Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 24
1.
Alzheimers Dement ; 20(5): 3472-3484, 2024 May.
Article En | MEDLINE | ID: mdl-38591250

INTRODUCTION: The course of depressive symptoms and dementia risk is unclear, as are potential structural neuropathological common causes. METHODS: Utilizing joint latent class mixture models, we identified longitudinal trajectories of annually assessed depressive symptoms and dementia risk over 21 years in 957 older women (baseline age 72.7 years old) from the Women's Health Initiative Memory Study. In a subsample of 569 women who underwent structural magnetic resonance imaging, we examined whether estimates of cerebrovascular disease and Alzheimer's disease (AD)-related neurodegeneration were associated with identified trajectories. RESULTS: Five trajectories of depressive symptoms and dementia risk were identified. Compared to women with minimal symptoms, women who reported mild and stable and emerging depressive symptoms were at the highest risk of developing dementia and had more cerebrovascular disease and AD-related neurodegeneration. DISCUSSION: There are heterogeneous profiles of depressive symptoms and dementia risk. Common neuropathological factors may contribute to both depression and dementia. Highlights The progression of depressive symptoms and concurrent dementia risk is heterogeneous. Emerging depressive symptoms may be a prodromal symptom of dementia. Cerebrovascular disease and AD are potentially shared neuropathological factors.


Dementia , Depression , Magnetic Resonance Imaging , Humans , Female , Aged , Dementia/pathology , Dementia/epidemiology , Longitudinal Studies , Brain/pathology , Brain/diagnostic imaging , Cerebrovascular Disorders/pathology , Alzheimer Disease/pathology , Disease Progression , Risk Factors
2.
Eur J Cancer ; 202: 113975, 2024 May.
Article En | MEDLINE | ID: mdl-38442645

The first-line treatment choice of EGFRIs plus doublet chemotherapy vs. bevacizumab plus doublet chemotherapy remains a topic of interest for patients with left-sided RAS WT mCRC. We conducted a systematic literature review and meta-analysis of clinical trial data published between 2015 and 2024. We evaluated the relative efficacy and safety of first-line EGFRIs plus doublet chemotherapy (FOLFIRI or FOLFOX) vs. bevacizumab plus doublet chemotherapy for patients with RAS WT left-sided mCRC, as well as in all- and right-sided tumors. We identified eight trials with 2624 patients. Five trials reported outcomes by tumor sidedness. In the left-sided population, overall survival (OS) (Hazard Ratio (HR) = 0.80, 95% Confidence Interval (CI): 0.71-0.90) and objective response rate (ORR) (Odds ratio [OR]=1.61, 95% CI: 1.30-1.99) favored EGFRI plus chemotherapy, while no statistically significant differences were observed for progression-free survival (PFS) (HR=0.93, 95% CI: 0.84-1.04) or resection rate (RR). Similar results were found in the all-sided population. In the right-sided population, PFS favored bevacizumab plus chemotherapy (HR=1.45, 95% CI: 1.19-1.78), while no statistically significant differences were observed for OS (HR=1.17, 95% CI: 0.95-1.44), ORR (OR=0.99, 95% CI: 0.69-1.41), and RR. Early tumor shrinkage in the all-sided population favored EGFRI plus chemotherapy (OR=1.72; 95% CI: 1.36-2.17); limited data precluded evaluation by sidedness. Safety was available in 6 trials for all-sided tumors and 1 trial for left-sided tumors, each demonstrating typical class-specific adverse events. This most comprehensive meta-analysis indicates a benefit for first-line EGFRI plus chemotherapy over bevacizumab plus chemotherapy in patients with left-sided RAS WT mCRC.


Colorectal Neoplasms , Adult , Humans , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bevacizumab/adverse effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , ErbB Receptors
3.
medRxiv ; 2023 Nov 29.
Article En | MEDLINE | ID: mdl-38076972

Exposure to ambient air pollution, especially particulate matter with aerodynamic diameter <2.5 µm (PM2.5) and nitrogen dioxide (NO2), are environmental risk factors for Alzheimer's disease and related dementia. The medial temporal lobe (MTL) is an important brain region subserving episodic memory that atrophies with age, during the Alzheimer's disease continuum, and is vulnerable to the effects of cerebrovascular disease. Despite the importance of air pollution it is unclear whether exposure leads to atrophy of the MTL and by what pathways. Here we conducted a longitudinal study examining associations between ambient air pollution exposure and MTL atrophy and whether putative air pollution exposure effects resembled Alzheimer's disease-related neurodegeneration or cerebrovascular disease-related neurodegeneration. Participants included older women (n = 627; aged 71-87) who underwent two structural brain MRI scans (MRI-1: 2005-6; MRI-2: 2009-10) as part of the Women's Health Initiative Memory Study of Magnetic Resonance Imaging. Regionalized universal kriging was used to estimate annual concentrations of PM2.5 and NO2 at residential locations aggregated to 3-year averages prior to MRI-1. The outcome was 5-year standardized change in MTL volumes. Mediators included voxel-based MRI measures of the spatial pattern of neurodegeneration of Alzheimer's disease (Alzheimer's disease pattern similarity scores [AD-PS]) and whole-brain white matter small-vessel ischemic disease (WM-SVID) volume as a proxy of global cerebrovascular damage. Structural equation models were constructed to examine whether the associations between exposures with MTL atrophy were mediated by the initial level or concurrent change in AD-PS score or WM-SVID while adjusting for sociodemographic, lifestyle, clinical characteristics, and intracranial volume. Living in locations with higher PM2.5 (per interquartile range [IQR]=3.17µg/m3) or NO2 (per IQR=6.63ppb) was associated with greater MTL atrophy (ßPM2.5 = -0.29, 95% confidence interval [CI]=[-0.41,-0.18]; ßNO2 =-0.12, 95%CI=[-0.23,-0.02]). Greater PM2.5 was associated with larger increases in AD-PS (ßPM2.5 = 0.23, 95%CI=[0.12,0.33]) over time, which partially mediated associations with MTL atrophy (indirect effect= -0.10; 95%CI=[-0.15, -0.05]), explaining approximately 32% of the total effect. NO2 was positively associated with AD-PS at MRI-1 (ßNO2=0.13, 95%CI=[0.03,0.24]), which partially mediated the association with MTL atrophy (indirect effect= -0.01, 95% CI=[-0.03,-0.001]). Global WM-SVID at MRI-1 or concurrent change were not significant mediators between exposures and MTL atrophy. Findings support the mediating role of Alzheimer's disease-related neurodegeneration contributing to MTL atrophy associated with late-life exposures to air pollutants. Alzheimer's disease-related neurodegeneration only partially explained associations between exposure and MTL atrophy suggesting the role of multiple neuropathological processes underlying air pollution neurotoxicity on brain aging.

4.
medRxiv ; 2023 Nov 29.
Article En | MEDLINE | ID: mdl-38077091

Background: Ambient air pollution exposures increase risk for Alzheimer's disease (AD) and related dementias, possibly due to structural changes in the medial temporal lobe (MTL). However, existing MRI studies examining exposure effects on the MTL were cross-sectional and focused on the hippocampus, yielding mixed results. Method: To determine whether air pollution exposures were associated with MTL atrophy over time, we conducted a longitudinal study including 653 cognitively unimpaired community-dwelling older women from the Women's Health Initiative Memory Study with two MRI brain scans (MRI-1: 2005-6; MRI-2: 2009-10; Mage at MRI-1=77.3±3.5years). Using regionalized universal kriging models, exposures at residential locations were estimated as 3-year annual averages of fine particulate matter (PM2.5) and nitrogen dioxide (NO2) prior to MRI-1. Bilateral gray matter volumes of the hippocampus, amygdala, parahippocampal gyrus (PHG), and entorhinal cortex (ERC) were summed to operationalize the MTL. We used linear regressions to estimate exposure effects on 5-year volume changes in the MTL and its subregions, adjusting for intracranial volume, sociodemographic, lifestyle, and clinical characteristics. Results: On average, MTL volume decreased by 0.53±1.00cm3 over 5 years. For each interquartile increase of PM2.5 (3.26µg/m3) and NO2 (6.77ppb), adjusted MTL volume had greater shrinkage by 0.32cm3 (95%CI=[-0.43, -0.21]) and 0.12cm3 (95%CI=[-0.22, -0.01]), respectively. The exposure effects did not differ by APOE ε4 genotype, sociodemographic, and cardiovascular risk factors, and remained among women with low-level PM2.5 exposure. Greater PHG atrophy was associated with higher PM2.5 (b=-0.24, 95%CI=[-0.29, -0.19]) and NO2 exposures (b=-0.09, 95%CI=[-0.14, -0.04]). Higher exposure to PM2.5 but not NO2 was also associated with greater ERC atrophy. Exposures were not associated with amygdala or hippocampal atrophy. Conclusion: In summary, higher late-life PM2.5 and NO2 exposures were associated with greater MTL atrophy over time in cognitively unimpaired older women. The PHG and ERC - the MTL cortical subregions where AD neuropathologies likely begin, may be preferentially vulnerable to air pollution neurotoxicity.

5.
Lung Cancer ; 181: 107260, 2023 07.
Article En | MEDLINE | ID: mdl-37285629

INTRODUCTION: The KRAS G12C mutation has recently become a druggable target in non-small cell lung cancer (NSCLC). In this observational study, we present real-world clinicopathological characteristics, treatment patterns, and survival outcomes data in patients with KRAS mutation-positive advanced NSCLC (aNSCLC), including those with KRAS G12C and KRAS non-G12C mutations, who received docetaxel as standard-of-care treatment in the second-line and beyond (2L+). METHODS: US-based electronic health record-derived de-identified databases were used to assess clinicopathological characteristics and outcomes in adult aNSCLC patients with KRAS mutations treated with 2L+ docetaxel between January 1, 2011, and March 31, 2021. The primary endpoints were median real-world overall survival OS (rwOS) and median real-world progression-free survival (rwPFS), which were estimated in 2L, third-line, fourth-line, and 2L+ analysis sets among patients who had a 6-month minimum opportunity for follow-up and were not taking a clinical trial drug. RESULTS: Of the 677 patients with KRAS-mutant aNSCLC (KRAS mutant cohort) treated with 2L+ docetaxel, 295 (43.6%) had KRAS G12C mutation (KRAS G12C cohort) and 382 (56.4%) had KRAS non-G12C mutation (KRAS non-G12C cohort). Across all cohorts, approximately 47%, 35%, 14-15%, and 6-9% of patients received 2L, third-line, fourth-line, and fifth- or later-line docetaxel, respectively. In the KRAS G12C cohort, ∼68% of patients were treated with a PD-1/PD-L1 inhibitor prior to 2L+ docetaxel. Most 2L+ docetaxel regimens in the KRAS G12C cohort were combinations (59.5%), primarily with ramucirumab (45.2%). In the KRAS G12C cohort, the median rwOS and median rwPFS after 2L+ docetaxel were 6.0 (95% CI, 4.9-7.1) and 3.4 (95% CI, 2.7-4.2) months, respectively, with similar trends observed in other cohorts and lines of therapy. CONCLUSIONS: Real-world outcomes were poor in patients with KRAS G12C-mutated aNSCLC treated with 2L+ docetaxel. Targeted and more efficacious treatment options in these patients are warranted.


Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Adult , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Docetaxel/therapeutic use , Proto-Oncogene Proteins p21(ras)/genetics , Lung Neoplasms/drug therapy , Taxoids , Mutation
6.
Sci Total Environ ; 823: 153642, 2022 Jun 01.
Article En | MEDLINE | ID: mdl-35122843

Exposures to fine particulate matter (PM2.5) and nitrogen dioxide (NO2) have been associated with the emergence of depressive symptoms in older adulthood, although most studies used cross-sectional outcome measures. Elucidating the brain structures mediating the adverse effects can strengthen the causal role between air pollution and increasing depressive symptoms. We evaluated whether smaller volumes of brain structures implicated in late-life depression mediate associations between ambient air pollution exposure and changes in depressive symptoms. This prospective study included 764 community-dwelling older women (aged 81.6 ± 3.6 in 2008-2010) from the Women's Health Initiative Memory Study (WHIMS) Magnetic Resonance Imaging study (WHIMS-MRI; 2005-06) and WHIMS-Epidemiology of Cognitive Health Outcomes (WHIMS-ECHO; 2008-16). Three-year average annual mean concentrations (scaled by interquartile range [IQR]) of ambient PM2.5 (in µg/m3; IQR = 3.14 µg/m3) and NO2 (in ppb; IQR = 7.80 ppb) before WHIMS-MRI were estimated at participants' addresses via spatiotemporal models. Mediators included structural brain MRI-derived grey matter volumes of the prefrontal cortex and structures of the limbic-cortical-striatal-pallidal-thalamic circuit. Depressive symptoms were assessed annually by the 15-item Geriatric Depression Scale. Structural equation models were constructed to estimate associations between exposure, structural brain volumes, and depressive symptoms. Increased exposures (by each IQR) were associated with greater annual increases in depressive symptoms (ßPM2.5 = 0.022; 95% Confidence Interval (CI) = 0.003, 0.042; ßNO2 = 0.019; 95% CI = 0.001, 0.037). The smaller volume of prefrontal cortex associated with exposures partially mediated the associations of increased depressive symptoms with NO2 (8%) and PM2.5 (13%), and smaller insula volume associated with NO2 contributed modestly (13%) to the subsequent increase in depressive symptoms. We demonstrate the first evidence that the smaller volumes of the prefrontal cortex and insula may mediate the subsequent increases in depressive symptoms associated with late-life exposures to NO2 and PM2.5.


Air Pollutants , Air Pollution , Aged , Aged, 80 and over , Air Pollutants/analysis , Air Pollution/adverse effects , Air Pollution/analysis , Cross-Sectional Studies , Depression/epidemiology , Environmental Exposure/analysis , Female , Humans , Nitrogen Dioxide/analysis , Particulate Matter/analysis , Prefrontal Cortex/diagnostic imaging , Prospective Studies
7.
PLoS Med ; 19(2): e1003893, 2022 02.
Article En | MEDLINE | ID: mdl-35113870

BACKGROUND: Late-life exposure to ambient air pollution is a modifiable risk factor for dementia, but epidemiological studies have shown inconsistent evidence for cognitive decline. Air quality (AQ) improvement has been associated with improved cardiopulmonary health and decreased mortality, but to the best of our knowledge, no studies have examined the association with cognitive function. We examined whether AQ improvement was associated with slower rate of cognitive decline in older women aged 74 to 92 years. METHODS AND FINDINGS: We studied a cohort of 2,232 women residing in the 48 contiguous US states that were recruited from more than 40 study sites located in 24 states and Washington, DC from the Women's Health Initiative (WHI) Memory Study (WHIMS)-Epidemiology of Cognitive Health Outcomes (WHIMS-ECHO) study. They were predominantly non-Hispanic White women and were dementia free at baseline in 2008 to 2012. Measures of annual (2008 to 2018) cognitive function included the modified Telephone Interview for Cognitive Status (TICSm) and the telephone-based California Verbal Learning Test (CVLT). We used regionalized universal kriging models to estimate annual concentrations (1996 to 2012) of fine particulate matter (PM2.5) and nitrogen dioxide (NO2) at residential locations. Estimates were aggregated to the 3-year average immediately preceding (recent exposure) and 10 years prior to (remote exposure) WHIMS-ECHO enrollment. Individual-level improved AQ was calculated as the reduction from remote to recent exposures. Linear mixed effect models were used to examine the associations between improved AQ and the rates of cognitive declines in TICSm and CVLT trajectories, adjusting for sociodemographic (age; geographic region; race/ethnicity; education; income; and employment), lifestyle (physical activity; smoking; and alcohol), and clinical characteristics (prior hormone use; hormone therapy assignment; depression; cardiovascular disease (CVD); hypercholesterolemia; hypertension; diabetes; and body mass index [BMI]). For both PM2.5 and NO2, AQ improved significantly over the 10 years before WHIMS-ECHO enrollment. During a median of 6.2 (interquartile range [IQR] = 5.0) years of follow-up, declines in both general cognitive status (ß = -0.42/year, 95% CI: -0.44, -0.40) and episodic memory (ß = -0.59/year, 95% CI: -0.64, -0.54) were observed. Greater AQ improvement was associated with slower decline in TICSm (ßPM2.5improvement = 0.026 per year for improved PM2.5 by each IQR = 1.79 µg/m3 reduction, 95% CI: 0.001, 0.05; ßNO2improvement = 0.034 per year for improved NO2 by each IQR = 3.92 parts per billion [ppb] reduction, 95% CI: 0.01, 0.06) and CVLT (ßPM2.5 improvement = 0.070 per year for improved PM2.5 by each IQR = 1.79 µg/m3 reduction, 95% CI: 0.02, 0.12; ßNO2improvement = 0.060 per year for improved NO2 by each IQR = 3.97 ppb reduction, 95% CI: 0.005, 0.12) after adjusting for covariates. The respective associations with TICSm and CVLT were equivalent to the slower decline rate found with 0.9 to 1.2 and1.4 to 1.6 years of younger age and did not significantly differ by age, region, education, Apolipoprotein E (ApoE) e4 genotypes, or cardiovascular risk factors. The main limitations of this study include measurement error in exposure estimates, potential unmeasured confounding, and limited generalizability. CONCLUSIONS: In this study, we found that greater improvement in long-term AQ in late life was associated with slower cognitive declines in older women. This novel observation strengthens the epidemiologic evidence of an association between air pollution and cognitive aging.


Air Pollutants/adverse effects , Air Pollution/adverse effects , Cognitive Dysfunction/epidemiology , Environmental Exposure/adverse effects , Independent Living/trends , Interviews as Topic/methods , Aged , Aged, 80 and over , Air Pollutants/analysis , Air Pollution/analysis , Cognitive Dysfunction/prevention & control , Cognitive Dysfunction/psychology , Cohort Studies , Environmental Exposure/prevention & control , Female , Follow-Up Studies , Humans , Independent Living/psychology , Longitudinal Studies , Particulate Matter/adverse effects , Particulate Matter/analysis , Quality Improvement , United States/epidemiology , Verbal Learning/physiology
8.
Proc Natl Acad Sci U S A ; 119(2)2022 01 11.
Article En | MEDLINE | ID: mdl-34983871

Late-life ambient air pollution is a risk factor for brain aging, but it remains unknown if improved air quality (AQ) lowers dementia risk. We studied a geographically diverse cohort of older women dementia free at baseline in 2008 to 2012 (n = 2,239, aged 74 to 92). Incident dementia was centrally adjudicated annually. Yearly mean concentrations of fine particulate matter (PM2.5) and nitrogen dioxide (NO2) were estimated using regionalized national universal kriging models and averaged over the 3-y period before baseline (recent exposure) and 10 y earlier (remote exposure). Reduction from remote to recent exposures was used as the indicator of improved AQ. Cox proportional hazard ratios (HRs) for dementia risk associated with AQ measures were estimated, adjusting for sociodemographic, lifestyle, and clinical characteristics. We identified 398 dementia cases during follow up (median = 6.1 y). PM2.5 and NO2 reduced significantly over the 10 y before baseline. Larger AQ improvement was associated with reduced dementia risks (HRPM2.5 0.80 per 1.78 µg/m3, 95% CI 0.71-0.91; HRNO2 0.80 per 3.91 parts per billion, 95% CI 0.71-0.90), equivalent to the lower risk observed in women 2.4 y younger at baseline. Higher PM2.5 at baseline was associated with higher dementia risk (HRPM2.5 1.16 per 2.90 µg/m3, 95% CI 0.98-1.38), but the lower dementia risk associated with improved AQ remained after further adjusting for recent exposure. The observed associations did not substantially differ by age, education, geographic region, Apolipoprotein E e4 genotypes, or cardiovascular risk factors. Long-term AQ improvement in late life was associated with lower dementia risk in older women.


Air Pollution/analysis , Dementia/epidemiology , Aged , Aged, 80 and over , Air Pollutants/analysis , Cohort Studies , Environmental Exposure/adverse effects , Female , Humans , Incidence , Nitrogen Dioxide , Particulate Matter/analysis , Proportional Hazards Models , Risk Factors
9.
J Gerontol A Biol Sci Med Sci ; 77(5): 977-985, 2022 05 05.
Article En | MEDLINE | ID: mdl-34383042

BACKGROUND: Whether racial/ethnic disparities in Alzheimer's disease (AD) risk may be explained by ambient fine particles (PM2.5) has not been studied. METHOD: We conducted a prospective, population-based study on a cohort of Black (n = 481) and White (n = 6 004) older women (aged 65-79) without dementia at enrollment (1995-1998). Cox models accounting for competing risk were used to estimate the hazard ratio (HR) for racial/ethnic disparities in AD (1996-2010) defined by Diagnostic and Statistical Manual of Mental Disorders, 4th edition and the association with time-varying annual average PM2.5 (1999-2010) estimated by spatiotemporal model. RESULTS: Over an average follow-up of 8.3 (±3.5) years with 158 incident cases (21 in Black women), the racial disparities in AD risk (range of adjusted HRBlack women = 1.85-2.41) observed in various models could not be explained by geographic region, age, socioeconomic characteristics, lifestyle factors, cardiovascular risk factors, and hormone therapy assignment. Estimated PM2.5 exposure was higher in Black (14.38 ± 2.21 µg/m3) than in White (12.55 ± 2.76 µg/m3) women, and further adjustment for the association between PM2.5 and AD (adjusted HRPM2.5 = 1.18-1.28) slightly reduced the racial disparities by 2%-6% (HRBlack women = 1.81-2.26). The observed association between PM2.5 and AD risk was ~2 times greater in Black (HRPM2.5 = 2.10-2.60) than in White (HRPM2.5 = 1.07-1.15) women (range of interaction ps: <.01-.01). We found similar results after further adjusting for social engagement (social strain, social support, social activity, living alone), stressful life events, Women's Health Initiative's clinic sites, and neighborhood socioeconomic characteristics. CONCLUSIONS: PM2.5 may contribute to racial/ethnic disparities in AD risk and its associated increase in AD risk was stronger among Black women.


Air Pollutants , Air Pollution , Alzheimer Disease , Aged , Air Pollutants/adverse effects , Air Pollutants/analysis , Alzheimer Disease/chemically induced , Alzheimer Disease/epidemiology , Environmental Exposure/adverse effects , Environmental Exposure/analysis , Female , Humans , Particulate Matter/adverse effects , Particulate Matter/analysis , Prospective Studies
10.
Child Psychiatry Hum Dev ; 53(2): 199-211, 2022 04.
Article En | MEDLINE | ID: mdl-33449264

The developmental course of antisocial behavior is often described in terms of qualitatively distinct trajectories. However, the genetic etiology of various trajectories is not well understood. We examined heterogeneity in the development of delinquent and aggressive behavior in 1532 twin youth using four waves of data collection, spanning ages 9-10 to 16-18. A latent class growth analysis was used to uncover relevant subgroups. For delinquent behavior, three latent classes emerged: Non-Delinquent, Low-Level Delinquent, and Persistent Delinquent. Liability for persistent delinquency had a substantial genetic origin (heritability = 67%), whereas genetic influences were negligible for lower-risk subgroups. Three classes of aggressive behavior were identified: Non-Aggressive, Moderate, and High. Moderate heritability spanned the entire continuum of risk for aggressive behavior. Thus, there are differences between aggressive behavior and non-aggressive delinquency with respect to heterogeneity of etiology. We conclude that persistent delinquency represents an etiologically distinct class of rule-breaking with strong genetic roots.


Juvenile Delinquency , Adolescent , Aggression , Antisocial Personality Disorder/genetics , Humans
11.
J Alzheimers Dis ; 84(4): 1691-1707, 2021.
Article En | MEDLINE | ID: mdl-34744078

BACKGROUND: Elucidating associations between exposures to ambient air pollutants and profiles of cognitive performance may provide insight into neurotoxic effects on the aging brain. OBJECTIVE: We examined associations between empirically derived profiles of cognitive performance and residential concentrations of particulate matter of aerodynamic diameter < 2.5 (PM2.5) and nitrogen dioxide (NO2) in older women. METHOD: Women (N = 2,142) from the Women's Health Initiative Study of Cognitive Aging completed a neuropsychological assessment measuring attention, visuospatial, language, and episodic memory abilities. Average yearly concentrations of PM2.5 and NO2 were estimated at the participant's addresses for the 3 years prior to the assessment. Latent profile structural equation models identified subgroups of women exhibiting similar profiles across tests. Multinomial regressions examined associations between exposures and latent profile classification, controlling for covariates. RESULT: Five latent profiles were identified: low performance across multiple domains (poor multi-domain; n = 282;13%), relatively poor verbal episodic memory (poor memory; n = 216; 10%), average performance across all domains (average multi-domain; n = 974; 45%), superior memory (n = 381; 18%), and superior attention (n = 332; 15%). Using women with average cognitive ability as the referent, higher PM2.5 (per interquartile range [IQR] = 3.64µg/m3) was associated with greater odds of being classified in the poor memory (OR = 1.29; 95% Confidence Interval [CI] = 1.10-1.52) or superior attention (OR = 1.30; 95% CI = 1.10-1.53) profiles. NO2 (per IQR = 9.86 ppb) was associated with higher odds of being classified in the poor memory (OR = 1.38; 95% CI = 1.17-1.63) and lower odds of being classified with superior memory (OR = 0.81; 95% CI = 0.67-0.97). CONCLUSION: Exposure to PM2.5 and NO2 are associated with patterns of cognitive performance characterized by worse verbal episodic memory relative to performance in other domains.


Air Pollutants/adverse effects , Air Pollution/adverse effects , Cognition/physiology , Environmental Exposure , Neuropsychological Tests/statistics & numerical data , Particulate Matter/adverse effects , Aged , Brain/physiology , Female , Humans , Nitrogen Dioxide/adverse effects
12.
Environ Health Perspect ; 129(9): 97009, 2021 09.
Article En | MEDLINE | ID: mdl-34516296

BACKGROUND: Episodic memory decline varies by age and underlying neuropathology. Whether ambient air pollution contributes to the heterogeneity of episodic memory decline in older populations remains unclear. OBJECTIVES: We estimated associations between air pollution exposures and episodic memory decline according to pollutant, exposure time window, age, and latent class subgroups defined by episodic memory trajectories. METHODS: Participants were from the Women's Health Initiative Memory Study-Epidemiology of Cognitive Health Outcomes. Older women (n=2,056; 74-92 years of age) completed annual (2008-2018) episodic memory assessments using the telephone-based California Verbal Learning Test (CVLT). We estimated 3-y average fine particulate matter [PM with an aerodynamic diameter of ≤2.5µm (PM2.5)] and nitrogen dioxide (NO2) exposures at baseline and 10 y earlier (recent and remote exposures, respectively), using regionalized national universal kriging. Separate latent class mixed models were used to estimate associations between interquartile range increases in exposures and CVLT trajectories in women ≤80 and >80 years of age, adjusting for covariates. RESULTS: Two latent classes were identified for women ≤80 years of age (n=828), "slow-decliners" {slope=-0.12/y [95% confidence interval (CI): -0.23, -0.01] and "fast-decliners" [slope=-1.79/y (95% CI: -2.08, -1.50)]}. In the slow-decliner class, but not the fast-decliner class, PM2.5 exposures were associated with a greater decline in CVLT scores over time, with a stronger association for recent vs. remote exposures [-0.16/y (95% CI: -2.08, -0.03) per 2.88 µg/m3 and -0.11/y (95% CI: -0.22, 0.01) per 3.27 µg/m3, respectively]. Among women ≥80 years of age (n=1,128), the largest latent class comprised "steady-decliners" [slope=-1.35/y (95% CI: -1.53, -1.17)], whereas the second class, "cognitively resilient", had no decline in CVLT on average. PM2.5 was not associated with episodic memory decline in either class. A 6.25-ppb increase in recent NO2 was associated with nonsignificant acceleration of episodic memory decline in the ≤80-y-old fast-decliner class [-0.21/y (95% CI: -0.45, 0.04)], and in the >80-y-old cognitively resilient class [-0.10/y (95% CI: -0.24, 0.03)] and steady-decliner class [-0.11/y (95% CI: -0.27, 0.05)]. Associations with recent NO2 exposure in women >80 years of age were stronger and statistically significant when 267 women with incident probable dementia were excluded [e.g., -0.12/y (95% CI: -0.22, -0.02) for the cognitively resilient class]. In contrast with changes in CVLT over time, there were no associations between exposures and CVLT scores during follow-up in any subgroup. DISCUSSION: In a community-dwelling U.S. population of older women, associations between late-life exposure to ambient air pollution and episodic memory decline varied by age-related cognitive trajectories, exposure time windows, and pollutants. https://doi.org/10.1289/EHP7668.


Air Pollutants , Air Pollution , Memory, Episodic , Aged , Air Pollutants/analysis , Air Pollution/analysis , Environmental Exposure , Female , Humans , Particulate Matter/analysis
13.
Environ Pollut ; 271: 116282, 2021 Feb 15.
Article En | MEDLINE | ID: mdl-33385889

The interrelationships among long-term ambient air pollution exposure, emotional distress and cognitive decline in older adulthood remain unclear. Long-term exposure may impact cognitive performance and subsequently impact emotional health. Conversely, exposure may initially be associated with emotional distress followed by declines in cognitive performance. Here we tested the inter-relationship between global cognitive ability, emotional distress, and exposure to PM2.5 (particulate matter with aerodynamic diameter <2.5 µm) and NO2 (nitrogen dioxide) in 6118 older women (aged 70.6 ± 3.8 years) from the Women's Health Initiative Memory Study. Annual exposure to PM2.5 (interquartile range [IQR] = 3.37 µg/m3) and NO2 (IQR = 9.00 ppb) was estimated at the participant's residence using regionalized national universal kriging models and averaged over the 3-year period before the baseline assessment. Using structural equation mediation models, a latent factor capturing emotional distress was constructed using item-level data from the 6-item Center for Epidemiological Studies Depression Scale and the Short Form Health Survey Emotional Well-Being scale at baseline and one-year follow-up. Trajectories of global cognitive performance, assessed by the Modified-Mini Mental State Examination (3MS) annually up to 12 years, were estimated. All effects reported were adjusted for important confounders. Increases in PM2.5 (ß = -0.144 per IQR; 95% CI = -0.261; -0.028) and NO2 (ß = -0.157 per IQR; 95% CI = -0.291; -0.022) were associated with lower initial 3MS performance. Lower 3MS performance was associated with increased emotional distress (ß = -0.008; 95% CI = -0.015; -0.002) over the subsequent year. Significant indirect effect of both exposures on increases in emotional distress mediated by exposure effects on worse global cognitive performance were present. No statistically significant indirect associations were found between exposures and 3MS trajectories putatively mediated by baseline emotional distress. Our study findings support cognitive aging processes as a mediator of the association between PM2.5 and NO2 exposure and emotional distress in later-life.


Air Pollutants , Air Pollution , Adult , Aged , Air Pollutants/analysis , Air Pollution/adverse effects , Air Pollution/analysis , Cognition , Environmental Exposure/adverse effects , Environmental Exposure/analysis , Female , Humans , Nitrogen Dioxide/analysis , Particulate Matter/analysis , Psychological Distress
14.
J Am Geriatr Soc ; 69(2): 474-484, 2021 02.
Article En | MEDLINE | ID: mdl-33205418

BACKGROUND/OBJECTIVES: Exposure to air pollution may contribute to both increasing depressive symptoms and decreasing episodic memory in older adulthood, but few studies have examined this hypothesis in a longitudinal context. Accordingly, we examined the association between air pollution and changes in depressive symptoms (DS) and episodic memory (EM) and their interrelationship in oldest-old (aged 80 and older) women. DESIGN: Prospective cohort data from the Women's Health Initiative Memory Study-Epidemiology of Cognitive Health Outcomes. SETTING: Geographically diverse community-dwelling population. PARTICIPANTS: A total of 1,583 dementia-free women aged 80 and older. MEASUREMENTS: Women completed up to six annual memory assessments (latent composite of East Boston Memory Test and Telephone Interview for Cognitive Status) and the 15-item Geriatric Depression Scale (GDS-15). We estimated 3-year average exposures to regional particulate matter with aerodynamic diameter below 2.5 µm (PM2.5 ) (interquartile range [IQR] = 3.35 µg/m3 ) and gaseous nitrogen dioxide (NO2 ) (IQR = 9.55 ppb) at baseline and during a remote period 10 years earlier, using regionalized national universal kriging. RESULTS: Latent change structural equation models examined whether residing in areas with higher pollutant levels was associated with annual changes in standardized EM and DS while adjusting for potential confounders. Remote NO2 (ß = .287 per IQR; P = .002) and PM2.5 (ß = .170 per IQR; P = .019) exposure was significantly associated with larger increases in standardized DS, although the magnitude of the difference, less than 1 point on the GDS-15, is of questionable clinical significance. Higher DS were associated with accelerated EM declines (ß = -.372; P = .001), with a significant indirect effect of remote NO2 and PM2.5 exposure on EM declines mediated by DS. There were no other significant indirect exposure effects. CONCLUSION: These findings in oldest-old women point to potential adverse effects of late-life exposure to air pollution on subsequent interplay between DS and EM, highlighting air pollution as an environmental health risk factor for older women.


Air Pollution , Depression , Environmental Exposure , Memory Disorders , Memory, Episodic , Aged, 80 and over , Air Pollution/adverse effects , Air Pollution/analysis , Cognition/physiology , Depression/diagnosis , Depression/epidemiology , Environmental Exposure/adverse effects , Environmental Exposure/prevention & control , Female , Geriatric Assessment/methods , Humans , Independent Living/psychology , Independent Living/statistics & numerical data , Memory Disorders/diagnosis , Memory Disorders/epidemiology , Mental Status Schedule , Particulate Matter/analysis , Risk Assessment , Risk Factors , United States/epidemiology
15.
Neurology ; 2020 11 18.
Article En | MEDLINE | ID: mdl-33208540

OBJECTIVE: To examine whether late-life exposure to PM2.5 (particulate matter with aerodynamic diameters <2.5-µm) contributes to progressive brain atrophy predictive of Alzheimer's disease (AD) using a community-dwelling cohort of women (aged 70-89) with up to two brain MRI scans (MRI-1: 2005-6; MRI-2: 2010-11). METHODS: AD pattern similarity (AD-PS) scores, developed by supervised machine learning and validated with MRI data from the AD Neuroimaging Initiative, was used to capture high-dimensional gray matter atrophy in brain areas vulnerable to AD (e.g., amygdala, hippocampus, parahippocampal gyrus, thalamus, inferior temporal lobe areas and midbrain). Based on participants' addresses and air monitoring data, we implemented a spatiotemporal model to estimate 3-year average exposure to PM2.5 preceding MRI-1. General linear models were used to examine the association between PM2.5 and AD-PS scores (baseline and 5-year standardized change), accounting for potential confounders and white matter lesion volumes. RESULTS: For 1365 women aged 77.9±3.7 years in 2005-6, there was no association between PM2.5 and baseline AD-PS score in cross-sectional analyses (ß=-0.004; 95% CI: -0.019, 0.011). Longitudinally, each interquartile range increase of PM2.5 (2.82-µg/m3) was associated with increased AD-PS scores during the follow-up, equivalent to a 24% (hazard ratio=1.24; 95% CI: 1.14, 1.34) increase in AD risk over 5-years (n=712; aged 77.4±3.5 years). This association remained after adjustment for socio-demographics, intracranial volume, lifestyle, clinical characteristics, and white matter lesions, and was present with levels below US regulatory standards (<12-µg/m3). CONCLUSIONS: Late-life exposure to PM2.5 is associated with increased neuroanatomical risk of AD, which may not be explained by available indicators of cerebrovascular damage.

16.
Brain ; 143(1): 289-302, 2020 01 01.
Article En | MEDLINE | ID: mdl-31746986

Evidence suggests exposure to particulate matter with aerodynamic diameter <2.5 µm (PM2.5) may increase the risk for Alzheimer's disease and related dementias. Whether PM2.5 alters brain structure and accelerates the preclinical neuropsychological processes remains unknown. Early decline of episodic memory is detectable in preclinical Alzheimer's disease. Therefore, we conducted a longitudinal study to examine whether PM2.5 affects the episodic memory decline, and also explored the potential mediating role of increased neuroanatomic risk of Alzheimer's disease associated with exposure. Participants included older females (n = 998; aged 73-87) enrolled in both the Women's Health Initiative Study of Cognitive Aging and the Women's Health Initiative Memory Study of Magnetic Resonance Imaging, with annual (1999-2010) episodic memory assessment by the California Verbal Learning Test, including measures of immediate free recall/new learning (List A Trials 1-3; List B) and delayed free recall (short- and long-delay), and up to two brain scans (MRI-1: 2005-06; MRI-2: 2009-10). Subjects were assigned Alzheimer's disease pattern similarity scores (a brain-MRI measured neuroanatomical risk for Alzheimer's disease), developed by supervised machine learning and validated with data from the Alzheimer's Disease Neuroimaging Initiative. Based on residential histories and environmental data on air monitoring and simulated atmospheric chemistry, we used a spatiotemporal model to estimate 3-year average PM2.5 exposure preceding MRI-1. In multilevel structural equation models, PM2.5 was associated with greater declines in immediate recall and new learning, but no association was found with decline in delayed-recall or composite scores. For each interquartile increment (2.81 µg/m3) of PM2.5, the annual decline rate was significantly accelerated by 19.3% [95% confidence interval (CI) = 1.9% to 36.2%] for Trials 1-3 and 14.8% (4.4% to 24.9%) for List B performance, adjusting for multiple potential confounders. Long-term PM2.5 exposure was associated with increased Alzheimer's disease pattern similarity scores, which accounted for 22.6% (95% CI: 1% to 68.9%) and 10.7% (95% CI: 1.0% to 30.3%) of the total adverse PM2.5 effects on Trials 1-3 and List B, respectively. The observed associations remained after excluding incident cases of dementia and stroke during the follow-up, or further adjusting for small-vessel ischaemic disease volumes. Our findings illustrate the continuum of PM2.5 neurotoxicity that contributes to early decline of immediate free recall/new learning at the preclinical stage, which is mediated by progressive atrophy of grey matter indicative of increased Alzheimer's disease risk, independent of cerebrovascular damage.


Alzheimer Disease/epidemiology , Brain/diagnostic imaging , Environmental Exposure/statistics & numerical data , Memory, Episodic , Particulate Matter , Prodromal Symptoms , Aged , Aged, 80 and over , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/physiopathology , Alzheimer Disease/psychology , Cognitive Dysfunction/diagnostic imaging , Cognitive Dysfunction/epidemiology , Cognitive Dysfunction/physiopathology , Cognitive Dysfunction/psychology , Cohort Studies , Female , Humans , Longitudinal Studies , Magnetic Resonance Imaging , Prospective Studies , Risk Factors , United States/epidemiology
17.
Environ Int ; 135: 105196, 2020 02.
Article En | MEDLINE | ID: mdl-31881430

BACKGROUND: Emerging data suggests PM2.5 (particulate matter with aerodynamic diameter <2.5 µm) may be associated with both earlier declines in episodic memory (EM) and increased depressive symptoms in older adults. Although late-life depressive symptoms are associated with EM, no longitudinal studies have examined the inter-relationship among PM2.5, depressive symptoms and EM. METHODS: Older women (n = 2,202; aged 67-83 in 1999) enrolled in the Women's Health Initiative Study of Cognitive Aging completed up to eight annual assessments of depressive symptoms (15-item Geriatric Depression Scale) and EM (California Verbal Learning Test). A nationwide spatiotemporal model (1999-2010) was used to estimate ambient PM2.5 exposure at residential locations. Univariate and bivariate structural equation models (SEMs) for latent-change scores were used to examine how 3-year average PM2.5 preceding each assessment affects the temporal dynamics and bidirectional relations of annual changes in depressive symptoms and EM. RESULTS: In univariate SEMs, one inter-quartile (4.04 µg/m3) increment of 3-year PM2.5 was significantly (p < 0.05) associated with accelerated declines in verbal learning (List A trials 1-3: ß = -1.48) and free-recall memory (short-delay: ß = -1.43; long-delay: ß = -1.11), but not with change in depressive symptoms (ß = 0.12; p = 0.71). In bivariate SEMs, significant associations were observed between PM2.5 and accelerated declines in EM measures (ß = -1.44 to -0.99; p < 0.05) and between EM performance and changes in depressive symptoms (ß = -0.08 to -0.05; p < 0.05), with significant indirect PM2.5 effects on changes in depressive symptoms (ß = 0.08-0.10; p < 0.05). These findings were robust with adjustment for multiple demographic, lifestyle, and clinical factors, and remained after excluding subjects with dementia or mild cognitive impairment. No associations were found between PM2.5 and change in depressive symptoms or depressive symptoms and subsequent EM decline. CONCLUSIONS: Findings suggest that PM2.5 neurotoxicity may damage brain areas implicated in EM, followed by manifestation of depressive symptoms. Our data did not support depressive symptoms as the neuropsychological mediator of accelerated brain aging associated with PM2.5 exposure.


Air Pollutants , Depression , Memory, Episodic , Particulate Matter , Aged , Aged, 80 and over , Air Pollutants/toxicity , Environmental Exposure , Female , Humans , Longitudinal Studies , Particulate Matter/toxicity
18.
Front Public Health ; 7: 332, 2019.
Article En | MEDLINE | ID: mdl-31867298

Outdoor air pollution has been recognized as a novel environmental neurotoxin. Studies have begun to use brain Magnetic Resonance Imaging (MRI) to investigate how air pollution may adversely impact developing brains. A systematic review was conducted to evaluate and synthesize the reported evidence from MRI studies on how early-life exposure to outdoor air pollution affects neurodevelopment. Using PubMed and Web of Knowledge, we conducted a systematic search, followed by structural review of original articles with individual-level exposure data and that met other inclusion criteria. Six studies were identified, each sampled from 3 cohorts of children in Spain, The Netherlands, and the United States. All studies included a one-time assessment of brain MRI when children were 6-12 years old. Air pollutants from traffic and/or regional sources, including polycyclic aromatic hydrocarbons (PAHs), nitrogen dioxide, elemental carbon, particulate matter (<2.5 or <10 µm), and copper, were estimated prenatally (n = 1), during childhood (n = 3), or both (n = 2), using personal monitoring and urinary biomarkers (n = 1), air sampling at schools (n = 4), or a land-use regression (LUR) modeling based on residences (n = 2). Associations between exposure and brain were noted, including: smaller white matter surface area (n = 1) and microstructure (n = 1); region-specific patterns of cortical thinness (n = 1) and smaller volumes and/or less density within the caudate (n = 3); altered resting-state functional connectivity (n = 2) and brain activity to sensory stimuli (n = 1). Preliminary findings suggest that outdoor air pollutants may impact MRI brain structure and function, but limitations highlight that the design of future air pollution-neuroimaging studies needs to incorporate a developmental neurosciences perspective, considering the exposure timing, age of study population, and the most appropriate neurodevelopmental milestones.

19.
Am J Geriatr Psychiatry ; 27(10): 1083-1096, 2019 10.
Article En | MEDLINE | ID: mdl-31311712

OBJECTIVES: Although several environmental factors contribute to the etiology of late-life depressive symptoms, the role of ambient air pollution has been understudied. Experimental data support the neurotoxicity of airborne particulate matter with aerodynamic diameter of ≤2.5 µm (PM2.5), but it remains unclear whether long-term exposure is associated with late-life depressive symptoms. Our secondary aim was to explore whether the observed associations between exposure and depressive symptoms are explained by dementia risk. DESIGN, SETTING, AND PARTICIPANTS: Prospective community-dwelling cohort study from the Women's Health Initiative Study of Cognitive Aging (1999-2010). Our analyses included 1,989 older women (baseline age 73.3 ± 3.75) with no prior depression or cognitive impairment. MEASUREMENTS: Participants completed annual assessments of depressive symptoms (15-item Geriatric Depression Scale). Average ambient PM2.5 exposure at the residential location was estimated by spatiotemporal modeling for the 3-years preceding each neuropsychological assessment. Participants underwent separate annual examinations for incident dementia defined by DSM-IV. Latent-class mixture models examined the association between PM2.5 and identified trajectories of symptoms. RESULTS: Six trajectories of depressive symptoms were identified. Across all women, PM2.5 exposure was positively associated with depressive symptoms. The effect was especially strong in two clusters with sustained depressive symptoms (n = 625 sustained-mild [31%]; n = 125 sustained-moderate; [6%]). Among those with sustained-moderate symptoms, the estimated adverse effect of PM2.5 exposure was greater than that of hypertension. Among women without dementia, associations were modestly attenuated. CONCLUSION: Long-term exposure to ambient fine particles was associated with increased depressive symptoms among older women without prior depression or cognitive impairment.


Air Pollution/statistics & numerical data , Depression/epidemiology , Environmental Exposure/statistics & numerical data , Particulate Matter/analysis , Aged , Air Pollutants/analysis , Environmental Exposure/analysis , Female , Humans , Independent Living , Prospective Studies , Psychiatric Status Rating Scales , Randomized Controlled Trials as Topic
20.
Alzheimers Dement (N Y) ; 5: 118-128, 2019.
Article En | MEDLINE | ID: mdl-31011622

INTRODUCTION: In a geographically diverse sample of women, we asked whether cognitive reserve (CR) is best viewed as a general or cognitive domain-specific construct and whether some cognitive reserve domains but not others exert protective effects on risk of developing mild cognitive impairment (MCI) or dementia. METHODS: Estimates of general and domain-specific CR were derived via variance decomposition in 972 cognitively intact women from the Women's Health Initiative Study of Cognitive Aging and Women's Health Memory Study Magnetic Resonance Imaging. Women were then followed up for 13 years. RESULTS: General CR was the strongest predictor of reduced risk for both MCI and dementia, compared to domain-specific CR measures. Verbal memory, figural memory, and spatial CR were independently protective of MCI, but only verbal memory was independently associated with reduced risk for dementia. DISCUSSION: Cognitive reserve is a heterogenous construct with valid quantitative measures identifiable across different neuropsychological processes associated with MCI and dementia.

...