Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Patricia Aldea is active.

Publication


Featured researches published by Patricia Aldea.


Embo Molecular Medicine | 2009

Cerebrospinal fluid tau and ptau181 increase with cortical amyloid deposition in cognitively normal individuals: Implications for future clinical trials of Alzheimer's disease

Anne M. Fagan; Mark A. Mintun; Aarti R. Shah; Patricia Aldea; Catherine M. Roe; Robert H. Mach; Daniel S. Marcus; John C. Morris; David M. Holtzman

Alzheimers disease (AD) pathology is estimated to develop many years before detectable cognitive decline. Fluid and imaging biomarkers may identify people in early symptomatic and even preclinical stages, possibly when potential treatments can best preserve cognitive function. We previously reported that cerebrospinal fluid (CSF) levels of amyloid‐β42 (Aβ42) serve as an excellent marker for brain amyloid as detected by the amyloid tracer, Pittsburgh compound B (PIB). Using data from 189 cognitively normal participants, we now report a positive linear relationship between CSF tau/ptau181 (primary constituents of neurofibrillary tangles) with the amount of cortical amyloid. We observe a strong inverse relationship of cortical PIB binding with CSF Aβ42 but not for plasma Aβ species. Some individuals have low CSF Aβ42 but no cortical PIB binding. Together, these data suggest that changes in brain Aβ42 metabolism and amyloid formation are early pathogenic events in AD, and that significant disruptions in CSF tau metabolism likely occur after Aβ42 initially aggregates and increases as amyloid accumulates. These findings have important implications for preclinical AD diagnosis and treatment.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Regional variability of imaging biomarkers in autosomal dominant Alzheimer’s disease

Tammie L.S. Benzinger; Tyler Blazey; Clifford R. Jack; Robert A. Koeppe; Yi Su; Chengjie Xiong; Marcus E. Raichle; Abraham Z. Snyder; Beau M. Ances; Randall J. Bateman; Nigel J. Cairns; Anne M. Fagan; Alison Goate; Daniel S. Marcus; Paul S. Aisen; Jon Christensen; Lindsay Ercole; Russ C. Hornbeck; Angela M. Farrar; Patricia Aldea; Mateusz S. Jasielec; Christopher J. Owen; Xianyun Xie; Richard Mayeux; Adam M. Brickman; Eric McDade; William E. Klunk; Chester A. Mathis; John M. Ringman; Paul M. Thompson

Significance Beta-amyloid plaque accumulation, glucose hypometabolism, and neuronal atrophy are hallmarks of Alzheimer’s disease. However, the regional ordering of these biomarkers prior to dementia remains untested. In a cohort with Alzheimer’s disease mutations, we performed an integrated whole-brain analysis of three major imaging techniques: amyloid PET, [18F]fluro-deoxyglucose PET, and structural MRI. We found that most gray-matter structures with amyloid plaques later have hypometabolism followed by atrophy. Critically, however, not all regions lose metabolic function, and not all regions atrophy, even when there is significant amyloid deposition. These regional disparities have important implications for clinical trials of disease-modifying therapies. Major imaging biomarkers of Alzheimer’s disease include amyloid deposition [imaged with [11C]Pittsburgh compound B (PiB) PET], altered glucose metabolism (imaged with [18F]fluro-deoxyglucose PET), and structural atrophy (imaged by MRI). Recently we published the initial subset of imaging findings for specific regions in a cohort of individuals with autosomal dominant Alzheimer’s disease. We now extend this work to include a larger cohort, whole-brain analyses integrating all three imaging modalities, and longitudinal data to examine regional differences in imaging biomarker dynamics. The anatomical distribution of imaging biomarkers is described in relation to estimated years from symptom onset. Autosomal dominant Alzheimer’s disease mutation carrier individuals have elevated PiB levels in nearly every cortical region 15 y before the estimated age of onset. Reduced cortical glucose metabolism and cortical thinning in the medial and lateral parietal lobe appeared 10 and 5 y, respectively, before estimated age of onset. Importantly, however, a divergent pattern was observed subcortically. All subcortical gray-matter regions exhibited elevated PiB uptake, but despite this, only the hippocampus showed reduced glucose metabolism. Similarly, atrophy was not observed in the caudate and pallidum despite marked amyloid accumulation. Finally, before hypometabolism, a hypermetabolic phase was identified for some cortical regions, including the precuneus and posterior cingulate. Additional analyses of individuals in which longitudinal data were available suggested that an accelerated appearance of volumetric declines approximately coincides with the onset of the symptomatic phase of the disease.


Science Translational Medicine | 2016

Tau and Aβ imaging, CSF measures, and cognition in Alzheimer’s disease

Matthew R. Brier; Brian A. Gordon; Karl A. Friedrichsen; John E. McCarthy; Ari Stern; Jon Christensen; Christopher J. Owen; Patricia Aldea; Yi Su; Jason Hassenstab; Nigel J. Cairns; David M. Holtzman; Anne M. Fagan; John C. Morris; Tammie L.S. Benzinger; Beau M. Ances

PET imaging of pathological tau correlates more closely with Alzheimer’s disease–related cognitive impairment than does imaging of β-amyloid. A window into Alzheimer’s disease Alzheimer’s disease is pathologically defined by the accumulation of β-amyloid (Aβ) plaques and tau tangles. The cognitive and pathological correlates of Aβ deposition have been well studied owing to the availability of PET imaging ligands. Using newly available tau imaging agents, Brier et al. now explore relationships among tau pathology and Aβ with PET imaging, cerebrospinal fluid measures of disease, and cognition. Overall, tau imaging provided a more robust predictor of disease status than did Aβ imaging. Thus, whereas Aβ imaging provides a good marker for early disease state, tau imaging is a more robust predictor of disease progression. Alzheimer’s disease (AD) is characterized by two molecular pathologies: cerebral β-amyloidosis in the form of β-amyloid (Aβ) plaques and tauopathy in the form of neurofibrillary tangles, neuritic plaques, and neuropil threads. Until recently, only Aβ could be studied in humans using positron emission tomography (PET) imaging owing to a lack of tau PET imaging agents. Clinical pathological studies have linked tau pathology closely to the onset and progression of cognitive symptoms in patients with AD. We report PET imaging of tau and Aβ in a cohort of cognitively normal older adults and those with mild AD. Multivariate analyses identified unique disease-related stereotypical spatial patterns (topographies) for deposition of tau and Aβ. These PET imaging tau and Aβ topographies were spatially distinct but correlated with disease progression. Cerebrospinal fluid measures of tau, often used to stage preclinical AD, correlated with tau deposition in the temporal lobe. Tau deposition in the temporal lobe more closely tracked dementia status and was a better predictor of cognitive performance than Aβ deposition in any region of the brain. These data support models of AD where tau pathology closely tracks changes in brain function that are responsible for the onset of early symptoms in AD.


JAMA Neurology | 2016

Evaluation of Tau Imaging in Staging Alzheimer Disease and Revealing Interactions Between β-Amyloid and Tauopathy

Liang Wang; Tammie L.S. Benzinger; Yi Su; Jon Christensen; Karl A. Friedrichsen; Patricia Aldea; Jonathan McConathy; Nigel J. Cairns; Anne M. Fagan; John C. Morris; Beau M. Ances

IMPORTANCE In vivo tau imaging may become a diagnostic marker for Alzheimer disease (AD) and provides insights into the pathophysiology of AD. OBJECTIVE To evaluate the usefulness of [18F]-AV-1451 positron emission tomography (PET) imaging to stage AD and assess the associations among β-amyloid (Aβ), tau, and volume loss. DESIGN, SETTING, AND PARTICIPANTS An imaging study conducted at Knight Alzheimer Disease Research Center at Washington University in St Louis, Missouri. A total of 59 participants who were cognitively normal (CN) (Clinical Dementia Rating [CDR] score, 0) or had AD dementia (CDR score, >0) were included. MAIN OUTCOMES AND MEASURES Standardized uptake value ratio (SUVR) of [18F]-AV-1451 in the hippocampus and a priori-defined AD cortical signature regions, cerebrospinal fluid Aβ42, hippocampal volume, and AD signature cortical thickness. RESULTS Of the 59 participants, 38 (64%) were male; mean (SD) age was 74 (6) years. The [18F]-AV-1451 SUVR in the hippocampus and AD cortical signature regions distinguished AD from CN participants (area under the receiver operating characteristic curve range [95% CI], 0.89 [0.73-1.00] to 0.98 [0.92-1.00]). An [18F]-AV-1451 SUVR cutoff value of 1.19 (sensitivity, 100%; specificity, 86%) from AD cortical signature regions best separated cerebrospinal fluid Aβ42-positive (Aβ+) AD from cerebrospinal fluid Aβ42-negative (Aβ-) CN participants. This same cutoff also divided Aβ+ CN participants into low vs high tau groups. Moreover, the presence of Aβ+ was associated with an elevated [18F]-AV-1451 SUVR in AD cortical signature regions (Aβ+ participants: mean [SD], 1.3 [0.3]; Aβ- participants: 1.1 [0.1]; F = 4.3, P = .04) but not in the hippocampus. The presence of Aβ+ alone was not related to hippocampal volume or AD signature cortical thickness. An elevated [18F]-AV-1451 SUVR was associated with volumetric loss in both the hippocampus and AD cortical signature regions. The observed [18F]-AV-1451 SUVR volumetric association was modified by Aβ status in the hippocampus but not in AD cortical signature regions. An inverse association between hippocampal [18F]-AV-1451 SUVR and volume was seen in Aβ+ participants (R2 = 0.55; P < .001) but not Aβ- (R2 = 0; P = .97) participants. CONCLUSIONS AND RELEVANCE Use of [18F]-AV-1451 has a potential for staging of the preclinical and clinical phases of AD. β-Amyloid interacts with hippocampal and cortical tauopathy to affect neurodegeneration. In the absence of Aβ, hippocampal tau deposition may be insufficient for the neurodegenerative process that leads to AD.


NeuroImage | 2015

Partial volume correction in quantitative amyloid imaging.

Yi Su; Tyler Blazey; Abraham Z. Snyder; Marcus E. Raichle; Daniel S. Marcus; Beau M. Ances; Randall J. Bateman; Nigel J. Cairns; Patricia Aldea; Lisa Cash; Jon Christensen; Karl A. Friedrichsen; Russ C. Hornbeck; Angela M. Farrar; Christopher J. Owen; Richard Mayeux; Adam M. Brickman; William E. Klunk; Julie C. Price; Paul M. Thompson; Bernardino Ghetti; Andrew J. Saykin; Reisa A. Sperling; Keith Johnson; Peter R. Schofield; Virginia Buckles; John C. Morris; Tammie L.S. Benzinger

Amyloid imaging is a valuable tool for research and diagnosis in dementing disorders. As positron emission tomography (PET) scanners have limited spatial resolution, measured signals are distorted by partial volume effects. Various techniques have been proposed for correcting partial volume effects, but there is no consensus as to whether these techniques are necessary in amyloid imaging, and, if so, how they should be implemented. We evaluated a two-component partial volume correction technique and a regional spread function technique using both simulated and human Pittsburgh compound B (PiB) PET imaging data. Both correction techniques compensated for partial volume effects and yielded improved detection of subtle changes in PiB retention. However, the regional spread function technique was more accurate in application to simulated data. Because PiB retention estimates depend on the correction technique, standardization is necessary to compare results across groups. Partial volume correction has sometimes been avoided because it increases the sensitivity to inaccuracy in image registration and segmentation. However, our results indicate that appropriate PVC may enhance our ability to detect changes in amyloid deposition.


Brain | 2016

The relationship between cerebrospinal fluid markers of Alzheimer pathology and positron emission tomography tau imaging

Brian A. Gordon; Karl A. Friedrichsen; Matthew R. Brier; Tyler Blazey; Yi Su; Jon Christensen; Patricia Aldea; Jonathan McConathy; David M. Holtzman; Nigel J. Cairns; John C. Morris; Anne M. Fagan; Beau M. Ances; Tammie L.S. Benzinger

The two primary molecular pathologies in Alzheimers disease are amyloid-β plaques and tau-immunoreactive neurofibrillary tangles. Investigations into these pathologies have been restricted to cerebrospinal fluid assays, and positron emission tomography tracers that can image amyloid-β plaques. Tau tracers have recently been introduced into the field, although the utility of the tracer and its relationship to other Alzheimer biomarkers are still unknown. Here we examined tau deposition in 41 cognitively normal and 11 cognitively impaired older adults using the radioactive tau ligand (18)F-AV-1451 (previously known as T807) who also underwent a lumbar puncture to assess cerebrospinal fluid levels of total tau (t-tau), phosphorylated tau181 (p-tau181) and amyloid-β42 Voxel-wise statistical analyses examined spatial patterns of tau deposition associated with cognitive impairment. We then related the amount of tau tracer uptake to levels of cerebrospinal fluid biomarkers. All analyses controlled for age and gender and, when appropriate, the time between imaging and lumbar puncture assessments. Symptomatic individuals (Clinical Dementia Rating > 0) demonstrated markedly increased levels of tau tracer uptake. This elevation was most prominent in the temporal lobe and temporoparietal junction, but extended more broadly into parietal and frontal cortices. In the entire cohort, there were significant relationships among all cerebrospinal fluid biomarkers and tracer uptake, notably for tau-related cerebrospinal fluid markers. After controlling for levels of amyloid-β42, the correlations with tau uptake were r = 0.490 (P < 0.001) for t-tau and r = 0.492 (P < 0.001) for p-tau181 Within the cognitively normal cohort, levels of amyloid-β42, but not t-tau or p-tau181, were associated with elevated tracer binding that was confined primarily to the medial temporal lobe and adjacent neocortical regions. AV-1451 tau binding in the medial temporal, parietal, and frontal cortices is correlated with tau-related cerebrospinal fluid measures. In preclinical Alzheimers disease, there is focal tauopathy in the medial temporal lobes and adjacent cortices.


PLOS ONE | 2015

Quantitative amyloid imaging using image-derived arterial input function.

Yi Su; Tyler Blazey; Abraham Z. Snyder; Marcus E. Raichle; Russ C. Hornbeck; Patricia Aldea; John C. Morris; Tammie L.S. Benzinger

Amyloid PET imaging is an indispensable tool widely used in the investigation, diagnosis and monitoring of Alzheimer’s disease (AD). Currently, a reference region based approach is used as the mainstream quantification technique for amyloid imaging. This approach assumes the reference region is amyloid free and has the same tracer influx and washout kinetics as the regions of interest. However, this assumption may not always be valid. The goal of this work is to evaluate an amyloid imaging quantification technique that uses arterial region of interest as the reference to avoid potential bias caused by specific binding in the reference region. 21 participants, age 58 and up, underwent Pittsburgh compound B (PiB) PET imaging and MR imaging including a time-of-flight (TOF) MR angiography (MRA) scan and a structural scan. FreeSurfer based regional analysis was performed to quantify PiB PET data. Arterial input function was estimated based on coregistered TOF MRA using a modeling based technique. Regional distribution volume (VT) was calculated using Logan graphical analysis with estimated arterial input function. Kinetic modeling was also performed using the estimated arterial input function as a way to evaluate PiB binding (DVRkinetic) without a reference region. As a comparison, Logan graphical analysis was also performed with cerebellar cortex as reference to obtain DVRREF. Excellent agreement was observed between the two distribution volume ratio measurements (r>0.89, ICC>0.80). The estimated cerebellum VT was in line with literature reported values and the variability of cerebellum VT in the control group was comparable to reported variability using arterial sampling data. This study suggests that image-based arterial input function is a viable approach to quantify amyloid imaging data, without the need of arterial sampling or a reference region. This technique can be a valuable tool for amyloid imaging, particularly in population where reference normalization may not be accurate.


Alzheimers & Dementia | 2016

SIMILARITIES AND DIFFERENCES IN PATTERNS OF [F18]-AV-1451 AND [F18]-FDG IN FRONTOTEMPORAL DEMENTIA

Karl A. Friedrichsen; Nupur Ghoshal; Nelly Joseph-Mathurin; Shruti Mishra; Yi Su; Jon Christensen; Patricia Aldea; Jonathan McConathy; Brian A. Gordon; Beau M. Ances; Nigel J. Cairns; John C. Morris; Tammie L.S. Benzinger

cohort.Methods:18 participants were included. 80-100minute [F] AV-1451 images were normalized to SUVR units by cerebellar grey matter. Measures from antecedent cross-sectional [F]FDG-PET, baseline [F]Florbetapir PET and MRI that were approximately 3yrs before the [F]AV-1451 scan (mean 1⁄4 3.2yrs; range 1⁄4 2.05.1yrs), and two-year change in florbetapir and MRI measures were explored as independent predictors of [F]AV-1451 SUVR values extracted from the whole cortex and the medial temporal lobe (MTL). [F]Florbetapir and []FDG scans were pre-processed as described [1]. [F]Florbetapir scans were intensity normalized by the whole cerebellum (baseline) and a composite of the cerebral white matter, brainstem, and whole cerebellum (longitudinal). [F]FDG intensity was normalized to the pons. Baseline global cortical [F]Florbetapir and mean parietal [F]FDG SUVR, as well as 2yr annualized percent change (APC) in global [F]Florbetapir were extracted. Baseline and 2-year hippocampal volumes (HV) were estimated using Freesurfer v5.1. Step-wise linear models predicting cortical or MTL [F]AV-1451 SUVR with the following predictors were assessed: cortical [F]Florbetapir SUVR, annual percent change (APC) in cortical [F]Florbetapir SUVR, mean parietal [F]FDG SUVR, HV, APC in HV, diagnosis (control or MCI/AD), age, gender, and time between scans. Results:Mean cortical [F]AV-1451 uptake was predicted only by diagnosis and cross-sectional mean cortical [F]Florbetapir SUVR. However, MTL [F]AV-1451 uptake was predicted by both mean cross-sectional cortical [F]Florbetapir uptake and cross-sectional HV. Conclusions: Overall, [F]AV-1451 uptake (representative of tau deposition) was predicted by the extent of cortical amyloid deposition ([F]Florbetapir uptake) approximately three years prior. Hippocampal atrophy may interact with this process. [1] Risacher et al. (2015) Alzheimer’s & Dementia, (11): 1417.


Alzheimers & Dementia | 2016

RELATING PET AND CSF MEASURE OF TAU PATHOLOGY

Brian A. Gordon; Karl A. Friedrichsen; Matthew R. Brier; Tyler Blazey; Yi Su; Jon Christensen; Jonathan McConathy; Patricia Aldea; David M. Holtzman; Nigel J. Cairns; John C. Morris; Anne M. Fagan; Beau M. Ances; Tammie Ls. Benzinger

analyses to compare the three participant groups to each other within each PET modality. All results were reported at a FDR corrected threshold p <0.05. Results: Both Flutametamol and PiB showed greater signal throughout grey matter (GM) structures in AD vs. eCN or yCN as expected. (Figure) In all intragroup comparisons (yCN, eCN, and AD), greater white matter (WM) uptake was seen with Flutametamol vs. PiB. In yCN and eCN greater diffuse GM uptake was also seen with Flutametamol vs. PiB (although not as uniform and with a lower T-statistic (5) vs white matter (15)). Greater scalp and parotid uptake was seen in Flutametamol vs. PiB. Greater venous sinus signal was seen with PiB vs Flutametamol. When comparing yCN to eCN for each imaging drug, greater WM uptake was seen in eCN vs yCN. Conclusions: Flutametamol and PiB show similar GM uptake distributions in AD dementia vs CN participants. Differences in WM accumulation between the two amyloid tracers suggest quantitative differences will be apparent when using WM as a reference region. Both imaging drugs demonstrate an age dependent increase in WM binding.


JAMA Neurology | 2012

11C-PiB imaging of human immunodeficiency virus-associated neurocognitive disorder.

Beau M. Ances; Tammie L.S. Benzinger; Jon Christensen; Jewell B. Thomas; Rohit Venkat; Mengesha Teshome; Patricia Aldea; Anne M. Fagan; David M. Holtzman; John C. Morris; David B. Clifford

Collaboration


Dive into the Patricia Aldea's collaboration.

Top Co-Authors

Avatar

John C. Morris

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Beau M. Ances

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Jon Christensen

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Yi Su

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Nigel J. Cairns

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Karl A. Friedrichsen

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Tammie L.S. Benzinger

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Anne M. Fagan

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Brian A. Gordon

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

David M. Holtzman

Washington University in St. Louis

View shared research outputs
Researchain Logo
Decentralizing Knowledge