of parent grant, UK-ADC) This is a proposal for a University of Kentucky Alzheimer?s Disease Research Center (UK-ADRC) neuroimaging supplement. This purpose of the supplement is to provide neuroimaging data on the UK-ADRC?s deeply phenotyped clinical cohort to the NIH?s recently funded initiative, Standardized Centralized Alzheimer's and Related Dementias Neuroimaging (SCAN). In addition, a secondary purpose of this supplement is to identify neuroimaging correlates of two AD-mimics: limbic-predominant age-related TDP-43 encephalopathy (LATE) and primary age-related tauopathy (PART). The UK-ADRC is an experienced and collaborative ADRC originally funded in 1985. Our principal mission is to serve as the focal point for all AD-related activities at UK and this region of the United States, by providing an environment and core resources that catalyze innovative research, outreach, education, and clinical programs. Our signature resources include: 1) a cognitively normal group of ~500 subjects followed longitudinally, with all committed to brain autopsy upon death; 2) a strong program in clinical-neuropathological correlations and short postmortem interval autopsies; 3) an established program studying preclinical biomarkers of AD using neuroimaging and biofluids; 4) an integrated centralized database and innovative biostatistical expertise to characterize clinical and biological transitions; and 5) a successful and close partnership with the African-American community and increased participation of underrepresented individuals in our longitudinal cohort and ADRC-affiliated research studies and clinical trials. The overall scientific emphasis of the UK-ADRC continues to be on our interrelated themes: Transitions & Translation. Our well-characterized, longitudinal cohort and historically strong neuropathology program focused on normal aging, preclinical disease states and early cognitive transitions have been central to our success in defining early pathogenic mechanisms underlying the transitions from normal cognitive aging to impairment. The depth of expertise and collaborative nature of our investigators have also resulted in substantial progress on translation of that knowledge into new targets and novel therapeutic strategies. The UK-ADRC provides an infrastructure and environment that focuses on these integrated themes and advances AD research, education, outreach, and clinical programs through highly interactive and effective components: Administrative Core, Clinical Core, Data Management and Statistics Core, Neuropathology Core, Outreach and Recruitment Core, Biomarker Core, and Research Education Component. The UK-ADRC will make resources from these Cores available to support the success of this ADRC neuroimaging supplement.

Public Health Relevance

This supplement will fill a critical gap in the field by focusing on the development of neuroimaging biomarkers of two common AD mimics: LATE and PART. By imaging subjects who are A? negative and without significant cerebrovascular disease, we will identify MRI patterns characteristic of these AD mimics. We will share the UK SCAN neuroimaging data, combined with our deep clinical, biofluid, and genetic phenotyping of these longitudinally followed subjects, to stimulate additional research on AD mimics across the ADRC network.

Agency
National Institute of Health (NIH)
Institute
National Institute on Aging (NIA)
Type
Center Core Grants (P30)
Project #
3P30AG028383-15S1
Application #
10170755
Study Section
Special Emphasis Panel (ZAG1)
Program Officer
Elliott, Cerise
Project Start
2006-07-15
Project End
2021-06-30
Budget Start
2020-09-01
Budget End
2021-06-30
Support Year
15
Fiscal Year
2020
Total Cost
Indirect Cost
Name
University of Kentucky
Department
Pathology
Type
Schools of Medicine
DUNS #
939017877
City
Lexington
State
KY
Country
United States
Zip Code
40526
Barnes, Josephine; Bartlett, Jonathan W; Wolk, David A et al. (2018) Disease Course Varies According to Age and Symptom Length in Alzheimer's Disease. J Alzheimers Dis 64:631-642
Bradley-Whitman, Melissa A; Roberts, Kelly N; Abner, Erin L et al. (2018) A novel method for the rapid detection of post-translationally modified visinin-like protein 1 in rat models of brain injury. Brain Inj 32:363-380
Brown, Christopher A; Jiang, Yang; Smith, Charles D et al. (2018) Age and Alzheimer's pathology disrupt default mode network functioning via alterations in white matter microstructure but not hyperintensities. Cortex 104:58-74
Lin, Ming; Gong, Pinghua; Yang, Tao et al. (2018) Big Data Analytical Approaches to the NACC Dataset: Aiding Preclinical Trial Enrichment. Alzheimer Dis Assoc Disord 32:18-27
Burke, Shanna L; Maramaldi, Peter; Cadet, Tamara et al. (2018) Decreasing hazards of Alzheimer's disease with the use of antidepressants: mitigating the risk of depression and apolipoprotein E. Int J Geriatr Psychiatry 33:200-211
Kirson, Noam Y; Scott Andrews, J; Desai, Urvi et al. (2018) Patient Characteristics and Outcomes Associated with Receiving an Earlier Versus Later Diagnosis of Probable Alzheimer's Disease. J Alzheimers Dis 61:295-307
Nelson, Peter T; Abner, Erin L; Patel, Ela et al. (2018) The Amygdala as a Locus of Pathologic Misfolding in Neurodegenerative Diseases. J Neuropathol Exp Neurol 77:2-20
Lanzillotta, Chiara; Tramutola, Antonella; Meier, Shelby et al. (2018) Early and Selective Activation and Subsequent Alterations to the Unfolded Protein Response in Down Syndrome Mouse Models. J Alzheimers Dis 62:347-359
Gal, Jozsef; Chen, Jing; Katsumata, Yuriko et al. (2018) Detergent Insoluble Proteins and Inclusion Body-Like Structures Immunoreactive for PRKDC/DNA-PK/DNA-PKcs, FTL, NNT, and AIFM1 in the Amygdala of Cognitively Impaired Elderly Persons. J Neuropathol Exp Neurol 77:21-39
Goetzl, Edward J; Abner, Erin L; Jicha, Gregory A et al. (2018) Declining levels of functionally specialized synaptic proteins in plasma neuronal exosomes with progression of Alzheimer's disease. FASEB J 32:888-893

Showing the most recent 10 out of 471 publications