/2023] Open
Abstract
Background Despite the efforts of the neuroscience community, there are many published neuroimaging studies with data that are still not findable or accessible. Users face significant challenges in reusing neuroimaging data due to the lack of provenance metadata, such as experimental protocols, study instruments, and details about the study participants, which is also required for interoperability. To implement the FAIR guidelines for neuroimaging data, we have developed an iterative ontology engineering process and used it to create the NeuroBridge ontology. The NeuroBridge ontology is a computable model of provenance terms to implement FAIR principles and together with an international effort to annotate full text articles with ontology terms, the ontology enables users to locate relevant neuroimaging datasets. Methods Building on our previous work in metadata modeling, and in concert with an initial annotation of a representative corpus, we modeled diagnosis terms (e.g., schizophrenia, alcohol usage disorder), magnetic resonance imaging (MRI) scan types (T1-weighted, task-based, etc.), clinical symptom assessments (PANSS, AUDIT), and a variety of other assessments. We used the feedback of the annotation team to identify missing metadata terms, which were added to the NeuroBridge ontology, and we restructured the ontology to support both the final annotation of the corpus of neuroimaging articles by a second, independent set of annotators, as well as the functionalities of the NeuroBridge search portal for neuroimaging datasets. Results The NeuroBridge ontology consists of 660 classes with 49 properties with 3,200 axioms. The ontology includes mappings to existing ontologies, enabling the NeuroBridge ontology to be interoperable with other domain specific terminological systems. Using the ontology, we annotated 186 neuroimaging full-text articles describing the participant types, scanning, clinical and cognitive assessments. Conclusion The NeuroBridge ontology is the first computable metadata model that represents the types of data available in recent neuroimaging studies in schizophrenia and substance use disorders research; it can be extended to include more granular terms as needed. This metadata ontology is expected to form the computational foundation to help both investigators to make their data FAIR compliant and support users to conduct reproducible neuroimaging research.
Collapse
Affiliation(s)
- Satya S. Sahoo
- Case Western Reserve University, Cleveland, OH, United States
| | - Matthew D. Turner
- Department of Psychiatry and Behavioral Health, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Lei Wang
- Department of Psychiatry and Behavioral Health, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Jose Luis Ambite
- University of Southern California, Los Angeles, CA, United States
| | | | - Arcot Rajasekar
- University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Howard M. Lander
- University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Yue Wang
- University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Jessica A. Turner
- Department of Psychiatry and Behavioral Health, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
2
|
Moon SW, Zhao L, Matloff W, Hobel S, Berger R, Kwon D, Kim J, Toga AW, Dinov ID. Brain structure and allelic associations in Alzheimer's disease. CNS Neurosci Ther 2023; 29:1034-1048. [PMID: 36575854 PMCID: PMC10018103 DOI: 10.1111/cns.14073] [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: 01/08/2022] [Revised: 12/06/2022] [Accepted: 12/11/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD), the most prevalent form of dementia, affects 6.5 million Americans and over 50 million people globally. Clinical, genetic, and phenotypic studies of dementia provide some insights of the observed progressive neurodegenerative processes, however, the mechanisms underlying AD onset remain enigmatic. AIMS This paper examines late-onset dementia-related cognitive impairment utilizing neuroimaging-genetics biomarker associations. MATERIALS AND METHODS The participants, ages 65-85, included 266 healthy controls (HC), 572 volunteers with mild cognitive impairment (MCI), and 188 Alzheimer's disease (AD) patients. Genotype dosage data for AD-associated single nucleotide polymorphisms (SNPs) were extracted from the imputed ADNI genetics archive using sample-major additive coding. Such 29 SNPs were selected, representing a subset of independent SNPs reported to be highly associated with AD in a recent AD meta-GWAS study by Jansen and colleagues. RESULTS We identified the significant correlations between the 29 genomic markers (GMs) and the 200 neuroimaging markers (NIMs). The odds ratios and relative risks for AD and MCI (relative to HC) were predicted using multinomial linear models. DISCUSSION In the HC and MCI cohorts, mainly cortical thickness measures were associated with GMs, whereas the AD cohort exhibited different GM-NIM relations. Network patterns within the HC and AD groups were distinct in cortical thickness, volume, and proportion of White to Gray Matter (pct), but not in the MCI cohort. Multinomial linear models of clinical diagnosis showed precisely the specific NIMs and GMs that were most impactful in discriminating between AD and HC, and between MCI and HC. CONCLUSION This study suggests that advanced analytics provide mechanisms for exploring the interrelations between morphometric indicators and GMs. The findings may facilitate further clinical investigations of phenotypic associations that support deep systematic understanding of AD pathogenesis.
Collapse
Affiliation(s)
- Seok Woo Moon
- Department of Neuropsychiatry, Research Institute of Medical ScienceKonkuk University School of MedicineSeoulKorea
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USCCaliforniaLos AngelesUSA
| | - Lu Zhao
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USCCaliforniaLos AngelesUSA
| | - William Matloff
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USCCaliforniaLos AngelesUSA
| | - Sam Hobel
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USCCaliforniaLos AngelesUSA
| | - Ryan Berger
- Microbiology & ImmunologyUniversity of MichiganAnn ArborMichiganUSA
| | - Daehong Kwon
- Department of Biomedical Science and EngineeringKonkuk UniversitySeoulKorea
| | - Jaebum Kim
- Department of Biomedical Science and EngineeringKonkuk UniversitySeoulKorea
| | - Arthur W. Toga
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USCCaliforniaLos AngelesUSA
| | - Ivo D. Dinov
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USCCaliforniaLos AngelesUSA
- Department of Health Behavior and Biological Sciences, Statistics Online Computational Resource (SOCR), Michigan Institute for Data Science (MIDAS)University of MichiganAnn ArborMichiganUSA
- Department of StatisticsUniversity of CaliforniaLos AngelesCaliforniaUSA
| | | |
Collapse
|
3
|
Miller B, Kim SJ, Mehta HH, Cao K, Kumagai H, Thumaty N, Leelaprachakul N, Braniff RG, Jiao H, Vaughan J, Diedrich J, Saghatelian A, Arpawong TE, Crimmins EM, Ertekin-Taner N, Tubi MA, Hare ET, Braskie MN, Décarie-Spain L, Kanoski SE, Grodstein F, Bennett DA, Zhao L, Toga AW, Wan J, Yen K, Cohen P. Mitochondrial DNA variation in Alzheimer's disease reveals a unique microprotein called SHMOOSE. Mol Psychiatry 2023; 28:1813-1826. [PMID: 36127429 PMCID: PMC10027624 DOI: 10.1038/s41380-022-01769-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 08/15/2022] [Accepted: 08/26/2022] [Indexed: 01/22/2023]
Abstract
Mitochondrial DNA variants have previously associated with disease, but the underlying mechanisms have been largely elusive. Here, we report that mitochondrial SNP rs2853499 associated with Alzheimer's disease (AD), neuroimaging, and transcriptomics. We mapped rs2853499 to a novel mitochondrial small open reading frame called SHMOOSE with microprotein encoding potential. Indeed, we detected two unique SHMOOSE-derived peptide fragments in mitochondria by using mass spectrometry-the first unique mass spectrometry-based detection of a mitochondrial-encoded microprotein to date. Furthermore, cerebrospinal fluid (CSF) SHMOOSE levels in humans correlated with age, CSF tau, and brain white matter volume. We followed up on these genetic and biochemical findings by carrying out a series of functional experiments. SHMOOSE acted on the brain following intracerebroventricular administration, differentiated mitochondrial gene expression in multiple models, localized to mitochondria, bound the inner mitochondrial membrane protein mitofilin, and boosted mitochondrial oxygen consumption. Altogether, SHMOOSE has vast implications for the fields of neurobiology, Alzheimer's disease, and microproteins.
Collapse
Affiliation(s)
- Brendan Miller
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
| | - Su-Jeong Kim
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Hemal H Mehta
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Kevin Cao
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Hiroshi Kumagai
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Neehar Thumaty
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Naphada Leelaprachakul
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Regina Gonzalez Braniff
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Henry Jiao
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Joan Vaughan
- Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Jolene Diedrich
- Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Alan Saghatelian
- Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Thalida E Arpawong
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Eileen M Crimmins
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | | | - Meral A Tubi
- Imaging Genetics Center, Institute for Neuroimaging and Informatics, University of Southern California, Los Angeles, CA, USA
- Department of Neurology, University of Southern California, Los Angeles, CA, USA
| | - Evan T Hare
- Imaging Genetics Center, Institute for Neuroimaging and Informatics, University of Southern California, Los Angeles, CA, USA
- Department of Neurology, University of Southern California, Los Angeles, CA, USA
| | - Meredith N Braskie
- Imaging Genetics Center, Institute for Neuroimaging and Informatics, University of Southern California, Los Angeles, CA, USA
- Department of Neurology, University of Southern California, Los Angeles, CA, USA
| | - Léa Décarie-Spain
- Human and Evolutionary Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA, USA
| | - Scott E Kanoski
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
- Human and Evolutionary Biology Section, Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA, USA
| | - Francine Grodstein
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Lu Zhao
- Laboratory of Neuro Imaging, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of University of Southern California, Los Angeles, CA, USA
| | - Arthur W Toga
- Laboratory of Neuro Imaging, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of University of Southern California, Los Angeles, CA, USA
| | - Junxiang Wan
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Kelvin Yen
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Pinchas Cohen
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
4
|
Moon SW. Neuroimaging Genetics and Network Analysis in Alzheimer's Disease. Curr Alzheimer Res 2023; 20:526-538. [PMID: 37957920 DOI: 10.2174/0115672050265188231107072215] [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: 06/20/2023] [Revised: 07/22/2023] [Accepted: 08/13/2023] [Indexed: 11/15/2023]
Abstract
The issue of the genetics in brain imaging phenotypes serves as a crucial link between two distinct scientific fields: neuroimaging genetics (NG). The articles included here provide solid proof that this NG link has considerable synergy. There is a suitable collection of articles that offer a wide range of viewpoints on how genetic variations affect brain structure and function. They serve as illustrations of several study approaches used in contemporary genetics and neuroscience. Genome-wide association studies and candidate-gene association are two examples of genetic techniques. Cortical gray matter structural/volumetric measures from magnetic resonance imaging (MRI) are sources of information on brain phenotypes. Together, they show how various scientific disciplines have benefited from significant technological advances, such as the single-nucleotide polymorphism array in genetics and the development of increasingly higher-resolution MRI imaging. Moreover, we discuss NG's contribution to expanding our knowledge about the heterogeneity within Alzheimer's disease as well as the benefits of different network analyses.
Collapse
Affiliation(s)
- Seok Woo Moon
- Department of Psychiatry, Institute of Medical Science, Konkuk University School of Medicine, Chungju, Republic of Korea
| |
Collapse
|
5
|
Uselman TW, Medina CS, Gray HB, Jacobs RE, Bearer EL. Longitudinal manganese-enhanced magnetic resonance imaging of neural projections and activity. NMR IN BIOMEDICINE 2022; 35:e4675. [PMID: 35253280 PMCID: PMC11064873 DOI: 10.1002/nbm.4675] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 10/19/2021] [Accepted: 12/07/2021] [Indexed: 06/14/2023]
Abstract
Manganese-enhanced magnetic resonance imaging (MEMRI) holds exceptional promise for preclinical studies of brain-wide physiology in awake-behaving animals. The objectives of this review are to update the current information regarding MEMRI and to inform new investigators as to its potential. Mn(II) is a powerful contrast agent for two main reasons: (1) high signal intensity at low doses; and (2) biological interactions, such as projection tracing and neural activity mapping via entry into electrically active neurons in the living brain. High-spin Mn(II) reduces the relaxation time of water protons: at Mn(II) concentrations typically encountered in MEMRI, robust hyperintensity is obtained without adverse effects. By selectively entering neurons through voltage-gated calcium channels, Mn(II) highlights active neurons. Safe doses may be repeated over weeks to allow for longitudinal imaging of brain-wide dynamics in the same individual across time. When delivered by stereotactic intracerebral injection, Mn(II) enters active neurons at the injection site and then travels inside axons for long distances, tracing neuronal projection anatomy. Rates of axonal transport within the brain were measured for the first time in "time-lapse" MEMRI. When delivered systemically, Mn(II) enters active neurons throughout the brain via voltage-sensitive calcium channels and clears slowly. Thus behavior can be monitored during Mn(II) uptake and hyperintense signals due to Mn(II) uptake captured retrospectively, allowing pairing of behavior with neural activity maps for the first time. Here we review critical information gained from MEMRI projection mapping about human neuropsychological disorders. We then discuss results from neural activity mapping from systemic Mn(II) imaged longitudinally that have illuminated development of the tonotopic map in the inferior colliculus as well as brain-wide responses to acute threat and how it evolves over time. MEMRI posed specific challenges for image data analysis that have recently been transcended. We predict a bright future for longitudinal MEMRI in pursuit of solutions to the brain-behavior mystery.
Collapse
Affiliation(s)
- Taylor W. Uselman
- University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | | | - Harry B. Gray
- Beckman Institute, California Institute of Technology, Pasadena, California, USA
| | - Russell E. Jacobs
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Elaine L. Bearer
- University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
- Beckman Institute, California Institute of Technology, Pasadena, California, USA
| |
Collapse
|
6
|
White T, Blok E, Calhoun VD. Data sharing and privacy issues in neuroimaging research: Opportunities, obstacles, challenges, and monsters under the bed. Hum Brain Mapp 2022; 43:278-291. [PMID: 32621651 PMCID: PMC8675413 DOI: 10.1002/hbm.25120] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/12/2020] [Accepted: 06/22/2020] [Indexed: 12/19/2022] Open
Abstract
Collaborative networks and data sharing initiatives are broadening the opportunities for the advancement of science. These initiatives offer greater transparency in science, with the opportunity for external research groups to reproduce, replicate, and extend research findings. Further, larger datasets offer the opportunity to identify homogeneous patterns within subgroups of individuals, where these patterns may be obscured by the heterogeneity of the neurobiological measure in smaller samples. However, data sharing and data pooling initiatives are not without their challenges, especially with new laws that may at first glance appear quite restrictive for open science initiatives. Interestingly, what is key to some of these new laws (i.e, the European Union's general data protection regulation) is that they provide greater control of data to those who "give" their data for research purposes. Thus, the most important element in data sharing is allowing the participants to make informed decisions about how they want their data to be used, and, within the law of the specific country, to follow the participants' wishes. This framework encompasses obtaining thorough informed consent and allowing the participant to determine the extent that they want their data shared, many of the ethical and legal obstacles are reduced to just monsters under the bed. In this manuscript we discuss the many options and obstacles for data sharing, from fully open, to federated learning, to fully closed. Importantly, we highlight the intersection of data sharing, privacy, and data ownership and highlight specific examples that we believe are informative to the neuroimaging community.
Collapse
Affiliation(s)
- Tonya White
- Department of Child and Adolescent Psychiatry/PsychologyErasmus University Medical CenterRotterdamThe Netherlands
- Department of RadiologyErasmus University Medical CenterRotterdamThe Netherlands
| | - Elisabet Blok
- Department of Child and Adolescent Psychiatry/PsychologyErasmus University Medical CenterRotterdamThe Netherlands
| | - Vince D. Calhoun
- Tri‐institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS)Georgia State University, Georgia Institute of Technology, Emory UniversityAtlantaGeorgiaUSA
| |
Collapse
|
7
|
Turner JA, Calhoun VD, Thompson PM, Jahanshad N, Ching CRK, Thomopoulos SI, Verner E, Strauss GP, Ahmed AO, Turner MD, Basodi S, Ford JM, Mathalon DH, Preda A, Belger A, Mueller BA, Lim KO, van Erp TGM. ENIGMA + COINSTAC: Improving Findability, Accessibility, Interoperability, and Re-usability. Neuroinformatics 2022; 20:261-275. [PMID: 34846691 PMCID: PMC9149142 DOI: 10.1007/s12021-021-09559-y] [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] [Subscribe] [Scholar Register] [Accepted: 11/19/2021] [Indexed: 01/07/2023]
Abstract
The FAIR principles, as applied to clinical and neuroimaging data, reflect the goal of making research products Findable, Accessible, Interoperable, and Reusable. The use of the Collaborative Informatics and Neuroimaging Suite Toolkit for Anonymized Computation (COINSTAC) platform in the Enhancing Neuroimaging Genetics through Meta-Analysis (ENIGMA) consortium combines the technological approach of decentralized analyses with the sociological approach of sharing data. In addition, ENIGMA + COINSTAC provides a platform to facilitate the use of machine-actionable data objects. We first present how ENIGMA and COINSTAC support the FAIR principles, and then showcase their integration with a decentralized meta-analysis of sex differences in negative symptom severity in schizophrenia, and finally present ongoing activities and plans to advance FAIR principles in ENIGMA + COINSTAC. ENIGMA and COINSTAC currently represent efforts toward improved Access, Interoperability, and Reusability. We highlight additional improvements needed in these areas, as well as future connections to other resources for expanded Findability.
Collapse
Affiliation(s)
- Jessica A Turner
- Psychology Department, Georgia State University, Atlanta, GA, USA.
| | - Vince D Calhoun
- Psychology Department, Georgia State University, Atlanta, GA, USA
- Tri-Institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia State University, Georgia Institute of Technology, Emory University, Atlanta, GA, 30303, USA
| | - Paul M Thompson
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Neda Jahanshad
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Christopher R K Ching
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Sophia I Thomopoulos
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, USA
| | - Eric Verner
- Tri-Institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia State University, Georgia Institute of Technology, Emory University, Atlanta, GA, 30303, USA
| | - Gregory P Strauss
- Departments of Psychology and Neuroscience, University of Georgia, Athens, GA, USA
| | - Anthony O Ahmed
- Weill Cornell Medicine, Department of Psychiatry, White Plains, NY, 10605, USA
| | - Matthew D Turner
- Psychology Department, Georgia State University, Atlanta, GA, USA
| | - Sunitha Basodi
- Tri-Institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia State University, Georgia Institute of Technology, Emory University, Atlanta, GA, 30303, USA
| | - Judith M Ford
- Veterans Affairs San Francisco Healthcare System, San Francisco, CA, 94121, USA
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, 94121, USA
| | - Daniel H Mathalon
- Veterans Affairs San Francisco Healthcare System, San Francisco, CA, 94121, USA
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, 94121, USA
| | - Adrian Preda
- Department of Psychiatry and Human Behavior, University of California Irvine, University of California Irvine Medical Center, 101 The City Drive S, Orange, CA, 92868, USA
| | - Aysenil Belger
- Department of Psychiatry and Frank Porter Graham Child Development Institute, University of North Carolina at Chapel Hill, 105 Smith Level Road, Chapel Hill, NC, 27599-8180, USA
| | - Bryon A Mueller
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, 55414, USA
| | - Kelvin O Lim
- Department of Psychiatry and Behavioral Sciences, University of Minnesota, Minneapolis, MN, 55414, USA
| | - Theo G M van Erp
- Clinical Translational Neuroscience Laboratory, Department of Psychiatry and Human Behavior, University of California Irvine, 5251 California Ave, Irvine, CA, 92617, USA
- Center for the Neurobiology of Learning and Memory, University of California Irvine, 309 Qureshey Research Lab, Irvine, CA, 92697, USA
| |
Collapse
|
8
|
Zhao L, Batta I, Matloff W, O'Driscoll C, Hobel S, Toga AW. Neuroimaging PheWAS (Phenome-Wide Association Study): A Free Cloud-Computing Platform for Big-Data, Brain-Wide Imaging Association Studies. Neuroinformatics 2021; 19:285-303. [PMID: 32822005 DOI: 10.1007/s12021-020-09486-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Large-scale, case-control genome-wide association studies (GWASs) have revealed genetic variations associated with diverse neurological and psychiatric disorders. Recent advances in neuroimaging and genomic databases of large healthy and diseased cohorts have empowered studies to characterize effects of the discovered genetic factors on brain structure and function, implicating neural pathways and genetic mechanisms in the underlying biology. However, the unprecedented scale and complexity of the imaging and genomic data requires new advanced biomedical data science tools to manage, process and analyze the data. In this work, we introduce Neuroimaging PheWAS (phenome-wide association study): a web-based system for searching over a wide variety of brain-wide imaging phenotypes to discover true system-level gene-brain relationships using a unified genotype-to-phenotype strategy. This design features a user-friendly graphical user interface (GUI) for anonymous data uploading, study definition and management, and interactive result visualizations as well as a cloud-based computational infrastructure and multiple state-of-art methods for statistical association analysis and multiple comparison correction. We demonstrated the potential of Neuroimaging PheWAS with a case study analyzing the influences of the apolipoprotein E (APOE) gene on various brain morphological properties across the brain in the Alzheimer's Disease Neuroimaging Initiative (ADNI) cohort. Benchmark tests were performed to evaluate the system's performance using data from UK Biobank. The Neuroimaging PheWAS system is freely available. It simplifies the execution of PheWAS on neuroimaging data and provides an opportunity for imaging genetics studies to elucidate routes at play for specific genetic variants on diseases in the context of detailed imaging phenotypic data.
Collapse
Affiliation(s)
- Lu Zhao
- Laboratory of Neuro Imaging, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA
| | - Ishaan Batta
- Laboratory of Neuro Imaging, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA
| | - William Matloff
- Laboratory of Neuro Imaging, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA
| | - Caroline O'Driscoll
- Laboratory of Neuro Imaging, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA
| | - Samuel Hobel
- Laboratory of Neuro Imaging, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA
| | - Arthur W Toga
- Laboratory of Neuro Imaging, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
9
|
Tataranno ML, Vijlbrief DC, Dudink J, Benders MJNL. Precision Medicine in Neonates: A Tailored Approach to Neonatal Brain Injury. Front Pediatr 2021; 9:634092. [PMID: 34095022 PMCID: PMC8171663 DOI: 10.3389/fped.2021.634092] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 04/14/2021] [Indexed: 11/27/2022] Open
Abstract
Despite advances in neonatal care to prevent neonatal brain injury and neurodevelopmental impairment, predicting long-term outcome in neonates at risk for brain injury remains difficult. Early prognosis is currently based on cranial ultrasound (CUS), MRI, EEG, NIRS, and/or general movements assessed at specific ages, and predicting outcome in an individual (precision medicine) is not yet possible. New algorithms based on large databases and machine learning applied to clinical, neuromonitoring, and neuroimaging data and genetic analysis and assays measuring multiple biomarkers (omics) can fulfill the needs of modern neonatology. A synergy of all these techniques and the use of automatic quantitative analysis might give clinicians the possibility to provide patient-targeted decision-making for individualized diagnosis, therapy, and outcome prediction. This review will first focus on common neonatal neurological diseases, associated risk factors, and most common treatments. After that, we will discuss how precision medicine and machine learning (ML) approaches could change the future of prediction and prognosis in this field.
Collapse
Affiliation(s)
| | | | | | - Manon J. N. L. Benders
- Department of Neonatology, Wilhelmina Children's Hospital/University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
10
|
Lan H, Toga AW, Sepehrband F. Three-dimensional self-attention conditional GAN with spectral normalization for multimodal neuroimaging synthesis. Magn Reson Med 2021; 86:1718-1733. [PMID: 33961321 DOI: 10.1002/mrm.28819] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 04/05/2021] [Accepted: 04/07/2021] [Indexed: 01/03/2023]
Abstract
PURPOSE To develop a new 3D generative adversarial network that is designed and optimized for the application of multimodal 3D neuroimaging synthesis. METHODS We present a 3D conditional generative adversarial network (GAN) that uses spectral normalization and feature matching to stabilize the training process and ensure optimization convergence (called SC-GAN). A self-attention module was also added to model the relationships between widely separated image voxels. The performance of the network was evaluated on the data set from ADNI-3, in which the proposed network was used to predict PET images, fractional anisotropy, and mean diffusivity maps from multimodal MRI. Then, SC-GAN was applied on a multidimensional diffusion MRI experiment for superresolution application. Experiment results were evaluated by normalized RMS error, peak SNR, and structural similarity. RESULTS In general, SC-GAN outperformed other state-of-the-art GAN networks including 3D conditional GAN in all three tasks across all evaluation metrics. Prediction error of the SC-GAN was 18%, 24% and 29% lower compared to 2D conditional GAN for fractional anisotropy, PET and mean diffusivity tasks, respectively. The ablation experiment showed that the major contributors to the improved performance of SC-GAN are the adversarial learning and the self-attention module, followed by the spectral normalization module. In the superresolution multidimensional diffusion experiment, SC-GAN provided superior predication in comparison to 3D Unet and 3D conditional GAN. CONCLUSION In this work, an efficient end-to-end framework for multimodal 3D medical image synthesis (SC-GAN) is presented. The source code is also made available at https://github.com/Haoyulance/SC-GAN.
Collapse
Affiliation(s)
- Haoyu Lan
- Laboratory of NeuroImaging, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | | | - Arthur W Toga
- Laboratory of NeuroImaging, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Farshid Sepehrband
- Laboratory of NeuroImaging, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
11
|
Donahue EK, Murdos A, Jakowec MW, Sheikh-Bahaei N, Toga AW, Petzinger GM, Sepehrband F. Global and Regional Changes in Perivascular Space in Idiopathic and Familial Parkinson's Disease. Mov Disord 2021; 36:1126-1136. [PMID: 33470460 DOI: 10.1002/mds.28473] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 11/23/2020] [Accepted: 12/14/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The glymphatic system, including the perivascular space (PVS), plays a critical role in brain homeostasis. Although mounting evidence from Alzheimer's disease has supported the potential role of PVS in neurodegenerative disorders, its contribution in Parkinson's disease (PD) has not been fully elucidated. Although idiopathic (IPD) and familial PD (FPD) share similar pathophysiology in terms of protein aggregation, the differential impact of PVS on PD subtypes remains unknown. Our objective was to examine the differences in PVS volume fraction in IPD and FPD compared to healthy controls (HCs) and nonmanifest carriers (NMCs). METHODS A total of 470 individuals were analyzed from the Parkinson's Progression Markers Initiative database, including (1) IPD (n = 179), (2) FPD (LRRK2 [leucine-rich repeat kinase 2], glucocerebrosidase, or α-synuclein) (n = 67), (3) NMC (n = 101), and (4) HCs (n = 84). Total PVS volume fraction (%) was compared using parcellation and quantitation within greater white matter volume at global and regional levels in all cortical and subcortical white matter. RESULTS There was a significant increase in global and regional PVS volume fraction in PD versus non-PD, particularly in FPD versus NMC and LRRK2 FPD versus NMC. Regionally, FPD and NMC differed in the medial orbitofrontal region, as did LRRK2 FPD versus NMC. Non-PD and PD differed in the medial orbitofrontal region and the banks of the superior temporal regions. IPD and FPD differed in the cuneus and lateral occipital regions. CONCLUSIONS Our findings support the role of PVS in PD and highlight a potentially significant contribution of PVS to the pathophysiology of FPD, particularly LRRK2. © 2021 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Erin K Donahue
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Neuroscience Graduate Program, University of Southern California, Los Angeles, California, USA
| | - Amjad Murdos
- Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Michael W Jakowec
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Neuroscience Graduate Program, University of Southern California, Los Angeles, California, USA
| | - Nasim Sheikh-Bahaei
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Arthur W Toga
- Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Giselle M Petzinger
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Neuroscience Graduate Program, University of Southern California, Los Angeles, California, USA
| | - Farshid Sepehrband
- Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.,Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
12
|
Volumetric distribution of perivascular space in relation to mild cognitive impairment. Neurobiol Aging 2020; 99:28-43. [PMID: 33422892 DOI: 10.1016/j.neurobiolaging.2020.12.010] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 11/25/2020] [Accepted: 12/08/2020] [Indexed: 12/19/2022]
Abstract
Vascular contributions to early cognitive decline are increasingly recognized, prompting further investigation into the nature of related changes in perivascular spaces (PVS). Using magnetic resonance imaging, we show that, compared to a cognitively normal sample, individuals with early cognitive dysfunction have altered PVS presence and distribution, irrespective of Amyloid-β. Surprisingly, we noted lower PVS presence in the anterosuperior medial temporal lobe (asMTL) (1.29 times lower PVS volume fraction in cognitively impaired individuals, p < 0.0001), which was associated with entorhinal neurofibrillary tau tangle deposition (beta (standard error) = -0.98 (0.4); p = 0.014), one of the hallmarks of early Alzheimer's disease pathology. We also observed higher PVS volume fraction in centrum semi-ovale of the white matter, but only in female participants (1.47 times higher PVS volume fraction in cognitively impaired individuals, p = 0.0011). We also observed PVS changes in participants with history of hypertension (higher in the white matter and lower in the asMTL). Our results suggest that anatomically specific alteration of the PVS is an early neuroimaging feature of cognitive impairment in aging adults, which is differentially manifested in female.
Collapse
|
13
|
Montagne A, Nation DA, Sagare AP, Barisano G, Sweeney MD, Chakhoyan A, Pachicano M, Joe E, Nelson AR, D'Orazio LM, Buennagel DP, Harrington MG, Benzinger TLS, Fagan AM, Ringman JM, Schneider LS, Morris JC, Reiman EM, Caselli RJ, Chui HC, Tcw J, Chen Y, Pa J, Conti PS, Law M, Toga AW, Zlokovic BV. APOE4 leads to blood-brain barrier dysfunction predicting cognitive decline. Nature 2020; 581:71-76. [PMID: 32376954 PMCID: PMC7250000 DOI: 10.1038/s41586-020-2247-3] [Citation(s) in RCA: 722] [Impact Index Per Article: 144.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 04/01/2020] [Indexed: 01/13/2023]
Abstract
Vascular contributions to dementia and Alzheimer's disease are increasingly recognized1-6. Recent studies have suggested that breakdown of the blood-brain barrier (BBB) is an early biomarker of human cognitive dysfunction7, including the early clinical stages of Alzheimer's disease5,8-10. The E4 variant of apolipoprotein E (APOE4), the main susceptibility gene for Alzheimer's disease11-14, leads to accelerated breakdown of the BBB and degeneration of brain capillary pericytes15-19, which maintain BBB integrity20-22. It is unclear, however, whether the cerebrovascular effects of APOE4 contribute to cognitive impairment. Here we show that individuals bearing APOE4 (with the ε3/ε4 or ε4/ε4 alleles) are distinguished from those without APOE4 (ε3/ε3) by breakdown of the BBB in the hippocampus and medial temporal lobe. This finding is apparent in cognitively unimpaired APOE4 carriers and more severe in those with cognitive impairment, but is not related to amyloid-β or tau pathology measured in cerebrospinal fluid or by positron emission tomography23. High baseline levels of the BBB pericyte injury biomarker soluble PDGFRβ7,8 in the cerebrospinal fluid predicted future cognitive decline in APOE4 carriers but not in non-carriers, even after controlling for amyloid-β and tau status, and were correlated with increased activity of the BBB-degrading cyclophilin A-matrix metalloproteinase-9 pathway19 in cerebrospinal fluid. Our findings suggest that breakdown of the BBB contributes to APOE4-associated cognitive decline independently of Alzheimer's disease pathology, and might be a therapeutic target in APOE4 carriers.
Collapse
Affiliation(s)
- Axel Montagne
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Daniel A Nation
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Psychological Science, University of California, Irvine, Irvine, CA, USA
- Institute for Memory Disorders and Neurological Impairments, University of California, Irvine, Irvine, CA, USA
| | - Abhay P Sagare
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Giuseppe Barisano
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Melanie D Sweeney
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Ararat Chakhoyan
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Maricarmen Pachicano
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Elizabeth Joe
- Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Amy R Nelson
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Lina M D'Orazio
- Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | | | | | - Tammie L S Benzinger
- Department of Radiology, Washington University School of Medicine, St Louis, MO, USA
- The Hope Center for Neurodegenerative Disorders, Washington University School of Medicine, St Louis, MO, USA
| | - Anne M Fagan
- The Hope Center for Neurodegenerative Disorders, Washington University School of Medicine, St Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
- The Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St Louis, MO, USA
| | - John M Ringman
- Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Lon S Schneider
- Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of Southern California, Los Angeles, CA, USA
| | - John C Morris
- Department of Neurology, Washington University School of Medicine, St Louis, MO, USA
- The Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St Louis, MO, USA
| | | | | | - Helena C Chui
- Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Julia Tcw
- Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yining Chen
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Judy Pa
- Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Laboratory of Neuroimaging, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Peter S Conti
- Molecular Imaging Center, Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Meng Law
- Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Neuroscience and Radiology, Monash University, Alfred Health, Melbourne, Victoria, Australia
| | - Arthur W Toga
- Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Laboratory of Neuroimaging, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Berislav V Zlokovic
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
- Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
14
|
Anderson RJ, Cook JJ, Delpratt N, Nouls JC, Gu B, McNamara JO, Avants BB, Johnson GA, Badea A. Small Animal Multivariate Brain Analysis (SAMBA) - a High Throughput Pipeline with a Validation Framework. Neuroinformatics 2020; 17:451-472. [PMID: 30565026 PMCID: PMC6584586 DOI: 10.1007/s12021-018-9410-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
While many neuroscience questions aim to understand the human brain, much current knowledge has been gained using animal models, which replicate genetic, structural, and connectivity aspects of the human brain. While voxel-based analysis (VBA) of preclinical magnetic resonance images is widely-used, a thorough examination of the statistical robustness, stability, and error rates is hindered by high computational demands of processing large arrays, and the many parameters involved therein. Thus, workflows are often based on intuition or experience, while preclinical validation studies remain scarce. To increase throughput and reproducibility of quantitative small animal brain studies, we have developed a publicly shared, high throughput VBA pipeline in a high-performance computing environment, called SAMBA. The increased computational efficiency allowed large multidimensional arrays to be processed in 1–3 days—a task that previously took ~1 month. To quantify the variability and reliability of preclinical VBA in rodent models, we propose a validation framework consisting of morphological phantoms, and four metrics. This addresses several sources that impact VBA results, including registration and template construction strategies. We have used this framework to inform the VBA workflow parameters in a VBA study for a mouse model of epilepsy. We also present initial efforts towards standardizing small animal neuroimaging data in a similar fashion with human neuroimaging. We conclude that verifying the accuracy of VBA merits attention, and should be the focus of a broader effort within the community. The proposed framework promotes consistent quality assurance of VBA in preclinical neuroimaging, thus facilitating the creation and communication of robust results.
Collapse
Affiliation(s)
- Robert J Anderson
- Center for In Vivo Microscopy, Department of Radiology, Duke University Medical Center, Durham, NC, 27710, USA
| | - James J Cook
- Center for In Vivo Microscopy, Department of Radiology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Natalie Delpratt
- Center for In Vivo Microscopy, Department of Radiology, Duke University Medical Center, Durham, NC, 27710, USA.,Department of Biomedical Engineering, Duke University Medical Center, 3302, Durham, NC, 27710, USA
| | - John C Nouls
- Center for In Vivo Microscopy, Department of Radiology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Bin Gu
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, 27710, USA.,Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - James O McNamara
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, 27710, USA.,Department of Neurobiology, Duke University Medical Center, Durham, NC, 27710, USA.,Department of Neurology, Duke University Medical Center, Durham, NC, 27710, USA
| | | | - G Allan Johnson
- Center for In Vivo Microscopy, Department of Radiology, Duke University Medical Center, Durham, NC, 27710, USA.,Department of Biomedical Engineering, Duke University Medical Center, 3302, Durham, NC, 27710, USA
| | - Alexandra Badea
- Center for In Vivo Microscopy, Department of Radiology, Duke University Medical Center, Durham, NC, 27710, USA. .,Department of Biomedical Engineering, Duke University Medical Center, 3302, Durham, NC, 27710, USA.
| |
Collapse
|
15
|
Dinov ID. Modernizing the Methods and Analytics Curricula for Health Science Doctoral Programs. Front Public Health 2020; 8:22. [PMID: 32117857 PMCID: PMC7031195 DOI: 10.3389/fpubh.2020.00022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 01/23/2020] [Indexed: 12/24/2022] Open
Abstract
This perspective provides a rationale for redesigning and a framework for expanding the graduate health science analytics and biomedical doctoral program curricula. It responds to digital revolution pressures, ubiquitous proliferation of big biomedical data, substantial recent advances in scientific technologies, and rapid progress in health analytics. Specifically, the paper presents a set of common prerequisites, a proposal for core computational and data analytic curriculum, and a list of expected outcome competencies for graduates of doctoral health science and biomedical programs. The manuscript emphasizes the necessity for coordinated efforts of all stakeholders, including trainees, educators, academic institutions, funding agencies, and policy makers. Concrete recommendations are presented of how to ensure graduates with terminal health science analytics and biomedical degrees are trained and able to continuously self-learn, effectively communicate across disciplines, and promote adaptation and change to counteract the relentless pace of automation and the law of diminishing returns.
Collapse
Affiliation(s)
- Ivo D. Dinov
- Statistics Online Computational Resource, Department of Health Behavior and Biological Sciences, University of Michigan, Ann Arbor, MI, United States
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, United States
- Michigan Institute for Data Science, University of Michigan, Ann Arbor, MI, United States
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
16
|
Sta Cruz S, Dinov ID, Herting MM, González-Zacarías C, Kim H, Toga AW, Sepehrband F. Imputation Strategy for Reliable Regional MRI Morphological Measurements. Neuroinformatics 2020; 18:59-70. [PMID: 31054076 PMCID: PMC6829024 DOI: 10.1007/s12021-019-09426-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Regional morphological analysis represents a crucial step in most neuroimaging studies. Results from brain segmentation techniques are intrinsically prone to certain degrees of variability, mainly as results of suboptimal segmentation. To reduce this inherent variability, the errors are often identified through visual inspection and then corrected (semi)manually. Identification and correction of incorrect segmentation could be very expensive for large-scale studies. While identification of the incorrect results can be done relatively fast even with manual inspection, the correction step is extremely time-consuming, as it requires training staff to perform laborious manual corrections. Here we frame the correction phase of this problem as a missing data problem. Instead of manually adjusting the segmentation outputs, our computational approach aims to derive accurate morphological measures by machine learning imputation. Data imputation techniques may be used to replace missing or incorrect region average values with carefully chosen imputed values, all of which are computed based on other available multivariate information. We examined our approach of correcting segmentation outputs on a cohort of 970 subjects, which were undergone an extensive, time-consuming, manual post-segmentation correction. A random forest imputation technique recovered the gold standard results with a significant accuracy (r = 0.93, p < 0.0001; when 30% of the segmentations were considered incorrect in a non-random fashion). The random forest technique proved to be most effective for big data studies (N > 250).
Collapse
Affiliation(s)
- Shaina Sta Cruz
- Department of Communication Sciences and Disorders, California State University, Fullerton, CA, USA
- Public Health Graduate Program, University of California Merced, Merced, CA, USA
| | - Ivo D Dinov
- Laboratory of Neuro Imaging, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
- Statistics Online Computational Resource, Department of Health Behavior and Biological, Michigan Institute for Data Science, University of Michigan, Ann Arbor, MI, USA
| | - Megan M Herting
- Department of Preventive Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
- Department of Pediatrics, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Clio González-Zacarías
- Laboratory of Neuro Imaging, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
| | - Hosung Kim
- Laboratory of Neuro Imaging, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Arthur W Toga
- Laboratory of Neuro Imaging, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Farshid Sepehrband
- Laboratory of Neuro Imaging, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
17
|
Makkie M, Li X, Quinn S, Lin B, Ye J, Mon G, Liu T. A Distributed Computing Platform for fMRI Big Data Analytics. IEEE TRANSACTIONS ON BIG DATA 2019; 5:109-119. [PMID: 31240237 PMCID: PMC6592627 DOI: 10.1109/tbdata.2018.2811508] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Since the BRAIN Initiative and Human Brain Project began, a few efforts have been made to address the computational challenges of neuroscience Big Data. The promises of these two projects were to model the complex interaction of brain and behavior and to understand and diagnose brain diseases by collecting and analyzing large quanitites of data. Archiving, analyzing, and sharing the growing neuroimaging datasets posed major challenges. New computational methods and technologies have emerged in the domain of Big Data but have not been fully adapted for use in neuroimaging. In this work, we introduce the current challenges of neuroimaging in a big data context. We review our efforts toward creating a data management system to organize the large-scale fMRI datasets, and present our novel algorithms/methods for the distributed fMRI data processing that employs Hadoop and Spark. Finally, we demonstrate the significant performance gains of our algorithms/methods to perform distributed dictionary learning.
Collapse
Affiliation(s)
- Milad Makkie
- Department of Computer Science, University of Georgia, Athens, GA 30602
| | - Xiang Li
- Clincial Data Science Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114
| | - Shannon Quinn
- Department of Computer Science, University of Georgia, Athens, GA 30602
| | - Binbin Lin
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Ml 48109
| | - Jieping Ye
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Ml 48109
| | - Geoffrey Mon
- Department of Computer Science, University of Georgia, Athens, GA 30602
| | - Tianming Liu
- Department of Computer Science, University of Georgia, Athens, GA 30602
| |
Collapse
|
18
|
Zhou Y, Zhao L, Zhou N, Zhao Y, Marino S, Wang T, Sun H, Toga AW, Dinov ID. Predictive Big Data Analytics using the UK Biobank Data. Sci Rep 2019; 9:6012. [PMID: 30979917 PMCID: PMC6461626 DOI: 10.1038/s41598-019-41634-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 03/13/2019] [Indexed: 12/04/2022] Open
Abstract
The UK Biobank is a rich national health resource that provides enormous opportunities for international researchers to examine, model, and analyze census-like multisource healthcare data. The archive presents several challenges related to aggregation and harmonization of complex data elements, feature heterogeneity and salience, and health analytics. Using 7,614 imaging, clinical, and phenotypic features of 9,914 subjects we performed deep computed phenotyping using unsupervised clustering and derived two distinct sub-cohorts. Using parametric and nonparametric tests, we determined the top 20 most salient features contributing to the cluster separation. Our approach generated decision rules to predict the presence and progression of depression or other mental illnesses by jointly representing and modeling the significant clinical and demographic variables along with the derived salient neuroimaging features. We reported consistency and reliability measures of the derived computed phenotypes and the top salient imaging biomarkers that contributed to the unsupervised clustering. This clinical decision support system identified and utilized holistically the most critical biomarkers for predicting mental health, e.g., depression. External validation of this technique on different populations may lead to reducing healthcare expenses and improving the processes of diagnosis, forecasting, and tracking of normal and pathological aging.
Collapse
Affiliation(s)
- Yiwang Zhou
- Statistics Online Computational Resource (SOCR), Department of Health Behavior and Biological Sciences, University of Michigan, Ann Arbor, MI, USA.,Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Lu Zhao
- Laboratory of Neuro Imaging, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Nina Zhou
- Statistics Online Computational Resource (SOCR), Department of Health Behavior and Biological Sciences, University of Michigan, Ann Arbor, MI, USA.,Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Yi Zhao
- Statistics Online Computational Resource (SOCR), Department of Health Behavior and Biological Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Simeone Marino
- Statistics Online Computational Resource (SOCR), Department of Health Behavior and Biological Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Tuo Wang
- Statistics Online Computational Resource (SOCR), Department of Health Behavior and Biological Sciences, University of Michigan, Ann Arbor, MI, USA.,Department of Statistics, University of Michigan, Ann Arbor, MI, USA
| | - Hanbo Sun
- Statistics Online Computational Resource (SOCR), Department of Health Behavior and Biological Sciences, University of Michigan, Ann Arbor, MI, USA.,Department of Statistics, University of Michigan, Ann Arbor, MI, USA
| | - 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
| | - Ivo D Dinov
- Statistics Online Computational Resource (SOCR), Department of Health Behavior and Biological Sciences, University of Michigan, Ann Arbor, MI, USA. .,Laboratory of Neuro Imaging, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA. .,Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA. .,Michigan Institute for Data Science, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
19
|
Kiar G, Brown ST, Glatard T, Evans AC. A Serverless Tool for Platform Agnostic Computational Experiment Management. Front Neuroinform 2019; 13:12. [PMID: 30890927 PMCID: PMC6411646 DOI: 10.3389/fninf.2019.00012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 02/15/2019] [Indexed: 01/22/2023] Open
Abstract
Neuroscience has been carried into the domain of big data and high performance computing (HPC) on the backs of initiatives in data collection and an increasingly compute-intensive tools. While managing HPC experiments requires considerable technical acumen, platforms, and standards have been developed to ease this burden on scientists. While web-portals make resources widely accessible, data organizations such as the Brain Imaging Data Structure and tool description languages such as Boutiques provide researchers with a foothold to tackle these problems using their own datasets, pipelines, and environments. While these standards lower the barrier to adoption of HPC and cloud systems for neuroscience applications, they still require the consolidation of disparate domain-specific knowledge. We present Clowdr, a lightweight tool to launch experiments on HPC systems and clouds, record rich execution records, and enable the accessible sharing and re-launch of experimental summaries and results. Clowdr uniquely sits between web platforms and bare-metal applications for experiment management by preserving the flexibility of do-it-yourself solutions while providing a low barrier for developing, deploying and disseminating neuroscientific analysis.
Collapse
Affiliation(s)
- Gregory Kiar
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada
- Department of Biomedical Engineering, McGill University, Montreal, QC, Canada
| | - Shawn T. Brown
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Tristan Glatard
- Department of Computer Science, Concordia University, Montreal, QC, Canada
| | - Alan C. Evans
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada
- Department of Biomedical Engineering, McGill University, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| |
Collapse
|
20
|
Duncan D, Vespa P, Pitkänen A, Braimah A, Lapinlampi N, Toga AW. Big data sharing and analysis to advance research in post-traumatic epilepsy. Neurobiol Dis 2019; 123:127-136. [PMID: 29864492 PMCID: PMC6274619 DOI: 10.1016/j.nbd.2018.05.026] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 05/24/2018] [Accepted: 05/31/2018] [Indexed: 11/26/2022] Open
Abstract
We describe the infrastructure and functionality for a centralized preclinical and clinical data repository and analytic platform to support importing heterogeneous multi-modal data, automatically and manually linking data across modalities and sites, and searching content. We have developed and applied innovative image and electrophysiology processing methods to identify candidate biomarkers from MRI, EEG, and multi-modal data. Based on heterogeneous biomarkers, we present novel analytic tools designed to study epileptogenesis in animal model and human with the goal of tracking the probability of developing epilepsy over time.
Collapse
Affiliation(s)
- Dominique Duncan
- Laboratory of Neuro Imaging, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA.
| | - Paul Vespa
- Division of Neurosurgery and Department of Neurology, University of California at Los Angeles School of Medicine, Los Angeles, CA, USA
| | - Asla Pitkänen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Adebayo Braimah
- Laboratory of Neuro Imaging, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Niina Lapinlampi
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - 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
| |
Collapse
|
21
|
Nation DA, Sweeney MD, Montagne A, Sagare AP, D'Orazio LM, Pachicano M, Sepehrband F, Nelson AR, Buennagel DP, Harrington MG, Benzinger TLS, Fagan AM, Ringman JM, Schneider LS, Morris JC, Chui HC, Law M, Toga AW, Zlokovic BV. Blood-brain barrier breakdown is an early biomarker of human cognitive dysfunction. Nat Med 2019; 25:270-276. [PMID: 30643288 PMCID: PMC6367058 DOI: 10.1038/s41591-018-0297-y] [Citation(s) in RCA: 1000] [Impact Index Per Article: 166.7] [Reference Citation Analysis] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 11/07/2018] [Indexed: 11/09/2022]
Affiliation(s)
- Daniel A Nation
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.,Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.,Department of Psychology, University of Southern California, Los Angeles, CA, USA
| | - Melanie D Sweeney
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Axel Montagne
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Abhay P Sagare
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Lina M D'Orazio
- Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.,Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Maricarmen Pachicano
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Farshid Sepehrband
- Laboratory of Neuro Imaging, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Amy R Nelson
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | | | | | - Tammie L S Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA.,The Hope Center for Neurodegenerative Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Anne M Fagan
- The Hope Center for Neurodegenerative Disorders, Washington University School of Medicine, St. Louis, MO, USA.,Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.,The Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - John M Ringman
- Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.,Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Lon S Schneider
- Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.,Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.,Department of Psychiatry and Behavioral Sciences, University of Southern California, Los Angeles, CA, USA
| | - John C Morris
- The Hope Center for Neurodegenerative Disorders, Washington University School of Medicine, St. Louis, MO, USA.,Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Helena C Chui
- Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.,Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Meng Law
- Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.,Laboratory of Neuro Imaging, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.,Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Arthur W Toga
- Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.,Laboratory of Neuro Imaging, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Berislav V Zlokovic
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA. .,Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
22
|
Immonen R, Smith G, Brady RD, Wright D, Johnston L, Harris NG, Manninen E, Salo R, Branch C, Duncan D, Cabeen R, Ndode-Ekane XE, Gomez CS, Casillas-Espinosa PM, Ali I, Shultz SR, Andrade P, Puhakka N, Staba RJ, O'Brien TJ, Toga AW, Pitkänen A, Gröhn O. Harmonization of pipeline for preclinical multicenter MRI biomarker discovery in a rat model of post-traumatic epileptogenesis. Epilepsy Res 2019; 150:46-57. [PMID: 30641351 DOI: 10.1016/j.eplepsyres.2019.01.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 12/12/2018] [Accepted: 01/05/2019] [Indexed: 02/07/2023]
Abstract
Preclinical imaging studies of posttraumatic epileptogenesis (PTE) have largely been proof-of-concept studies with limited animal numbers, and thus lack the statistical power for biomarker discovery. Epilepsy Bioinformatics Study for Antiepileptogenic Therapy (EpiBioS4Rx) is a pioneering multicenter trial investigating preclinical imaging biomarkers of PTE. EpiBios4Rx faced the issue of harmonizing the magnetic resonance imaging (MRI) procedures and imaging data metrics prior to its execution. We present here the harmonization process between three preclinical MRI facilities at the University of Eastern Finland (UEF), the University of Melbourne (Melbourne), and the University of California, Los Angeles (UCLA), and evaluate the uniformity of the obtained MRI data. Adult, male rats underwent a lateral fluid percussion injury (FPI) and were followed by MRI 2 days, 9 days, 1 month, and 5 months post-injury. Ex vivo scans of fixed brains were conducted 7 months post-injury as an end point follow-up. Four MRI modalities were used: T2-weighted imaging, multi-gradient-echo imaging, diffusion-weighted imaging, and magnetization transfer imaging, and acquisition parameters for each modality were tailored to account for the different field strengths (4.7 T and 7 T) and different MR hardwares used at the three participating centers. Pilot data collection resulted in comparable image quality across sites. In interim analysis (of data obtained by April 30, 2018), the within-site variation of the quantified signal properties was low, while some differences between sites remained. In T2-weighted images the signal-to-noise ratios were high at each site, being 35 at UEF, 48 at Melbourne, and 32 at UCLA (p < 0.05). The contrast-to-noise ratios were similar between the sites (9, 10, and 8, respectively). Magnetization transfer ratio maps had identical white matter/ gray matter contrast between the sites, with white matter showing 15% higher MTR than gray matter despite different absolute MTR values (MTR both in white and gray matter was 3% lower in Melbourne than at UEF, p < 0.05). Diffusion-weighting yielded different degrees of signal attenuation across sites, being 83% at UEF, 76% in Melbourne, and 80% at UCLA (p < 0.05). Fractional anisotropy values differed as well, being 0.81 at UEF, 0.73 in Melbourne, and 0.84 at UCLA (p < 0.05). The obtained values in sham animals showed low variation within each site and no change over time, suggesting high repeatability of the measurements. Quality control scans with phantoms demonstrated stable hardware performance over time. Timing of post-TBI scans was designed to target specific phases of the dynamic pathology, and the execution at different centers was highly accurate. Besides a few outliers, the 2-day scans were done within an hour from the target time point. At day 9, most animals were scanned within an hour from the target time point, and all but 2 outliers within 24 h from the target. The 1-month post-TBI scans were done within 31 ± 3 days. MRI procedures and animal physiology during scans were similar between the sites. Taken together, the 10% inter-site difference in FA and 3% difference in MTR values should be included into analysis as a covariate or balanced out in post-processing in order to detect disease-related effects on brain structure at the same scale. However, for a MRI biomarker for post-traumatic epileptogenesis to have realistic chance of being successfully translated to validation in clinical trials, it would need to be a robust TBI-induced structural change which tolerates the inter-site methodological variability described here.
Collapse
Affiliation(s)
- Riikka Immonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FIN-70211 Kuopio, Finland.
| | - Gregory Smith
- Department of Neurology, David Geffen School of Medicine at University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Rhys D Brady
- Departments of Neuroscience and Neurology, Central Clinical School, Alfred Health, Monash University, Melbourne, Victoria, Australia; Departments of Medicine and Neurology, The Royal Melbourne Hospital, The University of Melbourne, Parkville, Victoria, Australia
| | - David Wright
- Departments of Neuroscience and Neurology, Central Clinical School, Alfred Health, Monash University, Melbourne, Victoria, Australia
| | - Leigh Johnston
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC 3052, Australia
| | - Neil G Harris
- Department of Neurology, David Geffen School of Medicine at University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Eppu Manninen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FIN-70211 Kuopio, Finland
| | - Raimo Salo
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FIN-70211 Kuopio, Finland
| | - Craig Branch
- Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Dominique Duncan
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA 90033, USA
| | - Ryan Cabeen
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA 90033, USA
| | - Xavier Ekolle Ndode-Ekane
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FIN-70211 Kuopio, Finland
| | - Cesar Santana Gomez
- Department of Neurology, David Geffen School of Medicine at University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Pablo M Casillas-Espinosa
- Departments of Neuroscience and Neurology, Central Clinical School, Alfred Health, Monash University, Melbourne, Victoria, Australia; Departments of Medicine and Neurology, The Royal Melbourne Hospital, The University of Melbourne, Parkville, Victoria, Australia
| | - Idrish Ali
- Departments of Neuroscience and Neurology, Central Clinical School, Alfred Health, Monash University, Melbourne, Victoria, Australia; Departments of Medicine and Neurology, The Royal Melbourne Hospital, The University of Melbourne, Parkville, Victoria, Australia
| | - Sandy R Shultz
- Departments of Neuroscience and Neurology, Central Clinical School, Alfred Health, Monash University, Melbourne, Victoria, Australia; Departments of Medicine and Neurology, The Royal Melbourne Hospital, The University of Melbourne, Parkville, Victoria, Australia
| | - Pedro Andrade
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FIN-70211 Kuopio, Finland
| | - Noora Puhakka
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FIN-70211 Kuopio, Finland
| | - Richard J Staba
- Department of Neurology, David Geffen School of Medicine at University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Terence J O'Brien
- Departments of Neuroscience and Neurology, Central Clinical School, Alfred Health, Monash University, Melbourne, Victoria, Australia; Departments of Medicine and Neurology, The Royal Melbourne Hospital, The University of Melbourne, Parkville, Victoria, Australia
| | - Arthur W Toga
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA 90033, USA
| | - Asla Pitkänen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FIN-70211 Kuopio, Finland
| | - Olli Gröhn
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FIN-70211 Kuopio, Finland
| |
Collapse
|
23
|
Irimia A, Lei X, Torgerson CM, Jacokes ZJ, Abe S, Van Horn JD. Support Vector Machines, Multidimensional Scaling and Magnetic Resonance Imaging Reveal Structural Brain Abnormalities Associated With the Interaction Between Autism Spectrum Disorder and Sex. Front Comput Neurosci 2018; 12:93. [PMID: 30534065 PMCID: PMC6276724 DOI: 10.3389/fncom.2018.00093] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Accepted: 11/02/2018] [Indexed: 11/28/2022] Open
Abstract
Despite substantial efforts, it remains difficult to identify reliable neuroanatomic biomarkers of autism spectrum disorder (ASD) based on magnetic resonance imaging (MRI) and diffusion tensor imaging (DTI). Studies which use standard statistical methods to approach this task have been hampered by numerous challenges, many of which are innate to the mathematical formulation and assumptions of general linear models (GLM). Although the potential of alternative approaches such as machine learning (ML) to identify robust neuroanatomic correlates of psychiatric disease has long been acknowledged, few studies have attempted to evaluate the abilities of ML to identify structural brain abnormalities associated with ASD. Here we use a sample of 110 ASD patients and 83 typically developing (TD) volunteers (95 females) to assess the suitability of support vector machines (SVMs, a robust type of ML) as an alternative to standard statistical inference for identifying structural brain features which can reliably distinguish ASD patients from TD subjects of either sex, thereby facilitating the study of the interaction between ASD diagnosis and sex. We find that SVMs can perform these tasks with high accuracy and that the neuroanatomic correlates of ASD identified using SVMs overlap substantially with those found using conventional statistical methods. Our results confirm and establish SVMs as powerful ML tools for the study of ASD-related structural brain abnormalities. Additionally, they provide novel insights into the volumetric, morphometric, and connectomic correlates of this epidemiologically significant disorder.
Collapse
Affiliation(s)
- Andrei Irimia
- Laboratory of Neuro Imaging, Keck School of Medicine, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, United States
- Ethel Percy Andrus Gerontology Center, Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, United States
| | - Xiaoyu Lei
- Laboratory of Neuro Imaging, Keck School of Medicine, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, United States
| | - Carinna M. Torgerson
- Laboratory of Neuro Imaging, Keck School of Medicine, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, United States
| | - Zachary J. Jacokes
- Laboratory of Neuro Imaging, Keck School of Medicine, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, United States
| | - Sumiko Abe
- Laboratory of Neuro Imaging, Keck School of Medicine, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, United States
| | - John D. Van Horn
- Laboratory of Neuro Imaging, Keck School of Medicine, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
24
|
Kalinin AA, Allyn-Feuer A, Ade A, Fon GV, Meixner W, Dilworth D, Husain SS, de Wet JR, Higgins GA, Zheng G, Creekmore A, Wiley JW, Verdone JE, Veltri RW, Pienta KJ, Coffey DS, Athey BD, Dinov ID. 3D Shape Modeling for Cell Nuclear Morphological Analysis and Classification. Sci Rep 2018; 8:13658. [PMID: 30209281 PMCID: PMC6135819 DOI: 10.1038/s41598-018-31924-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 08/29/2018] [Indexed: 02/08/2023] Open
Abstract
Quantitative analysis of morphological changes in a cell nucleus is important for the understanding of nuclear architecture and its relationship with pathological conditions such as cancer. However, dimensionality of imaging data, together with a great variability of nuclear shapes, presents challenges for 3D morphological analysis. Thus, there is a compelling need for robust 3D nuclear morphometric techniques to carry out population-wide analysis. We propose a new approach that combines modeling, analysis, and interpretation of morphometric characteristics of cell nuclei and nucleoli in 3D. We used robust surface reconstruction that allows accurate approximation of 3D object boundary. Then, we computed geometric morphological measures characterizing the form of cell nuclei and nucleoli. Using these features, we compared over 450 nuclei with about 1,000 nucleoli of epithelial and mesenchymal prostate cancer cells, as well as 1,000 nuclei with over 2,000 nucleoli from serum-starved and proliferating fibroblast cells. Classification of sets of 9 and 15 cells achieved accuracy of 95.4% and 98%, respectively, for prostate cancer cells, and 95% and 98% for fibroblast cells. To our knowledge, this is the first attempt to combine these methods for 3D nuclear shape modeling and morphometry into a highly parallel pipeline workflow for morphometric analysis of thousands of nuclei and nucleoli in 3D.
Collapse
Affiliation(s)
- Alexandr A Kalinin
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA.,Statistics Online Computational Resource (SOCR), Department of Health Behavior and Biological Sciences, University of Michigan School of Nursing, Ann Arbor, MI, USA
| | - Ari Allyn-Feuer
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Alex Ade
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Gordon-Victor Fon
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Walter Meixner
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - David Dilworth
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Syed S Husain
- Statistics Online Computational Resource (SOCR), Department of Health Behavior and Biological Sciences, University of Michigan School of Nursing, Ann Arbor, MI, USA
| | - Jeffrey R de Wet
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Gerald A Higgins
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Gen Zheng
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Amy Creekmore
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - John W Wiley
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - James E Verdone
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert W Veltri
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kenneth J Pienta
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Donald S Coffey
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Brian D Athey
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA. .,Michigan Institute for Data Science (MIDAS), University of Michigan, Ann Arbor, MI, USA.
| | - Ivo D Dinov
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA. .,Statistics Online Computational Resource (SOCR), Department of Health Behavior and Biological Sciences, University of Michigan School of Nursing, Ann Arbor, MI, USA. .,Michigan Institute for Data Science (MIDAS), University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
25
|
Sepehrband F, Lynch KM, Cabeen RP, Gonzalez-Zacarias C, Zhao L, D'Arcy M, Kesselman C, Herting MM, Dinov ID, Toga AW, Clark KA. Neuroanatomical morphometric characterization of sex differences in youth using statistical learning. Neuroimage 2018; 172:217-227. [PMID: 29414494 PMCID: PMC5967879 DOI: 10.1016/j.neuroimage.2018.01.065] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 01/10/2018] [Accepted: 01/25/2018] [Indexed: 12/31/2022] Open
Abstract
Exploring neuroanatomical sex differences using a multivariate statistical learning approach can yield insights that cannot be derived with univariate analysis. While gross differences in total brain volume are well-established, uncovering the more subtle, regional sex-related differences in neuroanatomy requires a multivariate approach that can accurately model spatial complexity as well as the interactions between neuroanatomical features. Here, we developed a multivariate statistical learning model using a support vector machine (SVM) classifier to predict sex from MRI-derived regional neuroanatomical features from a single-site study of 967 healthy youth from the Philadelphia Neurodevelopmental Cohort (PNC). Then, we validated the multivariate model on an independent dataset of 682 healthy youth from the multi-site Pediatric Imaging, Neurocognition and Genetics (PING) cohort study. The trained model exhibited an 83% cross-validated prediction accuracy, and correctly predicted the sex of 77% of the subjects from the independent multi-site dataset. Results showed that cortical thickness of the middle occipital lobes and the angular gyri are major predictors of sex. Results also demonstrated the inferential benefits of going beyond classical regression approaches to capture the interactions among brain features in order to better characterize sex differences in male and female youths. We also identified specific cortical morphological measures and parcellation techniques, such as cortical thickness as derived from the Destrieux atlas, that are better able to discriminate between males and females in comparison to other brain atlases (Desikan-Killiany, Brodmann and subcortical atlases).
Collapse
Affiliation(s)
- Farshid Sepehrband
- Laboratory of Neuro Imaging, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA.
| | - Kirsten M Lynch
- Laboratory of Neuro Imaging, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA; Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
| | - Ryan P Cabeen
- Laboratory of Neuro Imaging, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Clio Gonzalez-Zacarias
- Laboratory of Neuro Imaging, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA; Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
| | - Lu Zhao
- Laboratory of Neuro Imaging, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Mike D'Arcy
- USC Information Sciences Institute, University of Southern California, Los Angeles, CA, USA
| | - Carl Kesselman
- USC Information Sciences Institute, University of Southern California, Los Angeles, CA, USA
| | - Megan M Herting
- Department of Preventive Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA; Department of Pediatrics, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Ivo D Dinov
- Laboratory of Neuro Imaging, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA; Statistics Online Computational Resource, Department of Health Behavior and Biological, University of Michigan, Ann Arbor, MI, USA
| | - Arthur W Toga
- Laboratory of Neuro Imaging, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Kristi A Clark
- Laboratory of Neuro Imaging, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
26
|
Moon SW, Lee B, Choi YC. Changes in the Hippocampal Volume and Shape in Early-Onset Mild Cognitive Impairment. Psychiatry Investig 2018; 15:531-537. [PMID: 29695149 PMCID: PMC5976007 DOI: 10.30773/pi.2018.02.12] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 02/12/2018] [Indexed: 01/11/2023] Open
Abstract
OBJECTIVE The aim of this study was to examine the change in the hippocampal volume and shape in early-onset mild cognitive impairment (EO-MCI) associated with the APOE ε4 carrier state. METHODS This study had 50 subjects aged 55-63 years, all of whom were diagnosed with MCI at baseline via the Korean version of the Consortium to Establish a Registry for Alzheimer's Disease Assessment Packet. The EO-MCI patients were divided into the MCI continued (MCIcont) and Alzheimer's disease (AD) converted (ADconv) groups 2 years later. The hippocampal volume and shape were measured for all the subjects. The local shape analysis (LSA) was used to conduct based on the 2-year-interval magnetic resonance imaging scans. RESULTS There was a significant correlation between APOE ε4 allele and hippocampal volume atrophy. Over two years, the volume reduction in the left hippocampus was found to be faster than that in the right hippocampus, especially in the APOE ε4 carriers. LSA showed that the 2 subfields were significantly affected in the left hippocampus. CONCLUSION These results suggest that the possession of APOE ε4 allele may lead to greater predilection for left hippocampal atrophy in EO-MCI, and some specific subfields of the hippocampus may be more prominently involved.
Collapse
Affiliation(s)
- Seok Woo Moon
- Department of Psychiatry, Konkuk University School of Medicine, Chungju, Republic of Korea
| | - Boram Lee
- Department of Psychiatry, Graduate School of Konkuk University, Seoul, Republic of Korea
| | - Young Chil Choi
- Department of Radiology, Konkuk University School of Medicine, Chungju, Republic of Korea
| |
Collapse
|
27
|
Leandrou S, Petroudi S, Kyriacou PA, Reyes-Aldasoro CC, Pattichis CS. Quantitative MRI Brain Studies in Mild Cognitive Impairment and Alzheimer's Disease: A Methodological Review. IEEE Rev Biomed Eng 2018; 11:97-111. [PMID: 29994606 DOI: 10.1109/rbme.2018.2796598] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Classifying and predicting Alzheimer's disease (AD) in individuals with memory disorders through clinical and psychometric assessment is challenging, especially in mild cognitive impairment (MCI) subjects. Quantitative structural magnetic resonance imaging acquisition methods in combination with computer-aided diagnosis are currently being used for the assessment of AD. These acquisitions methods include voxel-based morphometry, volumetric measurements in specific regions of interest (ROIs), cortical thickness measurements, shape analysis, and texture analysis. This review evaluates the aforementioned methods in the classification of cases into one of the following three groups: normal controls, MCI, and AD subjects. Furthermore, the performance of the methods is assessed on the prediction of conversion from MCI to AD. In parallel, it is also assessed which ROIs are preferred in both classification and prognosis through the different states of the disease. Structural changes in the early stages of the disease are more pronounced in the medial temporal lobe, especially in the entorhinal cortex, whereas with disease progression, both entorhinal cortex and hippocampus offer similar discriminative power. However, for the conversion from MCI subjects to AD, entorhinal cortex provides better predictive accuracies rather than other structures, such as the hippocampus.
Collapse
|
28
|
Hinojosa-Rodríguez M, Harmony T, Carrillo-Prado C, Van Horn JD, Irimia A, Torgerson C, Jacokes Z. Clinical neuroimaging in the preterm infant: Diagnosis and prognosis. Neuroimage Clin 2017; 16:355-368. [PMID: 28861337 PMCID: PMC5568883 DOI: 10.1016/j.nicl.2017.08.015] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Revised: 08/11/2017] [Accepted: 08/12/2017] [Indexed: 01/30/2023]
Abstract
Perinatal care advances emerging over the past twenty years have helped to diminish the mortality and severe neurological morbidity of extremely and very preterm neonates (e.g., cystic Periventricular Leukomalacia [c-PVL] and Germinal Matrix Hemorrhage - Intraventricular Hemorrhage [GMH-IVH grade 3-4/4]; 22 to < 32 weeks of gestational age, GA). However, motor and/or cognitive disabilities associated with mild-to-moderate white and gray matter injury are frequently present in this population (e.g., non-cystic Periventricular Leukomalacia [non-cystic PVL], neuronal-axonal injury and GMH-IVH grade 1-2/4). Brain research studies using magnetic resonance imaging (MRI) report that 50% to 80% of extremely and very preterm neonates have diffuse white matter abnormalities (WMA) which correspond to only the minimum grade of severity. Nevertheless, mild-to-moderate diffuse WMA has also been associated with significant affectations of motor and cognitive activities. Due to increased neonatal survival and the intrinsic characteristics of diffuse WMA, there is a growing need to study the brain of the premature infant using non-invasive neuroimaging techniques sensitive to microscopic and/or diffuse lesions. This emerging need has led the scientific community to try to bridge the gap between concepts or ideas from different methodologies and approaches; for instance, neuropathology, neuroimaging and clinical findings. This is evident from the combination of intense pre-clinical and clinicopathologic research along with neonatal neurology and quantitative neuroimaging research. In the following review, we explore literature relating the most frequently observed neuropathological patterns with the recent neuroimaging findings in preterm newborns and infants with perinatal brain injury. Specifically, we focus our discussions on the use of neuroimaging to aid diagnosis, measure morphometric brain damage, and track long-term neurodevelopmental outcomes.
Collapse
Affiliation(s)
- Manuel Hinojosa-Rodríguez
- Unidad de Investigación en Neurodesarrollo, Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus Juriquilla, Mexico
| | - Thalía Harmony
- Unidad de Investigación en Neurodesarrollo, Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus Juriquilla, Mexico
| | - Cristina Carrillo-Prado
- Unidad de Investigación en Neurodesarrollo, Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Campus Juriquilla, Mexico
| | - John Darrell Van Horn
- USC Mark and Mary Stevens Neuroimaging and Informatics Institute, 2025 Zonal Avenue, SHN, Los Angeles, California 90033, USA
| | - Andrei Irimia
- USC Mark and Mary Stevens Neuroimaging and Informatics Institute, 2025 Zonal Avenue, SHN, Los Angeles, California 90033, USA
| | - Carinna Torgerson
- USC Mark and Mary Stevens Neuroimaging and Informatics Institute, 2025 Zonal Avenue, SHN, Los Angeles, California 90033, USA
| | - Zachary Jacokes
- USC Mark and Mary Stevens Neuroimaging and Informatics Institute, 2025 Zonal Avenue, SHN, Los Angeles, California 90033, USA
| |
Collapse
|
29
|
Medina CS, Manifold-Wheeler B, Gonzales A, Bearer EL. Automated Computational Processing of 3-D MR Images of Mouse Brain for Phenotyping of Living Animals. ACTA ACUST UNITED AC 2017; 119:29A.5.1-29A.5.38. [PMID: 28678440 DOI: 10.1002/cpmb.40] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Magnetic resonance (MR) imaging provides a method to obtain anatomical information from the brain in vivo that is not typically available by optical imaging because of this organ's opacity. MR is nondestructive and obtains deep tissue contrast with 100-µm3 voxel resolution or better. Manganese-enhanced MRI (MEMRI) may be used to observe axonal transport and localized neural activity in the living rodent and avian brain. Such enhancement enables researchers to investigate differences in functional circuitry or neuronal activity in images of brains of different animals. Moreover, once MR images of a number of animals are aligned into a single matrix, statistical analysis can be done comparing MR intensities between different multi-animal cohorts comprising individuals from different mouse strains or different transgenic animals, or at different time points after an experimental manipulation. Although preprocessing steps for such comparisons (including skull stripping and alignment) are automated for human imaging, no such automated processing has previously been readily available for mouse or other widely used experimental animals, and most investigators use in-house custom processing. This protocol describes a stepwise method to perform such preprocessing for mouse. © 2017 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
| | | | - Aaron Gonzales
- University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Elaine L Bearer
- University of New Mexico Health Sciences Center, Albuquerque, New Mexico.,Division of Biology, California Institute of Technology, Pasadena, California
| |
Collapse
|
30
|
Kiar G, Gorgolewski KJ, Kleissas D, Roncal WG, Litt B, Wandell B, Poldrack RA, Wiener M, Vogelstein RJ, Burns R, Vogelstein JT. Science in the cloud (SIC): A use case in MRI connectomics. Gigascience 2017; 6:1-10. [PMID: 28327935 PMCID: PMC5467033 DOI: 10.1093/gigascience/gix013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 02/13/2017] [Accepted: 03/01/2017] [Indexed: 01/08/2023] Open
Abstract
Modern technologies are enabling scientists to collect extraordinary amounts of complex and sophisticated data across a huge range of scales like never before. With this onslaught of data, we can allow the focal point to shift from data collection to data analysis. Unfortunately, lack of standardized sharing mechanisms and practices often make reproducing or extending scientific results very difficult. With the creation of data organization structures and tools that drastically improve code portability, we now have the opportunity to design such a framework for communicating extensible scientific discoveries. Our proposed solution leverages these existing technologies and standards, and provides an accessible and extensible model for reproducible research, called 'science in the cloud' (SIC). Exploiting scientific containers, cloud computing, and cloud data services, we show the capability to compute in the cloud and run a web service that enables intimate interaction with the tools and data presented. We hope this model will inspire the community to produce reproducible and, importantly, extensible results that will enable us to collectively accelerate the rate at which scientific breakthroughs are discovered, replicated, and extended.
Collapse
Affiliation(s)
- Gregory Kiar
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Center for Imaging Science, Johns Hopkins University, Baltimore, MD, USA
| | | | - Dean Kleissas
- Johns Hopkins University Applied Physics Lab, Columbia, MD, USA
| | - William Gray Roncal
- Johns Hopkins University Applied Physics Lab, Columbia, MD, USA
- Department of Computer Science, Johns Hopkins University, Baltimore, MD, USA
| | - Brian Litt
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Brian Wandell
- Department of Psychology, Stanford University, Stanford, CA, USA
- Center for Cognitive and Neurobiological Imaging, Stanford University, Stanford, CA, USA
| | | | - Martin Wiener
- Department of Psychology, George Mason University, Fairfax, VA, USA
| | | | - Randal Burns
- Department of Computer Science, Johns Hopkins University, Baltimore, MD, USA
| | - Joshua T. Vogelstein
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Center for Imaging Science, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
31
|
He KY, Ge D, He MM. Big Data Analytics for Genomic Medicine. Int J Mol Sci 2017; 18:ijms18020412. [PMID: 28212287 PMCID: PMC5343946 DOI: 10.3390/ijms18020412] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 02/08/2017] [Accepted: 02/09/2017] [Indexed: 12/25/2022] Open
Abstract
Genomic medicine attempts to build individualized strategies for diagnostic or therapeutic decision-making by utilizing patients’ genomic information. Big Data analytics uncovers hidden patterns, unknown correlations, and other insights through examining large-scale various data sets. While integration and manipulation of diverse genomic data and comprehensive electronic health records (EHRs) on a Big Data infrastructure exhibit challenges, they also provide a feasible opportunity to develop an efficient and effective approach to identify clinically actionable genetic variants for individualized diagnosis and therapy. In this paper, we review the challenges of manipulating large-scale next-generation sequencing (NGS) data and diverse clinical data derived from the EHRs for genomic medicine. We introduce possible solutions for different challenges in manipulating, managing, and analyzing genomic and clinical data to implement genomic medicine. Additionally, we also present a practical Big Data toolset for identifying clinically actionable genetic variants using high-throughput NGS data and EHRs.
Collapse
Affiliation(s)
- Karen Y He
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, OH 44106, USA.
| | | | - Max M He
- BioSciKin Co., Ltd., Nanjing 210042, China.
- Computation and Informatics in Biology and Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|
32
|
Paholpak P, Carr AR, Barsuglia JP, Barrows RJ, Jimenez E, Lee GJ, Mendez MF. Person-Based Versus Generalized Impulsivity Disinhibition in Frontotemporal Dementia and Alzheimer Disease. J Geriatr Psychiatry Neurol 2016; 29:344-351. [PMID: 27647788 DOI: 10.1177/0891988716666377] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND While much disinhibition in dementia results from generalized impulsivity, in behavioral variant frontotemporal dementia (bvFTD) disinhibition may also result from impaired social cognition. OBJECTIVE To deconstruct disinhibition and its neural correlates in bvFTD vs. early-onset Alzheimer's disease (eAD). METHODS Caregivers of 16 bvFTD and 21 matched-eAD patients completed the Frontal Systems Behavior Scale disinhibition items. The disinhibition items were further categorized into (1) "person-based" subscale which predominantly associated with violating social propriety and personal boundary and (2) "generalized-impulsivity" subscale which included nonspecific impulsive acts. Subscale scores were correlated with grey matter volumes from tensor-based morphometry on magnetic resonance images. RESULTS In comparison to the eAD patients, the bvFTD patients developed greater person-based disinhibition ( P < 0.001) but comparable generalized impulsivity. Severity of person-based disinhibition significantly correlated with the left anterior superior temporal sulcus (STS), and generalized-impulsivity correlated with the right orbitofrontal cortex (OFC) and the left anterior temporal lobe (aTL). CONCLUSIONS Person-based disinhibition was predominant in bvFTD and correlated with the left STS. In both dementia, violations of social propriety and personal boundaries involved fronto-parieto-temporal network of Theory of Mind, whereas nonspecific disinhibition involved the OFC and aTL.
Collapse
Affiliation(s)
- Pongsatorn Paholpak
- 1 Department of Neurology, David Geffen School of Medicine, University of California at Los Angeles, CA, USA.,2 Department of Psychiatry, Khon Kaen University, Khon Kaen, Thailand
| | - Andrew R Carr
- 1 Department of Neurology, David Geffen School of Medicine, University of California at Los Angeles, CA, USA.,3 Greater Los Angeles VA Healthcare System, West Los Angeles, CA, USA
| | | | - Robin J Barrows
- 1 Department of Neurology, David Geffen School of Medicine, University of California at Los Angeles, CA, USA.,3 Greater Los Angeles VA Healthcare System, West Los Angeles, CA, USA
| | - Elvira Jimenez
- 1 Department of Neurology, David Geffen School of Medicine, University of California at Los Angeles, CA, USA.,3 Greater Los Angeles VA Healthcare System, West Los Angeles, CA, USA.,4 Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine, University of California at Los Angeles, CA, USA
| | - Grace J Lee
- 5 Department of Psychology, School of Behavioral Health, Loma Linda University, Loma Linda, CA, USA
| | - Mario F Mendez
- 1 Department of Neurology, David Geffen School of Medicine, University of California at Los Angeles, CA, USA.,3 Greater Los Angeles VA Healthcare System, West Los Angeles, CA, USA.,4 Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine, University of California at Los Angeles, CA, USA
| |
Collapse
|
33
|
Fu KA, Nathan R, Dinov ID, Li J, Toga AW. T2-Imaging Changes in the Nigrosome-1 Relate to Clinical Measures of Parkinson's Disease. Front Neurol 2016; 7:174. [PMID: 27812347 PMCID: PMC5071353 DOI: 10.3389/fneur.2016.00174] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 09/27/2016] [Indexed: 01/23/2023] Open
Abstract
Background The nigrosome-1 region of the substantia nigra (SN) undergoes the greatest and earliest dopaminergic neuron loss in Parkinson’s disease (PD). As T2-weighted magnetic resonance imaging (MRI) scans are often collected with routine clinical MRI protocols, this investigation aims to determine whether T2-imaging changes in the nigrosome-1 are related to clinical measures of PD and to assess their potential as a more clinically accessible biomarker for PD. Methods Voxel intensity ratios were calculated for T2-weighted MRI scans from 47 subjects from the Parkinson’s Progression Markers Initiative database. Three approaches were used to delineate the SN and nigrosome-1: (1) manual segmentation, (2) automated segmentation, and (3) area voxel-based morphometry. Voxel intensity ratios were calculated from voxel intensity values taken from the nigrosome-1 and two areas of the remaining SN. Linear regression analyses were conducted relating voxel intensity ratios with the Movement Disorder Society-Unified Parkinson’s Disease Rating Scale (MDS-UPDRS) sub-scores for each subject. Results For manual segmentation, linear regression tests consistently identified the voxel intensity ratio derived from the dorsolateral SN and nigrosome-1 (IR2) as predictive of nBehav (p = 0.0377) and nExp (p = 0.03856). For automated segmentation, linear regression tests identified IR2 as predictive of Subscore IA (nBehav) (p = 0.01134), Subscore IB (nExp) (p = 0.00336), Score II (mExp) (p = 0.02125), and Score III (mSign) (p = 0.008139). For the voxel-based morphometric approach, univariate simple linear regression analysis identified IR2 as yielding significant results for nBehav (p = 0.003102), mExp (p = 0.0172), and mSign (p = 0.00393). Conclusion Neuroimaging biomarkers may be used as a proxy of changes in the nigrosome-1, measured by MDS-UPDRS scores as an indicator of the severity of PD. The voxel intensity ratio derived from the dorsolateral SN and nigrosome-1 was consistently predictive of non-motor complex behaviors in all three analyses and predictive of non-motor experiences of daily living, motor experiences of daily living, and motor signs of PD in two of the three analyses. These results suggest that T2 changes in the nigrosome-1 may relate to certain clinical measures of PD. T2 changes in the nigrosome-1 may be considered when developing a more accessible clinical diagnostic tool for patients with suspected PD.
Collapse
Affiliation(s)
- Katherine A Fu
- Laboratory of Neuro Imaging, Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA; Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Romil Nathan
- Laboratory of Neuro Imaging, Stevens Neuroimaging and Informatics Institute, University of Southern California , Los Angeles, CA , USA
| | - Ivo D Dinov
- Statistics Online Computational Resource, Health Behavior and Biological Sciences, University of Michigan , Ann Arbor, MI , USA
| | - Junning Li
- Laboratory of Neuro Imaging, Stevens Neuroimaging and Informatics Institute, University of Southern California , Los Angeles, CA , USA
| | - Arthur W Toga
- Laboratory of Neuro Imaging, Stevens Neuroimaging and Informatics Institute, University of Southern California , Los Angeles, CA , USA
| |
Collapse
|
34
|
Kim W, Chivukula S, Hauptman J, Pouratian N. Diffusion Tensor Imaging-Based Thalamic Segmentation in Deep Brain Stimulation for Chronic Pain Conditions. Stereotact Funct Neurosurg 2016; 94:225-234. [PMID: 27537848 DOI: 10.1159/000448079] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 06/29/2016] [Indexed: 11/19/2022]
Abstract
BACKGROUND/AIMS Thalamic deep brain stimulation (DBS) for the treatment of medically refractory pain has largely been abandoned on account of its inconsistent and oftentimes poor efficacy. Our aim here was to use diffusion tensor imaging (DTI)-based segmentation to assess the internal thalamic nuclei of patients who have undergone thalamic DBS for intractable pain and retrospectively correlate lead position with clinical outcome. METHODS DTI-based segmentation was performed on 5 patients who underwent sensory thalamus DBS for chronic pain. Postoperative computed tomography images obtained for electrode placement were fused with preoperative magnetic resonance images that had undergone DTI-based thalamic segmentation. Sensory thalamus maps of 4 patients were analyzed for lead positioning and interpatient variability. RESULTS Four patients who experienced significant pain relief following DBS demonstrated contact positions within the DTI-determined sensory thalamus or in its vicinity, whereas 1 patient who did not respond to stimulation did not. Only 4 voxels (2%) within the sensory thalamus were mutually shared among patients; 108 voxels (58%) were uniquely represented. CONCLUSIONS DTI-based segmentation of the thalamus can be used to confirm thalamic lead placement relative to the sensory thalamus and may serve as a useful tool to guide thalamic DBS electrode implantation in the future.
Collapse
Affiliation(s)
- Won Kim
- Department of Neurological Surgery, University of California, Los Angeles, Calif., USA
| | | | | | | |
Collapse
|
35
|
Plis SM, Sarwate AD, Wood D, Dieringer C, Landis D, Reed C, Panta SR, Turner JA, Shoemaker JM, Carter KW, Thompson P, Hutchison K, Calhoun VD. COINSTAC: A Privacy Enabled Model and Prototype for Leveraging and Processing Decentralized Brain Imaging Data. Front Neurosci 2016; 10:365. [PMID: 27594820 PMCID: PMC4990563 DOI: 10.3389/fnins.2016.00365] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 07/22/2016] [Indexed: 01/17/2023] Open
Abstract
The field of neuroimaging has embraced the need for sharing and collaboration. Data sharing mandates from public funding agencies and major journal publishers have spurred the development of data repositories and neuroinformatics consortia. However, efficient and effective data sharing still faces several hurdles. For example, open data sharing is on the rise but is not suitable for sensitive data that are not easily shared, such as genetics. Current approaches can be cumbersome (such as negotiating multiple data sharing agreements). There are also significant data transfer, organization and computational challenges. Centralized repositories only partially address the issues. We propose a dynamic, decentralized platform for large scale analyses called the Collaborative Informatics and Neuroimaging Suite Toolkit for Anonymous Computation (COINSTAC). The COINSTAC solution can include data missing from central repositories, allows pooling of both open and "closed" repositories by developing privacy-preserving versions of widely-used algorithms, and incorporates the tools within an easy-to-use platform enabling distributed computation. We present an initial prototype system which we demonstrate on two multi-site data sets, without aggregating the data. In addition, by iterating across sites, the COINSTAC model enables meta-analytic solutions to converge to "pooled-data" solutions (i.e., as if the entire data were in hand). More advanced approaches such as feature generation, matrix factorization models, and preprocessing can be incorporated into such a model. In sum, COINSTAC enables access to the many currently unavailable data sets, a user friendly privacy enabled interface for decentralized analysis, and a powerful solution that complements existing data sharing solutions.
Collapse
Affiliation(s)
- Sergey M. Plis
- The Mind Research Network, Lovelace Biomedical and Environmental Research InstituteAlbuquerque, NM, USA
| | - Anand D. Sarwate
- Department of Electrical and Computer Engineering, Rutgers, The State University of New JerseyPiscataway, NJ, USA
| | - Dylan Wood
- The Mind Research Network, Lovelace Biomedical and Environmental Research InstituteAlbuquerque, NM, USA
| | - Christopher Dieringer
- The Mind Research Network, Lovelace Biomedical and Environmental Research InstituteAlbuquerque, NM, USA
| | - Drew Landis
- The Mind Research Network, Lovelace Biomedical and Environmental Research InstituteAlbuquerque, NM, USA
| | - Cory Reed
- The Mind Research Network, Lovelace Biomedical and Environmental Research InstituteAlbuquerque, NM, USA
| | - Sandeep R. Panta
- The Mind Research Network, Lovelace Biomedical and Environmental Research InstituteAlbuquerque, NM, USA
| | - Jessica A. Turner
- The Mind Research Network, Lovelace Biomedical and Environmental Research InstituteAlbuquerque, NM, USA
- Department of Psychology and Neuroscience Institute, Georgia State UniversityAtlanta, GA, USA
| | - Jody M. Shoemaker
- The Mind Research Network, Lovelace Biomedical and Environmental Research InstituteAlbuquerque, NM, USA
| | - Kim W. Carter
- Telethon Kids Institute, The University of Western AustraliaSubiaco, WA, Australia
| | - Paul Thompson
- Departments of Neurology, Psychiatry, Engineering, Radiology, and Pediatrics, Imaging Genetics Center, Enhancing Neuroimaging and Genetics through Meta-Analysis Center for Worldwide Medicine, Imaging, and Genomics, University of Southern CaliforniaMarina del Rey, CA, USA
| | - Kent Hutchison
- Department of Psychology and Neuroscience, University of Colorado BoulderBoulder, CO, USA
| | - Vince D. Calhoun
- The Mind Research Network, Lovelace Biomedical and Environmental Research InstituteAlbuquerque, NM, USA
- Department of Electrical and Computer Engineering, University of New MexicoAlbuquerque, NM, USA
| |
Collapse
|
36
|
Dinov ID, Heavner B, Tang M, Glusman G, Chard K, Darcy M, Madduri R, Pa J, Spino C, Kesselman C, Foster I, Deutsch EW, Price ND, Van Horn JD, Ames J, Clark K, Hood L, Hampstead BM, Dauer W, Toga AW. Predictive Big Data Analytics: A Study of Parkinson's Disease Using Large, Complex, Heterogeneous, Incongruent, Multi-Source and Incomplete Observations. PLoS One 2016; 11:e0157077. [PMID: 27494614 PMCID: PMC4975403 DOI: 10.1371/journal.pone.0157077] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 05/24/2016] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND A unique archive of Big Data on Parkinson's Disease is collected, managed and disseminated by the Parkinson's Progression Markers Initiative (PPMI). The integration of such complex and heterogeneous Big Data from multiple sources offers unparalleled opportunities to study the early stages of prevalent neurodegenerative processes, track their progression and quickly identify the efficacies of alternative treatments. Many previous human and animal studies have examined the relationship of Parkinson's disease (PD) risk to trauma, genetics, environment, co-morbidities, or life style. The defining characteristics of Big Data-large size, incongruency, incompleteness, complexity, multiplicity of scales, and heterogeneity of information-generating sources-all pose challenges to the classical techniques for data management, processing, visualization and interpretation. We propose, implement, test and validate complementary model-based and model-free approaches for PD classification and prediction. To explore PD risk using Big Data methodology, we jointly processed complex PPMI imaging, genetics, clinical and demographic data. METHODS AND FINDINGS Collective representation of the multi-source data facilitates the aggregation and harmonization of complex data elements. This enables joint modeling of the complete data, leading to the development of Big Data analytics, predictive synthesis, and statistical validation. Using heterogeneous PPMI data, we developed a comprehensive protocol for end-to-end data characterization, manipulation, processing, cleaning, analysis and validation. Specifically, we (i) introduce methods for rebalancing imbalanced cohorts, (ii) utilize a wide spectrum of classification methods to generate consistent and powerful phenotypic predictions, and (iii) generate reproducible machine-learning based classification that enables the reporting of model parameters and diagnostic forecasting based on new data. We evaluated several complementary model-based predictive approaches, which failed to generate accurate and reliable diagnostic predictions. However, the results of several machine-learning based classification methods indicated significant power to predict Parkinson's disease in the PPMI subjects (consistent accuracy, sensitivity, and specificity exceeding 96%, confirmed using statistical n-fold cross-validation). Clinical (e.g., Unified Parkinson's Disease Rating Scale (UPDRS) scores), demographic (e.g., age), genetics (e.g., rs34637584, chr12), and derived neuroimaging biomarker (e.g., cerebellum shape index) data all contributed to the predictive analytics and diagnostic forecasting. CONCLUSIONS Model-free Big Data machine learning-based classification methods (e.g., adaptive boosting, support vector machines) can outperform model-based techniques in terms of predictive precision and reliability (e.g., forecasting patient diagnosis). We observed that statistical rebalancing of cohort sizes yields better discrimination of group differences, specifically for predictive analytics based on heterogeneous and incomplete PPMI data. UPDRS scores play a critical role in predicting diagnosis, which is expected based on the clinical definition of Parkinson's disease. Even without longitudinal UPDRS data, however, the accuracy of model-free machine learning based classification is over 80%. The methods, software and protocols developed here are openly shared and can be employed to study other neurodegenerative disorders (e.g., Alzheimer's, Huntington's, amyotrophic lateral sclerosis), as well as for other predictive Big Data analytics applications.
Collapse
Affiliation(s)
- Ivo D. Dinov
- Statistics Online Computational Resource, School of Nursing, Michigan Institute for Data Science, University of Michigan, Ann Arbor, Michigan, United States of America
- Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, California, United States of America
- Udall Center of Excellence for Parkinson’s Disease Research, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Ben Heavner
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - Ming Tang
- Statistics Online Computational Resource, School of Nursing, Michigan Institute for Data Science, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Gustavo Glusman
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - Kyle Chard
- Computation Institute, University of Chicago and Argonne National Laboratory, Chicago, Illinois, United States of America
| | - Mike Darcy
- Information Sciences Institute, University of Southern California, Los Angeles, California, United States of America
| | - Ravi Madduri
- Computation Institute, University of Chicago and Argonne National Laboratory, Chicago, Illinois, United States of America
| | - Judy Pa
- Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, California, United States of America
| | - Cathie Spino
- Udall Center of Excellence for Parkinson’s Disease Research, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Carl Kesselman
- Information Sciences Institute, University of Southern California, Los Angeles, California, United States of America
| | - Ian Foster
- Computation Institute, University of Chicago and Argonne National Laboratory, Chicago, Illinois, United States of America
| | - Eric W. Deutsch
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - Nathan D. Price
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - John D. Van Horn
- Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, California, United States of America
| | - Joseph Ames
- Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, California, United States of America
| | - Kristi Clark
- Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, California, United States of America
| | - Leroy Hood
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - Benjamin M. Hampstead
- Department of Psychiatry and Michigan Alzheimer’s Disease Center, University of Michigan, Ann Arbor, Michigan, United States of America
- Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan, United States of America
| | - William Dauer
- Udall Center of Excellence for Parkinson’s Disease Research, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Arthur W. Toga
- Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, California, United States of America
| |
Collapse
|
37
|
Panchal H, Paholpak P, Lee G, Carr A, Barsuglia JP, Mather M, Jimenez E, Mendez MF. Neuropsychological and Neuroanatomical Correlates of the Social Norms Questionnaire in Frontotemporal Dementia Versus Alzheimer's Disease. Am J Alzheimers Dis Other Demen 2016; 31:326-32. [PMID: 26646114 PMCID: PMC10852706 DOI: 10.1177/1533317515617722] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Traditional neuropsychological batteries may not distinguish early behavioral variant frontotemporal dementia (bvFTD) from Alzheimer's disease (AD) without the inclusion of a social behavioral measure. We compared 33 participants, 15 bvFTD, and 18 matched patients with early-onset AD (eAD), on the Social Norms Questionnaire (SNQ), neuropsychological tests and 3-dimensional T1-weighted magnetic resonance imaging (MRI). The analyses included correlations of SNQ results (total score, overendorsement or "overadhere" errors, and violations or "break" errors) with neuropsychological results and tensor-based morphometry regions of interest. Patients with BvFTD had significantly lower SNQ total scores and higher overadhere errors than patients with eAD. On neuropsychological measures, the SNQ total scores correlated significantly with semantic knowledge and the overadhere subscores with executive dysfunction. On MRI analysis, the break subscores significantly correlated with lower volume of lateral anterior temporal lobes (aTL). The results also suggest that endorsement of social norm violations corresponds to the role of the right aTL in social semantic knowledge.
Collapse
Affiliation(s)
- Hemali Panchal
- VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA Department of Neurology, Los Angeles, CA, USA
| | - Pongsatorn Paholpak
- VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA Department of Neurology, Los Angeles, CA, USA Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles, CA, USA Department of Psychiatry, Khon Kaen University, Khon Khaen, Thailand
| | - Grace Lee
- Department of Psychology, School of Behavioral Health, Loma Linda, CA, USA
| | - Andrew Carr
- VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | | | - Michelle Mather
- VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA Department of Neurology, Los Angeles, CA, USA
| | - Elvira Jimenez
- VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA Department of Neurology, Los Angeles, CA, USA Department of Psychiatry & Biobehavioral Sciences, Los Angeles, CA, USA
| | - Mario F Mendez
- VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA Department of Neurology, Los Angeles, CA, USA Department of Psychiatry & Biobehavioral Sciences, Los Angeles, CA, USA
| |
Collapse
|
38
|
Dinov ID. Methodological challenges and analytic opportunities for modeling and interpreting Big Healthcare Data. Gigascience 2016; 5:12. [PMID: 26918190 PMCID: PMC4766610 DOI: 10.1186/s13742-016-0117-6] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 02/09/2016] [Indexed: 11/25/2022] Open
Abstract
Managing, processing and understanding big healthcare data is challenging, costly and demanding. Without a robust fundamental theory for representation, analysis and inference, a roadmap for uniform handling and analyzing of such complex data remains elusive. In this article, we outline various big data challenges, opportunities, modeling methods and software techniques for blending complex healthcare data, advanced analytic tools, and distributed scientific computing. Using imaging, genetic and healthcare data we provide examples of processing heterogeneous datasets using distributed cloud services, automated and semi-automated classification techniques, and open-science protocols. Despite substantial advances, new innovative technologies need to be developed that enhance, scale and optimize the management and processing of large, complex and heterogeneous data. Stakeholder investments in data acquisition, research and development, computational infrastructure and education will be critical to realize the huge potential of big data, to reap the expected information benefits and to build lasting knowledge assets. Multi-faceted proprietary, open-source, and community developments will be essential to enable broad, reliable, sustainable and efficient data-driven discovery and analytics. Big data will affect every sector of the economy and their hallmark will be 'team science'.
Collapse
Affiliation(s)
- Ivo D. Dinov
- Statistics Online Computational Resource (SOCR), Health Behavior and Biological Sciences, Michigan Institute for Data Science, University of Michigan, 426 N. Ingalls, Ann Arbor, MI 49109 USA
| |
Collapse
|
39
|
Computational analysis in epilepsy neuroimaging: A survey of features and methods. NEUROIMAGE-CLINICAL 2016; 11:515-529. [PMID: 27114900 PMCID: PMC4833048 DOI: 10.1016/j.nicl.2016.02.013] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 02/11/2016] [Accepted: 02/22/2016] [Indexed: 12/15/2022]
Abstract
Epilepsy affects 65 million people worldwide, a third of whom have seizures that are resistant to anti-epileptic medications. Some of these patients may be amenable to surgical therapy or treatment with implantable devices, but this usually requires delineation of discrete structural or functional lesion(s), which is challenging in a large percentage of these patients. Advances in neuroimaging and machine learning allow semi-automated detection of malformations of cortical development (MCDs), a common cause of drug resistant epilepsy. A frequently asked question in the field is what techniques currently exist to assist radiologists in identifying these lesions, especially subtle forms of MCDs such as focal cortical dysplasia (FCD) Type I and low grade glial tumors. Below we introduce some of the common lesions encountered in patients with epilepsy and the common imaging findings that radiologists look for in these patients. We then review and discuss the computational techniques introduced over the past 10 years for quantifying and automatically detecting these imaging findings. Due to large variations in the accuracy and implementation of these studies, specific techniques are traditionally used at individual centers, often guided by local expertise, as well as selection bias introduced by the varying prevalence of specific patient populations in different epilepsy centers. We discuss the need for a multi-institutional study that combines features from different imaging modalities as well as computational techniques to definitively assess the utility of specific automated approaches to epilepsy imaging. We conclude that sharing and comparing these different computational techniques through a common data platform provides an opportunity to rigorously test and compare the accuracy of these tools across different patient populations and geographical locations. We propose that these kinds of tools, quantitative imaging analysis methods and open data platforms for aggregating and sharing data and algorithms, can play a vital role in reducing the cost of care, the risks of invasive treatments, and improve overall outcomes for patients with epilepsy. We introduce common epileptogenic lesions encountered in patients with drug resistant epilepsy. We discuss state of the art computational techniques used to detect lesions. There is a need for multi-institutional studies that combine these techniques. Clinically validated pipelines alongside the advances in imaging and electrophysiology will improve outcomes.
Collapse
Key Words
- DRE, drug resistant epilepsy
- DTI, diffusion tensor imaging
- DWI, diffusion weighted imaging
- Drug resistant epilepsy
- Epilepsy
- FCD, focal cortical dysplasia
- FLAIR, fluid-attenuated inversion recovery
- Focal cortical dysplasia
- GM, gray matter
- GW, gray-white junction
- HARDI, high angular resolution diffusion imaging
- MEG, magnetoencephalography
- MRS, magnetic resonance spectroscopy imaging
- Machine learning
- Malformations of cortical development
- Multimodal neuroimaging
- PET, positron emission tomography
- PNH, periventricular nodular heterotopia
- SBM, surface-based morphometry
- T1W, T1-weighted MRI
- T2W, T2-weighted MRI
- VBM, voxel-based morphometry
- WM, white matter
Collapse
|
40
|
Torgerson CM, Quinn C, Dinov I, Liu Z, Petrosyan P, Pelphrey K, Haselgrove C, Kennedy DN, Toga AW, Van Horn JD. Interacting with the National Database for Autism Research (NDAR) via the LONI Pipeline workflow environment. Brain Imaging Behav 2016; 9:89-103. [PMID: 25666423 DOI: 10.1007/s11682-015-9354-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Under the umbrella of the National Database for Clinical Trials (NDCT) related to mental illnesses, the National Database for Autism Research (NDAR) seeks to gather, curate, and make openly available neuroimaging data from NIH-funded studies of autism spectrum disorder (ASD). NDAR has recently made its database accessible through the LONI Pipeline workflow design and execution environment to enable large-scale analyses of cortical architecture and function via local, cluster, or "cloud"-based computing resources. This presents a unique opportunity to overcome many of the customary limitations to fostering biomedical neuroimaging as a science of discovery. Providing open access to primary neuroimaging data, workflow methods, and high-performance computing will increase uniformity in data collection protocols, encourage greater reliability of published data, results replication, and broaden the range of researchers now able to perform larger studies than ever before. To illustrate the use of NDAR and LONI Pipeline for performing several commonly performed neuroimaging processing steps and analyses, this paper presents example workflows useful for ASD neuroimaging researchers seeking to begin using this valuable combination of online data and computational resources. We discuss the utility of such database and workflow processing interactivity as a motivation for the sharing of additional primary data in ASD research and elsewhere.
Collapse
Affiliation(s)
- Carinna M Torgerson
- Laboratory of Neuro Imaging and The Institute for Neuroimaging and Informatics, Keck School of Medicine of USC, University of Southern California, 2001 North Soto Street - SSB1-Room 102, Los Angeles, CA, 90032, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Fan Q, Witzel T, Nummenmaa A, Van Dijk KRA, Van Horn JD, Drews MK, Somerville LH, Sheridan MA, Santillana RM, Snyder J, Hedden T, Shaw EE, Hollinshead MO, Renvall V, Zanzonico R, Keil B, Cauley S, Polimeni JR, Tisdall D, Buckner RL, Wedeen VJ, Wald LL, Toga AW, Rosen BR. MGH-USC Human Connectome Project datasets with ultra-high b-value diffusion MRI. Neuroimage 2016; 124:1108-1114. [PMID: 26364861 PMCID: PMC4651764 DOI: 10.1016/j.neuroimage.2015.08.075] [Citation(s) in RCA: 136] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Revised: 08/21/2015] [Accepted: 08/24/2015] [Indexed: 11/26/2022] Open
Abstract
The MGH-USC CONNECTOM MRI scanner housed at the Massachusetts General Hospital (MGH) is a major hardware innovation of the Human Connectome Project (HCP). The 3T CONNECTOM scanner is capable of producing a magnetic field gradient of up to 300 mT/m strength for in vivo human brain imaging, which greatly shortens the time spent on diffusion encoding, and decreases the signal loss due to T2 decay. To demonstrate the capability of the novel gradient system, data of healthy adult participants were acquired for this MGH-USC Adult Diffusion Dataset (N=35), minimally preprocessed, and shared through the Laboratory of Neuro Imaging Image Data Archive (LONI IDA) and the WU-Minn Connectome Database (ConnectomeDB). Another purpose of sharing the data is to facilitate methodological studies of diffusion MRI (dMRI) analyses utilizing high diffusion contrast, which perhaps is not easily feasible with standard MR gradient system. In addition, acquisition of the MGH-Harvard-USC Lifespan Dataset is currently underway to include 120 healthy participants ranging from 8 to 90 years old, which will also be shared through LONI IDA and ConnectomeDB. Here we describe the efforts of the MGH-USC HCP consortium in acquiring and sharing the ultra-high b-value diffusion MRI data and provide a report on data preprocessing and access. We conclude with a demonstration of the example data, along with results of standard diffusion analyses, including q-ball Orientation Distribution Function (ODF) reconstruction and tractography.
Collapse
Affiliation(s)
- Qiuyun Fan
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, 149 13th Street, Charlestown 02129, MA, USA
| | - Thomas Witzel
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, 149 13th Street, Charlestown 02129, MA, USA
| | - Aapo Nummenmaa
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, 149 13th Street, Charlestown 02129, MA, USA
| | - Koene R A Van Dijk
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, 149 13th Street, Charlestown 02129, MA, USA; Harvard University Department of Psychology, Center for Brain Science, Cambridge, MA, USA
| | - John D Van Horn
- Laboratory of Neuro Imaging, Institute for Neuroimaging and Informatics, Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Michelle K Drews
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, 149 13th Street, Charlestown 02129, MA, USA; Harvard University Department of Psychology, Center for Brain Science, Cambridge, MA, USA
| | - Leah H Somerville
- Harvard University Department of Psychology, Center for Brain Science, Cambridge, MA, USA
| | - Margaret A Sheridan
- Division of Developmental Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rosario M Santillana
- Division of Developmental Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jenna Snyder
- Division of Developmental Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Trey Hedden
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, 149 13th Street, Charlestown 02129, MA, USA
| | - Emily E Shaw
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, 149 13th Street, Charlestown 02129, MA, USA
| | - Marisa O Hollinshead
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, 149 13th Street, Charlestown 02129, MA, USA; Harvard University Department of Psychology, Center for Brain Science, Cambridge, MA, USA
| | - Ville Renvall
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, 149 13th Street, Charlestown 02129, MA, USA; Department of Neuroscience and Biomedical Engineering, Aalto University School of Science, Espoo, Finland
| | - Roberta Zanzonico
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, 149 13th Street, Charlestown 02129, MA, USA
| | - Boris Keil
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, 149 13th Street, Charlestown 02129, MA, USA
| | - Stephen Cauley
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, 149 13th Street, Charlestown 02129, MA, USA
| | - Jonathan R Polimeni
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, 149 13th Street, Charlestown 02129, MA, USA
| | - Dylan Tisdall
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, 149 13th Street, Charlestown 02129, MA, USA
| | - Randy L Buckner
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, 149 13th Street, Charlestown 02129, MA, USA; Harvard University Department of Psychology, Center for Brain Science, Cambridge, MA, USA
| | - Van J Wedeen
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, 149 13th Street, Charlestown 02129, MA, USA
| | - Lawrence L Wald
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, 149 13th Street, Charlestown 02129, MA, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Arthur W Toga
- Laboratory of Neuro Imaging, Institute for Neuroimaging and Informatics, Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Bruce R Rosen
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Harvard Medical School, Massachusetts General Hospital, 149 13th Street, Charlestown 02129, MA, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
42
|
Barrows RJ, Barsuglia J, Paholpak P, Eknoyan D, Sabodash V, Lee GJ, Mendez MF. Executive Abilities as Reflected by Clock Hand Placement: Frontotemporal Dementia Versus Early-Onset Alzheimer Disease. J Geriatr Psychiatry Neurol 2015; 28:239-48. [PMID: 26251109 DOI: 10.1177/0891988715598228] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The clock-drawing test (CDT) is widely used in clinical practice to diagnose and distinguish patients with dementia. It remains unclear, however, whether the CDT can distinguish among the early-onset dementias. Accordingly, we examined the ability of both quantitative and qualitative CDT analyses to distinguish behavioral variant frontotemporal dementia (bvFTD) and early-onset Alzheimer disease (eAD), the 2 most common neurodegenerative dementias with onset <65 years of age. We hypothesized that executive aspects of the CDT would discriminate between these 2 disorders. The study compared 15 patients with bvFTD and 16 patients with eAD on the CDT using 2 different scales and correlated the findings with neuropsychological testing and magnetic resonance imaging. The total CDT scores did not discriminate bvFTD and eAD; however, specific analysis of executive hand placement items successfully distinguished the groups, with eAD exhibiting greater errors than bvFTD. The performance on those executive hand placement items correlated with measures of naming as well as visuospatial and executive function. On tensor-based morphometry of the magnetic resonance images, executive hand placement correlated with right frontal volume. These findings suggest that lower performance on executive hand placement items occurs with involvement of the right dorsolateral frontal-parietal network for executive control in eAD, a network disproportionately affected in AD of early onset. Rather than the total performance on the clock task, the analysis of specific errors, such as executive hand placement, may be useful for early differentiation of eAD, bvFTD, and other conditions.
Collapse
Affiliation(s)
- Robin J Barrows
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA Greater Los Angeles VA Healthcare System, West Los Angeles, CA, USA
| | - Joseph Barsuglia
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA Greater Los Angeles VA Healthcare System, West Los Angeles, CA, USA
| | - Pongsatorn Paholpak
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA Greater Los Angeles VA Healthcare System, West Los Angeles, CA, USA Department of Psychiatry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Donald Eknoyan
- Greater Los Angeles VA Healthcare System, West Los Angeles, CA, USA Department of Psychiatry, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Valeriy Sabodash
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA Greater Los Angeles VA Healthcare System, West Los Angeles, CA, USA
| | - Grace J Lee
- Department of Psychology, School of Behavioral Health, Loma Linda University, Loma Linda, CA, USA
| | - Mario F Mendez
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA Greater Los Angeles VA Healthcare System, West Los Angeles, CA, USA
| |
Collapse
|
43
|
Toga AW, Foster I, Kesselman C, Madduri R, Chard K, Deutsch EW, Price ND, Glusman G, Heavner BD, Dinov ID, Ames J, Van Horn J, Kramer R, Hood L. Big biomedical data as the key resource for discovery science. J Am Med Inform Assoc 2015; 22:1126-31. [PMID: 26198305 PMCID: PMC5009918 DOI: 10.1093/jamia/ocv077] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 05/07/2015] [Accepted: 05/15/2015] [Indexed: 12/19/2022] Open
Abstract
Modern biomedical data collection is generating exponentially more data in a multitude of formats. This flood of complex data poses significant opportunities to discover and understand the critical interplay among such diverse domains as genomics, proteomics, metabolomics, and phenomics, including imaging, biometrics, and clinical data. The Big Data for Discovery Science Center is taking an "-ome to home" approach to discover linkages between these disparate data sources by mining existing databases of proteomic and genomic data, brain images, and clinical assessments. In support of this work, the authors developed new technological capabilities that make it easy for researchers to manage, aggregate, manipulate, integrate, and model large amounts of distributed data. Guided by biological domain expertise, the Center's computational resources and software will reveal relationships and patterns, aiding researchers in identifying biomarkers for the most confounding conditions and diseases, such as Parkinson's and Alzheimer's.
Collapse
Affiliation(s)
- Arthur W Toga
- Laboratory of Neuro Imaging, USC Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA
| | - Ian Foster
- Computation Institute, University of Chicago and Argonne National Laboratory, Chicago, IL, USA
| | - Carl Kesselman
- Information Sciences Institute, University of Southern California, Los Angeles, CA, USA
| | - Ravi Madduri
- Computation Institute, University of Chicago and Argonne National Laboratory, Chicago, IL, USA
| | - Kyle Chard
- Computation Institute, University of Chicago and Argonne National Laboratory, Chicago, IL, USA
| | | | | | | | | | - Ivo D Dinov
- Statistics Online Computational Resource (SOCR), UMSN, University of Michigan, Ann Arbor, MI, USA
| | - Joseph Ames
- Laboratory of Neuro Imaging, USC Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA
| | - John Van Horn
- Laboratory of Neuro Imaging, USC Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA
| | | | - Leroy Hood
- Institute for Systems Biology, Seattle, WA, USA
| |
Collapse
|
44
|
Moon SW, Dinov ID, Hobel S, Zamanyan A, Choi YC, Shi R, Thompson PM, Toga AW. Structural Brain Changes in Early-Onset Alzheimer's Disease Subjects Using the LONI Pipeline Environment. J Neuroimaging 2015; 25:728-37. [PMID: 25940587 PMCID: PMC4537660 DOI: 10.1111/jon.12252] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 03/20/2015] [Accepted: 03/22/2015] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND AND PURPOSE This study investigates 36 subjects aged 55-65 from the Alzheimer's Disease Neuroimaging Initiative (ADNI) database to expand our knowledge of early-onset (EO) Alzheimer's Disease (EO-AD) using neuroimaging biomarkers. METHODS Nine of the subjects had EO-AD, and 27 had EO mild cognitive impairment (EO-MCI). The structural ADNI data were parcellated using BrainParser, and the 15 most discriminating neuroimaging markers between the two cohorts were extracted using the Global Shape Analysis (GSA) Pipeline workflow. Then the Local Shape Analysis (LSA) Pipeline workflow was used to conduct local (per-vertex) post-hoc statistical analyses of the shape differences based on the participants' diagnoses (EO-MCI+EO-AD). Tensor-based Morphometry (TBM) and multivariate regression models were used to identify the significance of the structural brain differences based on the participants' diagnoses. RESULTS The significant between-group regional differences using GSA were found in 15 neuroimaging markers. The results of the LSA analysis workflow were based on the subject diagnosis, age, years of education, apolipoprotein E (ε4), Mini-Mental State Examination, visiting times, and logical memory as regressors. All the variables had significant effects on the regional shape measures. Some of these effects survived the false discovery rate (FDR) correction. Similarly, the TBM analysis showed significant effects on the Jacobian displacement vector fields, but these effects were reduced after FDR correction. CONCLUSIONS These results may explain some of the differences between EO-AD and EO-MCI, and some of the characteristics of the EO cognitive impairment subjects.
Collapse
Affiliation(s)
- Seok Woo Moon
- Department of Psychiatry, Konkuk University School of Medicine, Seoul 143-701, Korea
| | - Ivo D. Dinov
- Laboratory of Neuro Imaging, Institute for Neuroimaging and Informatics, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA 90032, USA
- University of Michigan, School of Nursing, Ann Arbor, MI 48109, USA
| | - Sam Hobel
- Laboratory of Neuro Imaging, Institute for Neuroimaging and Informatics, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA 90032, USA
| | - Alen Zamanyan
- Laboratory of Neuro Imaging, Institute for Neuroimaging and Informatics, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA 90032, USA
| | - Young Chil Choi
- Department of Radiology, Konkuk University School of Medicine, Seoul 143-701, Korea
| | - Ran Shi
- Laboratory of Neuro Imaging, Institute for Neuroimaging and Informatics, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA 90032, USA
| | - Paul M. Thompson
- Laboratory of Neuro Imaging, Institute for Neuroimaging and Informatics, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA 90032, USA
| | - Arthur W. Toga
- Laboratory of Neuro Imaging, Institute for Neuroimaging and Informatics, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA 90032, USA
| | | |
Collapse
|
45
|
Masters M, Bruner E, Queer S, Traynor S, Senjem J. Analysis of the volumetric relationship among human ocular, orbital and fronto-occipital cortical morphology. J Anat 2015; 227:460-73. [PMID: 26250048 DOI: 10.1111/joa.12364] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2015] [Indexed: 11/29/2022] Open
Abstract
Recent research on the visual system has focused on investigating the relationship among eye (ocular), orbital, and visual cortical anatomy in humans. This issue is relevant in evolutionary and medical fields. In terms of evolution, only in modern humans and Neandertals are the orbits positioned beneath the frontal lobes, with consequent structural constraints. In terms of medicine, such constraints can be associated with minor deformation of the eye, vision defects, and patterns of integration among these features, and in association with the frontal lobes, are important to consider in reconstructive surgery. Further study is therefore necessary to establish how these variables are related, and to what extent ocular size is associated with orbital and cerebral cortical volumes. Relationships among these anatomical components were investigated using magnetic resonance images from a large sample of 83 individuals, which also included each subject's body height, age, sex, and uncorrected visual acuity score. Occipital and frontal gyri volumes were calculated using two different cortical parcellation tools in order to provide a better understanding of how the eye and orbit vary in relation to visual cortical gyri, and frontal cortical gyri which are not directly related to visual processing. Results indicated that ocular and orbital volumes were weakly correlated, and that eye volume explains only a small proportion of the variance in orbital volume. Ocular and orbital volumes were also found to be equally and, in most cases, more highly correlated with five frontal lobe gyri than with occipital lobe gyri associated with V1, V2, and V3 of the visual cortex. Additionally, after accounting for age and sex variation, the relationship between ocular and total visual cortical volume was no longer statistically significant, but remained significantly related to total frontal lobe volume. The relationship between orbital and visual cortical volumes remained significant for a number of occipital lobe gyri even after accounting for these cofactors, but was again found to be more highly correlated with the frontal cortex than with the occipital cortex. These results indicate that eye volume explains only a small amount of variation in orbital and visual cortical volume, and that the eye and orbit are generally more structurally associated with the frontal lobes than they are functionally associated with the visual cortex of the occipital lobes. Results also demonstrate that these components of the visual system are highly complex and influenced by a multitude of factors in humans.
Collapse
Affiliation(s)
| | - Emiliano Bruner
- Centro Nacional de Investigación sobre la Evolución Humana, Burgos, Spain
| | | | | | - Jess Senjem
- University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
46
|
Kini LG, Davis KA, Wagenaar JB. Data integration: Combined imaging and electrophysiology data in the cloud. Neuroimage 2015; 124:1175-1181. [PMID: 26044858 DOI: 10.1016/j.neuroimage.2015.05.075] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 04/29/2015] [Accepted: 05/26/2015] [Indexed: 11/30/2022] Open
Abstract
There has been an increasing effort to correlate electrophysiology data with imaging in patients with refractory epilepsy over recent years. IEEG.org provides a free-access, rapidly growing archive of imaging data combined with electrophysiology data and patient metadata. It currently contains over 1200 human and animal datasets, with multiple data modalities associated with each dataset (neuroimaging, EEG, EKG, de-identified clinical and experimental data, etc.). The platform is developed around the concept that scientific data sharing requires a flexible platform that allows sharing of data from multiple file formats. IEEG.org provides high- and low-level access to the data in addition to providing an environment in which domain experts can find, visualize, and analyze data in an intuitive manner. Here, we present a summary of the current infrastructure of the platform, available datasets and goals for the near future.
Collapse
Affiliation(s)
- Lohith G Kini
- Department of Bioengineering, University of Pennsylvania, 240 Skirkanich Hall, 210 South 33rd Street, Philadelphia, PA 19104-6321, USA.
| | - Kathryn A Davis
- Department of Neurology, Hospital of the University of Pennsylvania, 3400 Spruce Street, 3 West Gates Bldg, Philadelphia PA 19104, USA.
| | - Joost B Wagenaar
- Department of Neurology, Hospital of the University of Pennsylvania, 3400 Spruce Street, 3 West Gates Bldg, Philadelphia PA 19104, USA.
| |
Collapse
|
47
|
Cui Z, Zhao C, Gong G. Parallel workflow tools to facilitate human brain MRI post-processing. Front Neurosci 2015; 9:171. [PMID: 26029043 PMCID: PMC4429548 DOI: 10.3389/fnins.2015.00171] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 04/27/2015] [Indexed: 02/04/2023] Open
Abstract
Multi-modal magnetic resonance imaging (MRI) techniques are widely applied in human brain studies. To obtain specific brain measures of interest from MRI datasets, a number of complex image post-processing steps are typically required. Parallel workflow tools have recently been developed, concatenating individual processing steps and enabling fully automated processing of raw MRI data to obtain the final results. These workflow tools are also designed to make optimal use of available computational resources and to support the parallel processing of different subjects or of independent processing steps for a single subject. Automated, parallel MRI post-processing tools can greatly facilitate relevant brain investigations and are being increasingly applied. In this review, we briefly summarize these parallel workflow tools and discuss relevant issues.
Collapse
Affiliation(s)
- Zaixu Cui
- State Key Laboratory of Cognitive Neuroscience and Learning and IDG/McGovern Institute for Brain Research, Beijing Normal University Beijing, China
| | - Chenxi Zhao
- State Key Laboratory of Cognitive Neuroscience and Learning and IDG/McGovern Institute for Brain Research, Beijing Normal University Beijing, China
| | - Gaolang Gong
- State Key Laboratory of Cognitive Neuroscience and Learning and IDG/McGovern Institute for Brain Research, Beijing Normal University Beijing, China
| |
Collapse
|
48
|
Craddock RC, Tungaraza RL, Milham MP. Connectomics and new approaches for analyzing human brain functional connectivity. Gigascience 2015; 4:13. [PMID: 25810900 PMCID: PMC4373299 DOI: 10.1186/s13742-015-0045-x] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 01/18/2015] [Indexed: 11/10/2022] Open
Abstract
Estimating the functional interactions between brain regions and mapping those connections to corresponding inter-individual differences in cognitive, behavioral and psychiatric domains are central pursuits for understanding the human connectome. The number and complexity of functional interactions within the connectome and the large amounts of data required to study them position functional connectivity research as a “big data” problem. Maximizing the degree to which knowledge about human brain function can be extracted from the connectome will require developing a new generation of neuroimaging analysis algorithms and tools. This review describes several outstanding problems in brain functional connectomics with the goal of engaging researchers from a broad spectrum of data sciences to help solve these problems. Additionally it provides information about open science resources consisting of raw and preprocessed data to help interested researchers get started.
Collapse
Affiliation(s)
- R Cameron Craddock
- Center for Biomedical Imaging and Neuromodulation, Nathan Kline Institute for Psychiatric Research, 140 Old Orangeburg Rd, Orangeburg, 10962 New York USA ; Center for the Developing Brain, Child Mind Institute, 445 Park Ave, New York, 10022 New York USA
| | - Rosalia L Tungaraza
- Center for Biomedical Imaging and Neuromodulation, Nathan Kline Institute for Psychiatric Research, 140 Old Orangeburg Rd, Orangeburg, 10962 New York USA
| | - Michael P Milham
- Center for Biomedical Imaging and Neuromodulation, Nathan Kline Institute for Psychiatric Research, 140 Old Orangeburg Rd, Orangeburg, 10962 New York USA ; Center for the Developing Brain, Child Mind Institute, 445 Park Ave, New York, 10022 New York USA
| |
Collapse
|
49
|
Torgerson CM, Irimia A, Goh SYM, Van Horn JD. The DTI connectivity of the human claustrum. Hum Brain Mapp 2015; 36:827-38. [PMID: 25339630 PMCID: PMC4324054 DOI: 10.1002/hbm.22667] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 09/29/2014] [Accepted: 10/13/2014] [Indexed: 01/18/2023] Open
Abstract
The origin, structure, and function of the claustrum, as well as its role in neural computation, have remained a mystery since its discovery in the 17th century. Assessing the in vivo connectivity of the claustrum may bring forth useful insights with relevance to model the overall functionality of the claustrum itself. Using structural and diffusion tensor neuroimaging in N = 100 healthy subjects, we found that the claustrum has the highest connectivity in the brain by regional volume. Network theoretical analyses revealed that (a) the claustrum is a primary contributor to global brain network architecture, and that (b) significant connectivity dependencies exist between the claustrum, frontal lobe, and cingulate regions. These results illustrate that the claustrum is ideally located within the human central nervous system (CNS) connectome to serve as the putative "gate keeper" of neural information for consciousness awareness. Our findings support and underscore prior theoretical contributions about the involvement of the claustrum in higher cognitive function and its relevance in devastating neurological disease.
Collapse
Affiliation(s)
- Carinna M. Torgerson
- The Institute for Neuroimaging and Informatics (INI) and Laboratory of Neuro Imaging [LONI]Keck School of Medicine of USC, University of Southern CaliforniaLos AngelesCalifornia
| | - Andrei Irimia
- The Institute for Neuroimaging and Informatics (INI) and Laboratory of Neuro Imaging [LONI]Keck School of Medicine of USC, University of Southern CaliforniaLos AngelesCalifornia
| | - S. Y. Matthew Goh
- The Institute for Neuroimaging and Informatics (INI) and Laboratory of Neuro Imaging [LONI]Keck School of Medicine of USC, University of Southern CaliforniaLos AngelesCalifornia
| | - John Darrell Van Horn
- The Institute for Neuroimaging and Informatics (INI) and Laboratory of Neuro Imaging [LONI]Keck School of Medicine of USC, University of Southern CaliforniaLos AngelesCalifornia
| |
Collapse
|
50
|
Sivagnanam S, Majumdar A, Yoshimoto K, Astakhov V, Bandrowski A, Martone M, Carnevale NT. Early experiences in developing and managing the neuroscience gateway. CONCURRENCY AND COMPUTATION : PRACTICE & EXPERIENCE 2015; 27:473-488. [PMID: 26523124 PMCID: PMC4624199 DOI: 10.1002/cpe.3283] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The last few decades have seen the emergence of computational neuroscience as a mature field where researchers are interested in modeling complex and large neuronal systems and require access to high performance computing machines and associated cyber infrastructure to manage computational workflow and data. The neuronal simulation tools, used in this research field, are also implemented for parallel computers and suitable for high performance computing machines. But using these tools on complex high performance computing machines remains a challenge because of issues with acquiring computer time on these machines located at national supercomputer centers, dealing with complex user interface of these machines, dealing with data management and retrieval. The Neuroscience Gateway is being developed to alleviate and/or hide these barriers to entry for computational neuroscientists. It hides or eliminates, from the point of view of the users, all the administrative and technical barriers and makes parallel neuronal simulation tools easily available and accessible on complex high performance computing machines. It handles the running of jobs and data management and retrieval. This paper shares the early experiences in bringing up this gateway and describes the software architecture it is based on, how it is implemented, and how users can use this for computational neuroscience research using high performance computing at the back end. We also look at parallel scaling of some publicly available neuronal models and analyze the recent usage data of the neuroscience gateway.
Collapse
Affiliation(s)
| | - Amit Majumdar
- University of California San Diego, La Jolla, CA, USA
| | | | | | | | | | | |
Collapse
|