1
|
Zhou J, Sun X, Wang K, Shen M, Yu J, Yao Q, Hong H, Tang C, Wang Q. What Information do Systemic Pathological Changes Bring to the Diagnosis and Treatment of Alzheimer's Disease? Neurosci Bull 2025:10.1007/s12264-025-01399-z. [PMID: 40257662 DOI: 10.1007/s12264-025-01399-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 02/21/2025] [Indexed: 04/22/2025] Open
Abstract
Alzheimer's disease (AD) is regarded as a neurodegenerative disease, and it has been proposed that AD may be a systemic disease. Studies have reported associations between non-neurological diseases and AD. The correlations between AD pathology and systemic (non-neurological) pathological changes are intricate, and the mechanisms underlying these correlations and their causality are unclear. In this article, we review the association between AD and disorders of other systems. In addition, we summarize the possible mechanisms associated with AD and disorders of other systems, mainly from the perspective of AD pathology. Regarding the relationship between AD and systemic pathological changes, we aim to provide a new outlook on the early warning signs and treatment of AD, such as establishing a diagnostic and screening system based on more accessible peripheral samples.
Collapse
Affiliation(s)
- Jinyue Zhou
- Health Science Center, The First Affiliated Hospital, Ningbo University, Ningbo, 315010, China
| | - Xiaoli Sun
- Department of Chemistry, Lishui University, Lishui, 32300, China
| | - Keren Wang
- Health Science Center, School of Public Health, Ningbo University, Ningbo, 315211, China
| | - Min Shen
- Reference Laboratory, Medical System Biotechnology Co., Ltd, Ningbo, 315104, China
| | - Jingbo Yu
- Health Science Center, The First Affiliated Hospital, Ningbo University, Ningbo, 315010, China
| | - Qi Yao
- Health Science Center, The First Affiliated Hospital, Ningbo University, Ningbo, 315010, China
| | - Hang Hong
- Health Science Center, School of Public Health, Ningbo University, Ningbo, 315211, China.
| | - Chunlan Tang
- Health Science Center, School of Public Health, Ningbo University, Ningbo, 315211, China.
| | - Qinwen Wang
- Health Science Center, The First Affiliated Hospital, Ningbo University, Ningbo, 315010, China.
| |
Collapse
|
2
|
Einstein D, Jurgens S, Howard E, Hayes JP. Inflammation following childhood maltreatment is associated with episodic memory decline in older adults. J Trauma Stress 2025. [PMID: 40082728 DOI: 10.1002/jts.23138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 12/17/2024] [Accepted: 01/03/2025] [Indexed: 03/16/2025]
Abstract
Childhood maltreatment is recognized as a risk factor for cognitive decline in adulthood. However, the mechanisms underlying this association, particularly the role of systemic inflammation, remain understudied. To address this gap, this study investigated the indirect effects of inflammation on the associations between childhood maltreatment and both episodic memory (EM) and executive functioning (EF) performance 10 years after inflammatory measurement in older adults. We selected 590 participants (Mage = 65.5 years) from the Midlife in the United States Study based on available childhood maltreatment, inflammation, and composite cognitive data. Spearman's rank correlations were calculated to test associations among childhood maltreatment, cognition, and inflammation. The results informed follow-up analyses testing the indirect effects of inflammation on the associations between childhood maltreatment and cognition. Correlations demonstrated that inflammation was associated with overall childhood maltreatment as well as with specific domains of childhood maltreatment (i.e., physical abuse, sexual abuse, emotional abuse, and physical neglect), ps = .002-.010. Inflammation was negatively associated with EF, p = .001, and EM, p = .028. Follow-up analyses revealed significant indirect pathways linking overall childhood maltreatment, β = -.0088, SE = 0.0058, 95% CI [-0.0223, -0.00000], to EM performance through inflammation, but no specific domain of maltreatment drove this association. The results suggest that inflammation may help explain links between childhood maltreatment exposure and EM deficits in adulthood. These results elucidate the importance of evaluating childhood maltreatment as a risk factor for later-life cognitive decline, particularly within the context of heightened inflammatory biomarkers.
Collapse
Affiliation(s)
- Dalia Einstein
- Psychology Department, The Ohio State University, Columbus, Ohio, USA
| | - Savana Jurgens
- Psychology Department, The Ohio State University, Columbus, Ohio, USA
| | - Erica Howard
- Psychology Department, The Ohio State University, Columbus, Ohio, USA
| | - Jasmeet P Hayes
- Psychology Department, The Ohio State University, Columbus, Ohio, USA
- Chronic Brain Injury Initiative, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
3
|
Li D, An B, Men L, Glittenberg M, Lutsey PL, Mielke MM, Yu F, Hoogeveen RC, Gottesman R, Zhang L, Meyer M, Sullivan K, Zantek N, Alonso A, Walker KA. The association of high-density lipoprotein cargo proteins with brain volume in older adults in the Atherosclerosis Risk in Communities (ARIC). J Alzheimers Dis 2025; 103:724-734. [PMID: 39772982 DOI: 10.1177/13872877241305806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
BACKGROUND High-density lipoprotein (HDL) modulates the blood-brain barrier and cerebrovascular integrity, likely influencing the risk of Alzheimer's disease (AD), neurodegeneration, and cognitive decline. OBJECTIVE This study aims to identify HDL protein cargo associated with brain amyloid deposition and brain volume in regions vulnerable to AD pathology in older adults. METHODS HDL was separated from the plasma of 65 non-demented participants of the Atherosclerosis Risk in Communities (ARIC) study using a fast protein liquid chromatography method. HDL cargo proteins were measured using a label-free, untargeted proteomic method based on mass spectrometry and data-independent acquisition. Linear regression with multiple imputations assessed the associations between each HDL cargo protein (log2-transformed) and brain amyloid deposition or temporal-parietal meta-ROI volume, adjusting for covariates. RESULTS The mean (SD) age of the participants was 76.3 (5.4) years old, 53.8% (35/65) female, 30.8% (20/65) black, and 28.1% (18/64, 1 missing) APOE4 carriers. We found few HDL cargo proteins associated with brain amyloid deposition and considerably more HDL cargo proteins associated with temporal-parietal meta-ROI volume. Two HDL cargo proteins mostly associated with temporoparietal meta-ROI volume were fibrinogen B (FGB) and plasminogen (PLG). A doubling of FGB in HDL was associated with a greater temporoparietal meta-ROI volume of 1638 mm3 (95% CI [688, 2589]). In comparison, a doubling of PLG in HDL was associated with a lower temporoparietal meta-ROI of 2025 mm3 (95% CI [-3669, -1034]). CONCLUSIONS This study suggests that HDL cargo proteins associated with temporal-parietal meta-ROI volume are involved in complement and coagulation pathways.
Collapse
Affiliation(s)
- Danni Li
- Department of Lab Medicine Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Binchong An
- Department of Lab Medicine Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Lu Men
- Department of Lab Medicine Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Matthew Glittenberg
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Pamela L Lutsey
- School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Michelle M Mielke
- Department of Epidemiology and Prevention, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Fang Yu
- Edson College of Nursing and Health Innovation, Arizona State University, Phoenix, AZ, USA
| | - Ron C Hoogeveen
- Division of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Rebecca Gottesman
- Stroke, Cognition, and Neuroepidemiology Section, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Lin Zhang
- School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Michelle Meyer
- Gillings School of Global Public Health, Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kevin Sullivan
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Nicole Zantek
- Department of Lab Medicine Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Alvaro Alonso
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Keenan A Walker
- Multimodal Imaging of Neurodegenerative Disease (MIND) Unit, National Institute of Aging, Intramural Research Program, Baltimore, MD, USA
| |
Collapse
|
4
|
How EH, Chin SM, Teo CH, Parhar IS, Soga T. Accelerated biological brain aging in major depressive disorder. Rev Neurosci 2024; 35:959-968. [PMID: 39002110 DOI: 10.1515/revneuro-2024-0025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 06/26/2024] [Indexed: 07/15/2024]
Abstract
Major depressive disorder (MDD) patients commonly encounter multiple types of functional disabilities, such as social, physical, and role functioning. MDD is related to an accreted risk of brain atrophy, aging-associated brain diseases, and mortality. Based on recently available studies, there are correlations between notable biological brain aging and MDD in adulthood. Despite several clinical and epidemiological studies that associate MDD with aging phenotypes, the underlying mechanisms in the brain remain unknown. The key areas in the study of biological brain aging in MDD are structural brain aging, impairment in functional connectivity, and the impact on cognitive function and age-related disorders. Various measurements have been used to determine the severity of brain aging, such as the brain age gap estimate (BrainAGE) or brain-predicted age difference (BrainPAD). This review summarized the current results of brain imaging data on the similarities between the manifestation of brain structural changes and the age-associated processes in MDD. This review also provided recent evidence of BrainPAD or BrainAGE scores in MDD, brain structural abnormalities, and functional connectivity, which are commonly observed between MDD and age-associated processes. It serves as a basis of current reference for future research on the potential areas of investigation for diagnostic, preventive, and potentially therapeutic purposes for brain aging in MDD.
Collapse
Affiliation(s)
- Eng Han How
- 65210 Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia , Jalan Lagoon Selatan, Bandar Sunway, 47500, Selangor, Malaysia
| | - Shar-Maine Chin
- 65210 Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia , Jalan Lagoon Selatan, Bandar Sunway, 47500, Selangor, Malaysia
| | - Chuin Hau Teo
- 65210 Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia , Jalan Lagoon Selatan, Bandar Sunway, 47500, Selangor, Malaysia
| | - Ishwar S Parhar
- Center Initiatives for Training International Researchers (CiTIR), University of Toyama, Gofuku, 930-8555 Toyama, Japan
| | - Tomoko Soga
- 65210 Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia , Jalan Lagoon Selatan, Bandar Sunway, 47500, Selangor, Malaysia
| |
Collapse
|
5
|
Aretz B, Doblhammer G, Heneka MT. The role of leukocytes in cognitive impairment due to long-term exposure to fine particulate matter: A large population-based mediation analysis. Alzheimers Dement 2024; 20:8715-8727. [PMID: 39412000 DOI: 10.1002/alz.14320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 09/12/2024] [Accepted: 09/13/2024] [Indexed: 12/25/2024]
Abstract
INTRODUCTION Our understanding of how fine particulate matter (PM2.5) impacts cognitive functioning is limited. Systemic inflammation processes may play a role in mediating this effect. METHODS This prospective cohort study used data from 66,254 participants aged 18+ between 2006 and 2015 from the Dutch Lifelines Cohort Study and Biobank. Causal mediation analysis was conducted to examine the impact of ambient PM2.5 exposure on cognitive processing time (CPT), using the change in white blood cell (WBC) count and its subtypes as potential mediators. RESULTS Heightened PM2.5 exposure was associated with slower CPT (total effect = 81.76 × 10-3, 95% confidence interval [CI] 59.51 × 10-3-105.31 × 10-3). The effect was partially mediated via increased WBC count (indirect effect [IE] = 0.42 × 10-3, 95% CI 0.07 × 10-3-0.90 × 10-3), particularly driven by an increase in monocytes (IE = 0.73 × 10-3, 95% CI 0.24 × 10-3-1.31 × 10-3). DISCUSSION Systemic inflammation processes may partially explain the harmful effects of PM2.5 on cognitive functioning, why lower levels of systemic inflammation may help contain its neurotoxic effects. HIGHLIGHTS The pathways leading to the neurotoxic effects of fine particulate matter (PM2.5) are poorly understood. We analyzed data from over 66,000 participants using causal pathway analysis. Increased white blood cell (WBC) count mediates the effect of PM2.5 on cognitive functioning. Monocyte count played a crucial role in this low-pollution setting. Systemic inflammation may contribute to the neurotoxic effects of PM2.5.
Collapse
Affiliation(s)
- Benjamin Aretz
- Institute of Sociology and Demography, University of Rostock, Rostock, Mecklenburg-Vorpommern, Germany
- Institute of General Practice and Family Medicine, University Hospital Bonn, Bonn, Nordrhein-Westfalen, Germany
| | - Gabriele Doblhammer
- Institute of Sociology and Demography, University of Rostock, Rostock, Mecklenburg-Vorpommern, Germany
- German Center for Neurodegenerative Diseases(DZNE), Demographic Studies, Bonn, Germany
| | - Michael T Heneka
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belvaux, Esch-sur-Alzette, Luxembourg
- Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
6
|
Tallino S, Etebari R, McDonough I, Leon H, Sepulveda I, Winslow W, Bartholomew SK, Perez SE, Mufson EJ, Velazquez R. Assessing the Benefit of Dietary Choline Supplementation Throughout Adulthood in the Ts65Dn Mouse Model of Down Syndrome. Nutrients 2024; 16:4167. [PMID: 39683562 DOI: 10.3390/nu16234167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 11/21/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND/OBJECTIVES Down syndrome (DS) is the most common cause of early-onset Alzheimer's disease (AD). Dietary choline has been proposed as a modifiable factor to improve the cognitive and pathological outcomes of AD and DS, especially as many do not reach adequate daily intake levels of choline. While lower circulating choline levels correlate with worse pathological measures in AD patients, choline status and intake in DS is widely understudied. Perinatal choline supplementation (Ch+) in the Ts65Dn mouse model of DS protects offspring against AD-relevant pathology and improves cognition. Further, dietary Ch+ in adult AD models also ameliorates pathology and improves cognition. However, dietary Ch+ in adult Ts65Dn mice has not yet been explored; thus, this study aimed to supply Ch+ throughout adulthood to determine the effects on cognition and DS co-morbidities. METHODS We fed trisomic Ts65Dn mice and disomic littermate controls either a choline normal (ChN; 1.1 g/kg) or a Ch+ (5 g/kg) diet from 4.5 to 14 months of age. RESULTS We found that Ch+ in adulthood failed to improve genotype-specific deficits in spatial learning. However, in both genotypes of female mice, Ch+ significantly improved cognitive flexibility in a reverse place preference task in the IntelliCage behavioral phenotyping system. Further, Ch+ significantly reduced weight gain and peripheral inflammation in female mice of both genotypes, and significantly improved glucose metabolism in male mice of both genotypes. CONCLUSIONS Our findings suggest that adulthood choline supplementation benefits behavioral and biological factors important for general well-being in DS and related to AD risk.
Collapse
Affiliation(s)
- Savannah Tallino
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Rachel Etebari
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
| | - Ian McDonough
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
| | - Hector Leon
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Isabella Sepulveda
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Wendy Winslow
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Samantha K Bartholomew
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Sylvia E Perez
- Barrow Neurological Institute, Phoenix, AZ 85013, USA
- Arizona Alzheimer's Consortium, Phoenix, AZ 85014, USA
| | - Elliott J Mufson
- Barrow Neurological Institute, Phoenix, AZ 85013, USA
- Arizona Alzheimer's Consortium, Phoenix, AZ 85014, USA
| | - Ramon Velazquez
- Banner Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ 85281, USA
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, USA
- Arizona Alzheimer's Consortium, Phoenix, AZ 85014, USA
| |
Collapse
|
7
|
McNeil CJ, Habota T, Sandu AL, Waiter G, Whalley H, Murray AD. The Influence of Birth Weight, Socio-Economic Status, and Adult Health on Brain Volumes during Ageing. Neuroepidemiology 2024:1-9. [PMID: 39401501 DOI: 10.1159/000541918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 09/24/2024] [Indexed: 11/14/2024] Open
Abstract
INTRODUCTION Greater late-life brain volumes are associated with resilience against dementia. We examined relationships between birth weight, lifelong socio-economic status, and health with late-life brain volumes. We hypothesised that early life factors directly affect late-life brain volumes. METHODS Adults aged 59-67 y underwent MRI and brain volumes were measured. Birth weight and lifelong health, and socio-economic status were quantified and the principal components of each extracted. Relationships were examined using regression and structural equation analysis. RESULTS Birth weight (β = 0.095, p = 0.017) and childhood socio-economic status (β = 0.091, p = 0.033, n = 280) were directly associated with brain volume. Childhood socio-economic status was further associated with grey matter volume (β = 0.04, p = 0.047). Adult health was linked to increased brain volume (β = 0.15, p = 0.003). CONCLUSION Birth weight and childhood socio-economic status are associated with whole and regional brain volume through direct mechanisms. Optimal fetal development, reduced childhood poverty, and good adult health could reduce brain atrophy and delay dementia onset in late-life.
Collapse
Affiliation(s)
| | - Tina Habota
- Aberdeen Biomedical Imaging Centre, University of Aberdeen, Aberdeen, UK
| | - Anca-Larisa Sandu
- Aberdeen Biomedical Imaging Centre, University of Aberdeen, Aberdeen, UK
| | - Gordon Waiter
- Aberdeen Biomedical Imaging Centre, University of Aberdeen, Aberdeen, UK
| | - Heather Whalley
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- Generation Scotland, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Alison D Murray
- Aberdeen Biomedical Imaging Centre, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
8
|
Shaw BC, Anders VR, Tinkey RA, Habean ML, Brock OD, Frostino BJ, Williams JL. Immunity impacts cognitive deficits across neurological disorders. J Neurochem 2024; 168:3512-3535. [PMID: 37899543 PMCID: PMC11056485 DOI: 10.1111/jnc.15999] [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: 08/01/2023] [Revised: 10/06/2023] [Accepted: 10/10/2023] [Indexed: 10/31/2023]
Abstract
Cognitive deficits are a common comorbidity with neurological disorders and normal aging. Inflammation is associated with multiple diseases including classical neurodegenerative dementias such as Alzheimer's disease (AD) and autoimmune disorders such as multiple sclerosis (MS), in which over half of all patients experience some form of cognitive deficits. Other degenerative diseases of the central nervous system (CNS) including frontotemporal lobe dementia (FTLD), and Parkinson's disease (PD) as well as traumatic brain injury (TBI) and psychological disorders like major depressive disorder (MDD), and even normal aging all have cytokine-associated reductions in cognitive function. Thus, there is likely commonality between these secondary cognitive deficits and inflammation. Neurological disorders are increasingly associated with substantial neuroinflammation, in which CNS-resident cells secrete cytokines and chemokines such as tumor necrosis factor (TNF)α and interleukins (ILs) including IL-1β and IL-6. CNS-resident cells also respond to a wide variety of cytokines and chemokines, which can have both direct effects on neurons by changing the expression of ion channels and perturbing electrical properties, as well as indirect effects through glia-glia and immune-glia cross-talk. There is significant overlap in these cytokine and chemokine expression profiles across diseases, with TNFα and IL-6 strongly associated with cognitive deficits in multiple disorders. Here, we review the involvement of various cytokines and chemokines in AD, MS, FTLD, PD, TBI, MDD, and normal aging in the absence of dementia. We propose that the neuropsychiatric phenotypes observed in these disorders may be at least partially attributable to a dysregulation of immunity resulting in pathological cytokine and chemokine expression from both CNS-resident and non-resident cells.
Collapse
Affiliation(s)
- Benjamin C. Shaw
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Victoria R. Anders
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Rachel A. Tinkey
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- School of Biomedical Sciences, Kent State University, Kent, OH, USA
- Brain Health Research Institute, Kent State University, Kent, OH, USA
| | - Maria L. Habean
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Neuroscience, Case Western Reserve University, Cleveland, OH, USA
| | - Orion D. Brock
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Molecular Medicine, Lerner Research Institute, Cleveland Clinic and Case Western Reserve University, Cleveland, OH, USA
| | - Benjamin J. Frostino
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- College of Science, University of Notre Dame, South Bend, IN, USA
| | - Jessica L. Williams
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- School of Biomedical Sciences, Kent State University, Kent, OH, USA
- Brain Health Research Institute, Kent State University, Kent, OH, USA
- Department of Neuroscience, Case Western Reserve University, Cleveland, OH, USA
- Molecular Medicine, Lerner Research Institute, Cleveland Clinic and Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
9
|
Yao L, Li MY, Wang KC, Liu YZ, Zheng HZ, Zhong Z, Ma SQ, Yang HM, Sun MM, He M, Huang HP, Wang HF. Abnormal resting-state functional connectivity of hippocampal subregions in type 2 diabetes mellitus-associated cognitive decline. Front Psychiatry 2024; 15:1360623. [PMID: 39376966 PMCID: PMC11456530 DOI: 10.3389/fpsyt.2024.1360623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 09/03/2024] [Indexed: 10/09/2024] Open
Abstract
Objective Type 2 diabetes mellitus (T2DM) over time predisposes to inflammatory responses and abnormalities in functional brain networks that damage learning, memory, or executive function. The hippocampus is a key region often reporting connectivity abnormalities in memory disorders. Here, we investigated peripheral inflammatory responses and resting-state functional connectivity (RSFC) changes characterized of hippocampal subregions in type 2 diabetes-associated cognitive decline (T2DACD). Methods The study included 16 patients with T2DM, 16 patients with T2DACD and 25 healthy controls (HCs). Subjects were assessed for cognitive performance, tested for the expression of inflammatory factors IL-6, IL-10 and TNF-α in peripheral serum, underwent resting-state functional magnetic resonance imaging scans, and analyzed for RSFC using the hippocampal subregions as seeds. We also calculated the correlation between cognitive performance and RSFC of hippocampal subregion, and analyzed the significantly altered RSFC values of T2DACD for Receiver Operating Characteristic (ROC) analysis. Results T2DACD patients showed a decline in their ability to complete cognitive assessment scales and experimental paradigms, and T2DM did not show abnormal cognitive performance. IL-6 expression was increased in peripheral serum in both T2DACD and T2DM. Compared with HCs, T2DACD showed abnormalities RSFC of the left anterior hippocampus with left precentral gyrus and left angular gyrus. T2DM showed abnormalities RSFC of the left middle hippocampus with right medial frontal gyrus, right anterior and middle hippocampus with left precuneus, left anterior hippocampus with right precuneus and right posterior middle temporal gyrus. Compared with T2DM, T2DACD showed abnormalities RSFC of the left posterior hippocampus and right middle hippocampus with left precuneus. In addition, RSFC in the left posterior hippocampus with left precuneus of T2DACD was positively correlated with Flanker conflict response time (r=0.766, P=0.001). In the ROC analysis, the significantly altered RSFC values of T2DACD achieved significant performance. Conclusions T2DACD showed a significant decrease in attentional inhibition and working memory, peripheral pro-inflammatory response increased, and abnormalities RSFC of the hippocampal subregions with default mode network and sensory-motor network. T2DM did not show a significant cognitive decline, but peripheral pro-inflammatory response increased and abnormalities RSFC of the hippocampus subregions occurred in the brain. In addition, the left precuneus may be a key brain region in the conversion of T2DM to T2DACD. The results of this study may provide a basis for the preliminary diagnosis of T2DACD.
Collapse
Affiliation(s)
- Lin Yao
- Institute of Acupuncture and Massage, Northeast Asian Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Meng-Yuan Li
- Institute of Acupuncture and Massage, Northeast Asian Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Kang-Cheng Wang
- College of Psychology, Shandong Normal University, Jinan, Shandong, China
| | - Yan-Ze Liu
- Acupuncture and Tuina Center, The Third Affiliated Clinical Hospital of Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Hai-Zhu Zheng
- College of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Zhen Zhong
- College of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Shi-Qi Ma
- College of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Hong-Mei Yang
- Institute of Acupuncture and Massage, Northeast Asian Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Meng-Meng Sun
- Institute of Acupuncture and Massage, Northeast Asian Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Min He
- Institute of Acupuncture and Massage, Northeast Asian Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Hai-Peng Huang
- Institute of Acupuncture and Massage, Northeast Asian Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Hong-Feng Wang
- Institute of Acupuncture and Massage, Northeast Asian Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| |
Collapse
|
10
|
Duggan MR, Peng Z, Sipilä PN, Lindbohm JV, Chen J, Lu Y, Davatzikos C, Erus G, Hohman TJ, Andrews SJ, Candia J, Tanaka T, Joynes CM, Alvarado CX, Nalls MA, Cordon J, Daya GN, An Y, Lewis A, Moghekar A, Palta P, Coresh J, Ferrucci L, Kivimäki M, Walker KA. Proteomics identifies potential immunological drivers of postinfection brain atrophy and cognitive decline. NATURE AGING 2024; 4:1263-1278. [PMID: 39143319 PMCID: PMC11408246 DOI: 10.1038/s43587-024-00682-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 07/11/2024] [Indexed: 08/16/2024]
Abstract
Infections have been associated with the incidence of Alzheimer disease and related dementias, but the mechanisms responsible for these associations remain unclear. Using a multicohort approach, we found that influenza, viral, respiratory, and skin and subcutaneous infections were associated with increased long-term dementia risk. These infections were also associated with region-specific brain volume loss, most commonly in the temporal lobe. We identified 260 out of 942 immunologically relevant proteins in plasma that were differentially expressed in individuals with an infection history. Of the infection-related proteins, 35 predicted volumetric changes in brain regions vulnerable to infection-specific atrophy. Several of these proteins, including PIK3CG, PACSIN2, and PRKCB, were related to cognitive decline and plasma biomarkers of dementia (Aβ42/40, GFAP, NfL, pTau-181). Genetic variants that influenced expression of immunologically relevant infection-related proteins, including ITGB6 and TLR5, predicted brain volume loss. Our findings support the role of infections in dementia risk and identify molecular mediators by which infections may contribute to neurodegeneration.
Collapse
Affiliation(s)
- Michael R Duggan
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Zhongsheng Peng
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Pyry N Sipilä
- Clinicum, Department of Public Health, University of Helsinki, Helsinki, Finland
- Finnish Institute of Occupational Health, Helsinki, Finland
| | - Joni V Lindbohm
- Clinicum, Department of Public Health, University of Helsinki, Helsinki, Finland
- Broad Institute of the MIT and Harvard University, The Klarman Cell Observatory, Cambridge, MA, USA
- Brain Sciences, University College London, London, UK
| | - Jingsha Chen
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Yifei Lu
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Guray Erus
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Timothy J Hohman
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Shea J Andrews
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Julián Candia
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Toshiko Tanaka
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Cassandra M Joynes
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, USA
| | - Chelsea X Alvarado
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, USA
- DataTecnica LLC, Washington, DC, USA
| | - Mike A Nalls
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, USA
- DataTecnica LLC, Washington, DC, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Jenifer Cordon
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Gulzar N Daya
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Yang An
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Alexandria Lewis
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Abhay Moghekar
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Priya Palta
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Mika Kivimäki
- Clinicum, Department of Public Health, University of Helsinki, Helsinki, Finland
- Brain Sciences, University College London, London, UK
| | - Keenan A Walker
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA.
| |
Collapse
|
11
|
Matsuo K, Nshihara H. Rebuilding insight into the pathophysiology of Alzheimer's disease through new blood-brain barrier models. Neural Regen Res 2024; 19:1954-1960. [PMID: 38227521 DOI: 10.4103/1673-5374.390978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 11/01/2023] [Indexed: 01/17/2024] Open
Abstract
The blood-brain barrier is a unique function of the microvasculature in the brain parenchyma that maintains homeostasis in the central nervous system. Blood-brain barrier breakdown is a common pathology in various neurological diseases, such as Alzheimer's disease, stroke, multiple sclerosis, and Parkinson's disease. Traditionally, it has been considered a consequence of neuroinflammation or neurodegeneration, but recent advanced imaging techniques and detailed studies in animal models show that blood-brain barrier breakdown occurs early in the disease process and may precede neuronal loss. Thus, the blood-brain barrier is attractive as a potential therapeutic target for neurological diseases that lack effective therapeutics. To elucidate the molecular mechanism underlying blood-brain barrier breakdown and translate them into therapeutic strategies for neurological diseases, there is a growing demand for experimental models of human origin that allow for functional assessments. Recently, several human induced pluripotent stem cell-derived blood-brain barrier models have been established and various in vitro blood-brain barrier models using microdevices have been proposed. Especially in the Alzheimer's disease field, the human evidence for blood-brain barrier dysfunction has been demonstrated and human induced pluripotent stem cell-derived blood-brain barrier models have suggested the putative molecular mechanisms of pathological blood-brain barrier. In this review, we summarize recent evidence of blood-brain barrier dysfunction in Alzheimer's disease from pathological analyses, imaging studies, animal models, and stem cell sources. Additionally, we discuss the potential future directions for blood-brain barrier research.
Collapse
Affiliation(s)
- Kinya Matsuo
- Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Hideaki Nshihara
- Department of Neurotherapeutics, Yamaguchi University, Ube, Japan
| |
Collapse
|
12
|
Duggan MR, Yang Z, Cui Y, Dark HE, Wen J, Erus G, Hohman TJ, Chen J, Lewis A, Moghekar A, Coresh J, Resnick SM, Davatzikos C, Walker KA. Proteomic analyses reveal plasma EFEMP1 and CXCL12 as biomarkers and determinants of neurodegeneration. Alzheimers Dement 2024; 20:6486-6505. [PMID: 39129354 PMCID: PMC11497673 DOI: 10.1002/alz.14142] [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: 04/18/2024] [Revised: 06/18/2024] [Accepted: 06/25/2024] [Indexed: 08/13/2024]
Abstract
INTRODUCTION Plasma proteomic analyses of unique brain atrophy patterns may illuminate peripheral drivers of neurodegeneration and identify novel biomarkers for predicting clinically relevant outcomes. METHODS We identified proteomic signatures associated with machine learning-derived aging- and Alzheimer's disease (AD) -related brain atrophy patterns in the Baltimore Longitudinal Study of Aging (n = 815). Using data from five cohorts, we examined whether candidate proteins were associated with AD endophenotypes and long-term dementia risk. RESULTS Plasma proteins associated with distinct patterns of age- and AD-related atrophy were also associated with plasma/cerebrospinal fluid (CSF) AD biomarkers, cognition, AD risk, as well as mid-life (20-year) and late-life (8-year) dementia risk. EFEMP1 and CXCL12 showed the most consistent associations across cohorts and were mechanistically implicated as determinants of brain structure using genetic methods, including Mendelian randomization. DISCUSSION Our findings reveal plasma proteomic signatures of unique aging- and AD-related brain atrophy patterns and implicate EFEMP1 and CXCL12 as important molecular drivers of neurodegeneration. HIGHLIGHTS Plasma proteomic signatures are associated with unique patterns of brain atrophy. Brain atrophy-related proteins predict clinically relevant outcomes across cohorts. Genetic variation underlying plasma EFEMP1 and CXCL12 influences brain structure. EFEMP1 and CXCL12 may be important molecular drivers of neurodegeneration.
Collapse
Affiliation(s)
- Michael R. Duggan
- Laboratory of Behavioral NeuroscienceNational Institute on Aging, National Institutes of HealthBaltimoreMarylandUSA
| | - Zhijian Yang
- Artificial Intelligence in Biomedical Imaging LaboratoryPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Yuhan Cui
- Artificial Intelligence in Biomedical Imaging LaboratoryPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Heather E. Dark
- Laboratory of Behavioral NeuroscienceNational Institute on Aging, National Institutes of HealthBaltimoreMarylandUSA
| | - Junhao Wen
- Laboratory of Artificial Intelligence and Biomedical ScienceKeck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Guray Erus
- Artificial Intelligence in Biomedical Imaging LaboratoryPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Timothy J. Hohman
- Vanderbilt Memory and Alzheimer's CenterVanderbilt University Medical CenterNashvilleTennesseeUSA
- Vanderbilt Genetics InstituteVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Jingsha Chen
- Department of EpidemiologyJohns Hopkins University Bloomberg School of Public HealthBaltimoreMarylandUSA
| | - Alexandria Lewis
- Department of NeurologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Abhay Moghekar
- Department of NeurologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Josef Coresh
- Departments of Population Health and MedicineNew York University Grossman School of MedicineNew YorkNew YorkUSA
| | - Susan M. Resnick
- Laboratory of Behavioral NeuroscienceNational Institute on Aging, National Institutes of HealthBaltimoreMarylandUSA
| | - Christos Davatzikos
- Artificial Intelligence in Biomedical Imaging LaboratoryPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Keenan A. Walker
- Laboratory of Behavioral NeuroscienceNational Institute on Aging, National Institutes of HealthBaltimoreMarylandUSA
| |
Collapse
|
13
|
Ma J, Subramaniam P, Yancey JR, Farrington AA, McGlade EC, Renshaw PF, Yurgelun-Todd DA. Elevated circulating soluble interleukin-2 receptor (sCD25) level is associated with prefrontal excitatory-inhibitory imbalance in individuals with chronic pain: A proton MRS study. Brain Behav Immun 2024; 120:1-9. [PMID: 38772429 PMCID: PMC11269041 DOI: 10.1016/j.bbi.2024.05.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 04/29/2024] [Accepted: 05/18/2024] [Indexed: 05/23/2024] Open
Abstract
Aberrant neuronal excitability in the anterior cingulate cortex (ACC) is implicated in cognitive and affective pain processing. Such excitability may be amplified by activated circulating immune cells, including T lymphocytes, that interact with the central nervous system. Here, we conducted a study of individuals with chronic pain using magnetic resonance spectroscopy (MRS) to investigate the clinical evidence for the interaction between peripheral immune activation and prefrontal excitatory-inhibitory imbalance. In thirty individuals with chronic musculoskeletal pain, we assessed markers of peripheral immune activation, including soluble interleukin-2 receptor alpha chain (sCD25) levels, as well as brain metabolites, including Glx (glutamate + glutamine) to GABA+ (γ-aminobutyric acid + macromolecules/homocarnosine) ratio in the ACC. We found that the circulating level of sCD25 was associated with prefrontal Glx/GABA+. Greater prefrontal Glx/GABA+ was associated with higher pain catastrophizing, evaluative pain ratings, and anxiodepressive symptoms. Further, the interaction effect of sCD25 and prefrontal Glx/GABA+ on pain catastrophizing was significant, indicating the joint association of these two markers with pain catastrophizing. Our results provide the first evidence suggesting that peripheral T cellular activation, as reflected by elevated circulating sCD25 levels, may be linked to prefrontal excitatory-inhibitory imbalance in individuals with chronic pain. The interaction between these two systems may play a role as a potential mechanism underlying pain catastrophizing. Further prospective and treatment studies are needed to elucidate the specific role of the immune and brain interaction in pain catastrophizing.
Collapse
Affiliation(s)
- Jiyoung Ma
- Department of Psychiatry, University of Utah School of Medicine, Salt Lake City, UT, USA; Diagnostic Neuroimaging Laboratory, Huntsman Mental Health Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Punitha Subramaniam
- Department of Psychiatry, University of Utah School of Medicine, Salt Lake City, UT, USA; Diagnostic Neuroimaging Laboratory, Huntsman Mental Health Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - James R Yancey
- Department of Psychiatry, University of Utah School of Medicine, Salt Lake City, UT, USA; Diagnostic Neuroimaging Laboratory, Huntsman Mental Health Institute, University of Utah School of Medicine, Salt Lake City, UT, USA; George E. Wahlen Department of Veterans Affairs Medical Center, VISN 19 Mental Illness Research, Education and Clinical Center, Salt Lake City, UT, USA
| | - Amy A Farrington
- Department of Psychiatry, University of Utah School of Medicine, Salt Lake City, UT, USA; Diagnostic Neuroimaging Laboratory, Huntsman Mental Health Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Erin C McGlade
- Department of Psychiatry, University of Utah School of Medicine, Salt Lake City, UT, USA; Diagnostic Neuroimaging Laboratory, Huntsman Mental Health Institute, University of Utah School of Medicine, Salt Lake City, UT, USA; George E. Wahlen Department of Veterans Affairs Medical Center, VISN 19 Mental Illness Research, Education and Clinical Center, Salt Lake City, UT, USA
| | - Perry F Renshaw
- Department of Psychiatry, University of Utah School of Medicine, Salt Lake City, UT, USA; Diagnostic Neuroimaging Laboratory, Huntsman Mental Health Institute, University of Utah School of Medicine, Salt Lake City, UT, USA; George E. Wahlen Department of Veterans Affairs Medical Center, VISN 19 Mental Illness Research, Education and Clinical Center, Salt Lake City, UT, USA
| | - Deborah A Yurgelun-Todd
- Department of Psychiatry, University of Utah School of Medicine, Salt Lake City, UT, USA; Diagnostic Neuroimaging Laboratory, Huntsman Mental Health Institute, University of Utah School of Medicine, Salt Lake City, UT, USA; George E. Wahlen Department of Veterans Affairs Medical Center, VISN 19 Mental Illness Research, Education and Clinical Center, Salt Lake City, UT, USA.
| |
Collapse
|
14
|
Tong Y, Jia Y, Gong A, Li F, Zeng R. Systemic inflammation in midlife is associated with late-life functional limitations. Sci Rep 2024; 14:17434. [PMID: 39075139 PMCID: PMC11286743 DOI: 10.1038/s41598-024-68724-w] [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: 12/23/2023] [Accepted: 07/26/2024] [Indexed: 07/31/2024] Open
Abstract
Systemic inflammation generally coexists with functional limitations that seriously affect quality of life. This study aimed to investigate the association between systemic inflammation in midlife and the risk of functional limitations in late-life. A total of 10,044 participants with an average age of 53.9 ± 5.7 years at baseline were included in a cohort study. At the last follow-up, the prevalence of impaired activities of daily living (ADLs), instrumental activities of daily living (IADLs), and lower extremity function (LEF) was 14.7%, 21.6%, and 50.3%, respectively. The values of four inflammatory biomarkers were used to calculate the inflammation composite score. Compared with the participants in the lowest quartile of the inflammation composite score (Q1), those in the highest quartile (Q4) exhibited an odds ratio (OR) of 1.589 and a 95% confidence interval (CI) of 1.335-1.892 for impaired ADLs, an OR of 1.426 and a 95% CI of 1.228-1.657 for impaired IADLs, and an OR of 1.728 and a 95% CI of 1.526-1.957 for impaired LEF. The association between systemic inflammation and functional limitations was partly mediated by cardiac and brain function. The present study provides evidence that systemic inflammation in midlife is associated with a higher risk of late-life functional limitations. Protecting vital organ functions in midlife may have a positive impact on reducing the risk of future functional limitations.Trial registration: www.clinicaltrials.gov ; Unique identifier: NCT00005131.
Collapse
Grants
- No. 2023YFS0027, 2023YFS0240, 2023YFS0074, 2023NSFSC1652, 2022YFS0279, 2021YFQ0062, 2022JDRC0148 Sichuan Province Science and Technology Support Program
- No. 2023YFS0027, 2023YFS0240, 2023YFS0074, 2023NSFSC1652, 2022YFS0279, 2021YFQ0062, 2022JDRC0148 Sichuan Province Science and Technology Support Program
- No. 2023YFS0027, 2023YFS0240, 2023YFS0074, 2023NSFSC1652, 2022YFS0279, 2021YFQ0062, 2022JDRC0148 Sichuan Province Science and Technology Support Program
- No. 2023YFS0027, 2023YFS0240, 2023YFS0074, 2023NSFSC1652, 2022YFS0279, 2021YFQ0062, 2022JDRC0148 Sichuan Province Science and Technology Support Program
- No. 2023YFS0027, 2023YFS0240, 2023YFS0074, 2023NSFSC1652, 2022YFS0279, 2021YFQ0062, 2022JDRC0148 Sichuan Province Science and Technology Support Program
- No. ZH2022-101 Health Commission of Sichuan Province
- No. ZH2022-101 Health Commission of Sichuan Province
- No. ZH2022-101 Health Commission of Sichuan Province
- No. ZH2022-101 Health Commission of Sichuan Province
- No. ZH2022-101 Health Commission of Sichuan Province
- No. HXHL21016 Sichuan University West China Nursing Discipline Development Special Fund Project
- No. HXHL21016 Sichuan University West China Nursing Discipline Development Special Fund Project
- No. HXHL21016 Sichuan University West China Nursing Discipline Development Special Fund Project
- No. HXHL21016 Sichuan University West China Nursing Discipline Development Special Fund Project
- No. HXHL21016 Sichuan University West China Nursing Discipline Development Special Fund Project
Collapse
Affiliation(s)
- Yao Tong
- Department of Cardiology, West China Hospital, Sichuan University, 37 Guoxue Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yu Jia
- General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Aobo Gong
- Department of Cardiology, West China Hospital, Sichuan University, 37 Guoxue Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Fanghui Li
- Department of Cardiology, West China Hospital, Sichuan University, 37 Guoxue Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Rui Zeng
- Department of Cardiology, West China Hospital, Sichuan University, 37 Guoxue Road, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
15
|
Bahorik AL, Hoang TD, Jacobs DR, Levine DA, Yaffe K. Association of Changes in C-Reactive Protein Level Trajectories Through Early Adulthood With Cognitive Function at Midlife: The CARDIA Study. Neurology 2024; 103:e209526. [PMID: 38959452 PMCID: PMC11226328 DOI: 10.1212/wnl.0000000000209526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 04/02/2024] [Indexed: 07/05/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Late-life inflammation has been linked to dementia risk and preclinical cognitive decline, but less is known about early adult inflammation and whether this could influence cognition in midlife. We aimed to identify inflammation levels through early adulthood and determine association of these trajectories with midlife cognition. METHODS We used data from the Coronary Artery Risk Development in Young Adults study to identify inflammation trajectories (C-reactive protein [CRP] level <10 mg/L) over 18 years through early adulthood (age range 24-58) in latent class analysis and to assess associations with cognition 5 years after the last CRP measurement (age range 47-63). Six cognitive domains were evaluated from tests of verbal memory, processing speed, executive function, verbal and category fluency, and global cognition; poor cognitive performance was defined as a decline of ≥1 SD less than the mean on each domain. The primary outcome was poor cognitive performance. Logistic regression was used to adjust for demographics, smoking, alcohol use, physical activity, and APOE 4 status. RESULTS Among 2,364 participants, the mean (SD) age was 50.2 (3.5) years; 55% were female, and 57% were White. Three CRP trajectories emerged over 18 years: lower stable (45%), moderate/increasing (16%), and consistently higher (39%). Compared with lower stable CRP, both consistently higher (adjusted odds ratio [aOR] 1.67, 95% CI 1.23-2.26) and moderately/increasing (aOR 2.04, 95% CI 1.40-2.96) CRP had higher odds of poor processing speed; consistently higher CRP additionally had higher odds of poor executive function (aOR 1.36, 95% CI 1.00-1.88). For memory (moderately/increasing aOR 1.36, 95% CI 1.00-1.88; consistently higher aOR 1.18, 95% CI 0.90-1.54), letter fluency (moderately/increasing aOR 1.00, 95% CI 0.69-1.43; consistently higher aOR 1.05, 95% CI 0.80-1.39), category fluency (moderately/increasing aOR 1.16, 95% CI 0.82-1.63; consistently higher aOR 1.11, 95% CI 0.85-1.45), or global cognition (moderately/increasing aOR 1.16, 95% CI 0.82-1.63; consistently higher aOR 1.11, 95% CI 0.85-1.45), no association was observed. DISCUSSION Consistently higher or moderate/increasing inflammation starting in early adulthood may lead to worse midlife executive function and processing speed. Study limitations include selection bias due to loss to follow-up and reliance on CRP as the only inflammatory marker. Inflammation is important for cognitive aging and may begin much earlier than previously known.
Collapse
Affiliation(s)
- Amber L Bahorik
- From the Department of Psychiatry and Behavioral Sciences (A.L.B., K.Y.), University of California, San Francisco; Northern California Institute Research for Research and Education (T.D.H.), San Francisco, CA; School of Public Health (D.R.J.), University of Minnesota, Minneapolis; Department of Internal Medicine (D.A.L.), and Department of Neurology (D.A.L.), University of Michigan, Ann Arbor; Department of Neurology (K.Y.), and Department of Epidemiology and Biostatistics (K.Y.), University of California, San Francisco
| | - Tina D Hoang
- From the Department of Psychiatry and Behavioral Sciences (A.L.B., K.Y.), University of California, San Francisco; Northern California Institute Research for Research and Education (T.D.H.), San Francisco, CA; School of Public Health (D.R.J.), University of Minnesota, Minneapolis; Department of Internal Medicine (D.A.L.), and Department of Neurology (D.A.L.), University of Michigan, Ann Arbor; Department of Neurology (K.Y.), and Department of Epidemiology and Biostatistics (K.Y.), University of California, San Francisco
| | - David R Jacobs
- From the Department of Psychiatry and Behavioral Sciences (A.L.B., K.Y.), University of California, San Francisco; Northern California Institute Research for Research and Education (T.D.H.), San Francisco, CA; School of Public Health (D.R.J.), University of Minnesota, Minneapolis; Department of Internal Medicine (D.A.L.), and Department of Neurology (D.A.L.), University of Michigan, Ann Arbor; Department of Neurology (K.Y.), and Department of Epidemiology and Biostatistics (K.Y.), University of California, San Francisco
| | - Deborah A Levine
- From the Department of Psychiatry and Behavioral Sciences (A.L.B., K.Y.), University of California, San Francisco; Northern California Institute Research for Research and Education (T.D.H.), San Francisco, CA; School of Public Health (D.R.J.), University of Minnesota, Minneapolis; Department of Internal Medicine (D.A.L.), and Department of Neurology (D.A.L.), University of Michigan, Ann Arbor; Department of Neurology (K.Y.), and Department of Epidemiology and Biostatistics (K.Y.), University of California, San Francisco
| | - Kristine Yaffe
- From the Department of Psychiatry and Behavioral Sciences (A.L.B., K.Y.), University of California, San Francisco; Northern California Institute Research for Research and Education (T.D.H.), San Francisco, CA; School of Public Health (D.R.J.), University of Minnesota, Minneapolis; Department of Internal Medicine (D.A.L.), and Department of Neurology (D.A.L.), University of Michigan, Ann Arbor; Department of Neurology (K.Y.), and Department of Epidemiology and Biostatistics (K.Y.), University of California, San Francisco
| |
Collapse
|
16
|
Frye BM, Negrey JD, Johnson CSC, Kim J, Barcus RA, Lockhart SN, Whitlow CT, Chiou KL, Snyder-Mackler N, Montine TJ, Craft S, Shively CA, Register TC. Mediterranean diet protects against a neuroinflammatory cortical transcriptome: Associations with brain volumetrics, peripheral inflammation, social isolation, and anxiety in nonhuman primates (Macaca fascicularis). Brain Behav Immun 2024; 119:681-692. [PMID: 38636565 PMCID: PMC12051215 DOI: 10.1016/j.bbi.2024.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/17/2024] [Accepted: 04/16/2024] [Indexed: 04/20/2024] Open
Abstract
Mediterranean diets may be neuroprotective and prevent cognitive decline relative to Western diets; however, the underlying biology is poorly understood. We assessed the effects of Western versus Mediterranean-like diets on RNAseq-generated transcriptional profiles in lateral temporal cortex and their relationships with longitudinal changes in neuroanatomy, circulating monocyte gene expression, and observations of social isolation and anxiety in 38 socially-housed, middle-aged female cynomolgus macaques (Macaca fascicularis). Diet resulted in differential expression of seven transcripts (FDR < 0.05). Cyclin dependent kinase 14 (CDK14), a proinflammatory regulator, was lower in the Mediterranean group. The remaining six transcripts [i.e., "lunatic fringe" (LFNG), mannose receptor C type 2 (MRC2), solute carrier family 3 member 2 (SLCA32), butyrophilin subfamily 2 member A1 (BTN2A1), katanin regulatory subunit B1 (KATNB1), and transmembrane protein 268 (TMEM268)] were higher in cortex of the Mediterranean group and generally associated with anti-inflammatory/neuroprotective pathways. KATNB1 encodes a subcomponent of katanin, important in maintaining microtubule homeostasis. BTN2A1 is involved in immunomodulation of γδ T-cells which have anti-neuroinflammatory and neuroprotective effects. CDK14, LFNG, MRC2, and SLCA32 are associated with inflammatory pathways. The latter four differentially expressed cortex transcripts were associated with peripheral monocyte transcript levels, neuroanatomical changes determined by MRI, and with social isolation and anxiety. These results provide important insights into the potential mechanistic processes linking diet, peripheral and central inflammation, and behavior. Collectively, our results provide evidence that, relative to Western diets, Mediterranean diets confer protection against peripheral and central inflammation which is reflected in preserved brain structure and socioemotional behavior. Ultimately, such protective effects may confer resilience to the development of neuropathology and associated disease.
Collapse
Affiliation(s)
- Brett M Frye
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC, USA; Department of Biology, Emory and Henry College, Emory, VA, USA; Wake Forest Alzheimer's Disease Research Center, Winston-Salem, NC, USA
| | - Jacob D Negrey
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC, USA; School of Anthropology, University of Arizona, Tucson, AZ, USA
| | | | - Jeongchul Kim
- Department of Radiology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Richard A Barcus
- Department of Radiology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Samuel N Lockhart
- Wake Forest Alzheimer's Disease Research Center, Winston-Salem, NC, USA; Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Christopher T Whitlow
- Wake Forest Alzheimer's Disease Research Center, Winston-Salem, NC, USA; Department of Radiology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Kenneth L Chiou
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, USA; School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Noah Snyder-Mackler
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, USA; School of Life Sciences, Arizona State University, Tempe, AZ, USA; School of Human Evolution and Social Change, Arizona State University, Tempe, AZ, USA
| | | | - Suzanne Craft
- Wake Forest Alzheimer's Disease Research Center, Winston-Salem, NC, USA; Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Carol A Shively
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC, USA; Wake Forest Alzheimer's Disease Research Center, Winston-Salem, NC, USA.
| | - Thomas C Register
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC, USA; Wake Forest Alzheimer's Disease Research Center, Winston-Salem, NC, USA.
| |
Collapse
|
17
|
Scholz R, Brösamle D, Yuan X, Beyer M, Neher JJ. Epigenetic control of microglial immune responses. Immunol Rev 2024; 323:209-226. [PMID: 38491845 DOI: 10.1111/imr.13317] [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: 12/18/2023] [Accepted: 03/02/2024] [Indexed: 03/18/2024]
Abstract
Microglia, the major population of brain-resident macrophages, are now recognized as a heterogeneous population comprising several cell subtypes with different (so far mostly supposed) functions in health and disease. A number of studies have performed molecular characterization of these different microglial activation states over the last years making use of "omics" technologies, that is transcriptomics, proteomics and, less frequently, epigenomics profiling. These approaches offer the possibility to identify disease mechanisms, discover novel diagnostic biomarkers, and develop new therapeutic strategies. Here, we focus on epigenetic profiling as a means to understand microglial immune responses beyond what other omics methods can offer, that is, revealing past and present molecular responses, gene regulatory networks and potential future response trajectories, and defining cell subtype-specific disease relevance through mapping non-coding genetic variants. We review the current knowledge in the field regarding epigenetic regulation of microglial identity and function, provide an exemplary analysis that demonstrates the advantages of performing joint transcriptomic and epigenomic profiling of single microglial cells and discuss how comprehensive epigenetic analyses may enhance our understanding of microglial pathophysiology.
Collapse
Affiliation(s)
- Rebekka Scholz
- Immunogenomics & Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Desirée Brösamle
- Biomedical Center (BMC), Biochemistry, Faculty of Medicine, LMU Munich, Munich, Germany
- Neuroimmunology and Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Xidi Yuan
- Biomedical Center (BMC), Biochemistry, Faculty of Medicine, LMU Munich, Munich, Germany
- Neuroimmunology and Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Marc Beyer
- Immunogenomics & Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases (DZNE) and University of Bonn and West German Genome Center, Bonn, Germany
| | - Jonas J Neher
- Biomedical Center (BMC), Biochemistry, Faculty of Medicine, LMU Munich, Munich, Germany
- Neuroimmunology and Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
18
|
Huang H, Li J, Shen J, Zhao T, Xiao R, Ma W. Dietary Inflammatory Index and Cognitive Function: Findings from a Cross-Sectional Study in Obese Chinese Township Population from 45 to 75 Years. J Inflamm Res 2024; 17:2365-2382. [PMID: 38651005 PMCID: PMC11034566 DOI: 10.2147/jir.s447300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 04/02/2024] [Indexed: 04/25/2024] Open
Abstract
Background and Objective Cognitive dysfunction is highly prevalent in obese people, and food is a key factor in obesity, and dietary inflammatory index (DII) can reflect whether diet has anti-inflammatory or pro-inflammatory potential. In addition, dietary fatty acid consumption is linked to inflammation, obesity, and cognitive impairment. Erythrocyte membrane fatty acids can reflect dietary fatty acid intake. Our hypothesis was that erythrocyte membrane fatty acids might have a significant impact on the relationship between DII and cognition in obese individuals, and we designed experiments to test the hypothesis. Methods In three villages in Beijing, we collected 579 respondents from individuals 45 to 75 years old and categorized them by body mass index. The Montreal Cognitive Assessment (MoCA) score and DII score was calculated and gas chromatography was used to measure the proportion of erythrocyte membrane fatty acids. The relationship between the DII score and cognition was examined using multiple linear regression and binary logistic regression. Mediation analysis can help to understand the causal chain between variables, deeply explore the internal relationship and mechanism of action between variables. So a multiple chain mediation model was developed to investigate the mediating factors between the DII score and cognitive association. Results According to adjusted linear regression, higher DII scores were linked to lower MoCA scores in the obese group. The negative correlation between DII score and cognitive function score remains in binary linear regression. We discovered through mediation analysis that erythrocyte membrane fatty acids mediate the detrimental link between DII and cognitive function in obese individuals. Conclusion We propose that higher DII scores in obese people are associated with a decline in cognitive function. In addition, this effect might be mediated via the fatty acids in the erythrocyte membrane.
Collapse
Affiliation(s)
- Hongying Huang
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, People’s Republic of China
| | - Jinchen Li
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, People’s Republic of China
| | - Jingyi Shen
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, People’s Republic of China
| | - Tong Zhao
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, People’s Republic of China
| | - Rong Xiao
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, People’s Republic of China
| | - Weiwei Ma
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, People’s Republic of China
| |
Collapse
|
19
|
Christova P, James LM, Georgopoulos AP. Negative association between neurovascular coupling and cortical gray matter volume during the lifespan. J Neurophysiol 2024; 131:778-784. [PMID: 38478986 PMCID: PMC11305651 DOI: 10.1152/jn.00005.2024] [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: 01/05/2024] [Revised: 02/20/2024] [Accepted: 03/13/2024] [Indexed: 04/16/2024] Open
Abstract
Recent studies have established the moment-to-moment turnover of the blood-oxygen-level-dependent signal (TBOLD) at resting state as a key measure of local cortical brain function. Here, we sought to extend that line of research by evaluating TBOLD in 70 cortical areas with respect to corresponding brain volume, age, and sex across the lifespan in 1,344 healthy participants including 633 from the Human Connectome Project (HCP)-Development cohort (294 males and 339 females, age range 8-21 yr) and 711 healthy participants from HCP-Aging cohort (316 males and 395 females, 36-90 yr old). In both groups, we found that 1) TBOLD increased with age, 2) volume decreased with age, and 3) TBOLD and volume were highly significantly negatively correlated, independent of age. The inverse association between TBOLD and volume was documented in nearly all 70 brain areas and for both sexes, with slightly stronger associations documented for males. The strong correspondence between TBOLD and volume across age and sex suggests a common influence such as chronic neuroinflammation contributing to reduced cortical volume and increased TBOLD across the lifespan.NEW & NOTEWORTHY We report a significant negative association between resting functional magnetic resonance imaging (fMRI) blood-oxygen-level-dependent (BOLD) signal turnover (TBOLD) and cortical gray matter volume across the lifespan, such that TBOLD increased whereas volume decreased. We attribute this association to a hypothesized chronic, low-grade neuroinflammation, probably induced by various neurotropic pathogens, including human herpes viruses known to be dormant in the brain in a latent state and reactivated by stress, fever, and various environmental exposures, such as ultraviolet light.
Collapse
Affiliation(s)
- Peka Christova
- Department of Veterans Affairs Health Care System, The Neuroimaging Research Group, Brain Sciences Center, Minneapolis, Minnesota, United States
- Department of Neuroscience, University of Minnesota Medical School, Minneapolis, Minnesota, United States
- Cognitive Sciences Center, University of Minnesota, Minneapolis, Minnesota, United States
| | - Lisa M James
- Department of Veterans Affairs Health Care System, The Neuroimaging Research Group, Brain Sciences Center, Minneapolis, Minnesota, United States
- Department of Neuroscience, University of Minnesota Medical School, Minneapolis, Minnesota, United States
- Cognitive Sciences Center, University of Minnesota, Minneapolis, Minnesota, United States
- Department of Psychiatry, University of Minnesota Medical School, Minneapolis, Minnesota, United States
| | - Apostolos P Georgopoulos
- Department of Veterans Affairs Health Care System, The Neuroimaging Research Group, Brain Sciences Center, Minneapolis, Minnesota, United States
- Department of Neuroscience, University of Minnesota Medical School, Minneapolis, Minnesota, United States
- Cognitive Sciences Center, University of Minnesota, Minneapolis, Minnesota, United States
- Department of Psychiatry, University of Minnesota Medical School, Minneapolis, Minnesota, United States
- Department of Neurology, University of Minnesota Medical School, Minneapolis, Minnesota, United States
| |
Collapse
|
20
|
Kundura L, Cezar R, Gimenez S, Pastore M, Reynes C, Sotto A, Reynes J, Allavena C, Meyer L, Makinson A, Corbeau P. Immune profiles of pre-frail people living with HIV-1: a prospective longitudinal study. Immun Ageing 2024; 21:20. [PMID: 38481213 PMCID: PMC10935995 DOI: 10.1186/s12979-024-00416-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 02/06/2024] [Indexed: 03/17/2024]
Abstract
BACKGROUND People living with HIV (PLWH) are at risk of frailty, which is predictive for death. As an overactivity of the immune system is thought to fuel frailty, we characterized the immune activation profiles linked to frailty. METHODS We quantified twenty-seven activation markers in forty-six virological responders (four females and forty-two males; median age, 74 years; median duration of infection, 24 years; median duration of undetectability, 13 years), whose frailty was determined according to the Fried criteria. T cell and NK cell activation was evaluated by flow cytometry, using a panel of cell surface markers. Soluble markers of inflammation, and monocyte activation and endothelial activation were measured by ELISA. The participants' immune activation was profiled by an unsupervised double hierarchical clustering analysis. We used ANOVA p-values to rank immunomarkers most related to Fried score. A Linear Discriminant Analysis (LDA) was performed to link immune activation markers to frailty. RESULTS 41% of the participants were pre-frail, including 24% with a Fried score of 1, and 17% with a Fried score of 2. ANOVA identified the 14 markers of T cell, monocyte, NK cell, endothelial activation, and inflammation the most linked to Fried 3 classes. The LDA performed with these 14 markers was capable of discriminating volunteers according to their Fried score. Two out of the 5 immune activation profiles revealed by the hierarchical clustering were linked to and predictive of pre-frailty. These two profiles were characterized by a low percentage of CD4 T cells and a high percentage of CD8 T cells, activated CD4 T cells, CD8 T cells, and NK cells, and inflammation. CONCLUSIONS We identified a particular immune activation profile associated with pre-frailty in PLWH. Profiling participants at risk of developing frailty might help to tailor the screening and prevention of medical complications fueled by loss of robustness. Further studies will indicate whether this frailty signature is specific or not of HIV infection, and whether it also precedes frailty in the general population.
Collapse
Affiliation(s)
- Lucy Kundura
- Institute of Human Genetics, CNRS-Montpellier University UMR9002, 141 rue de la Cardonille, Montpellier, 34396, France
| | - Renaud Cezar
- Immunology Department, Nîmes University Hospital, Place du Pr Debré, Nîmes, 30029, France
| | - Sandrine Gimenez
- Institute of Human Genetics, CNRS-Montpellier University UMR9002, 141 rue de la Cardonille, Montpellier, 34396, France
| | - Manuela Pastore
- Institute of Functional Genomics UMR5203 and BCM, CNRS-INSERM-Montpellier University, 141 rue de la Cardonille, Montpellier, 34396, France
| | - Christelle Reynes
- Institute of Functional Genomics UMR5203 and BCM, CNRS-INSERM-Montpellier University, 141 rue de la Cardonille, Montpellier, 34396, France
| | - Albert Sotto
- Infectious and Tropical Diseases Department, Nîmes University Hospital, Nîmes, France
- Montpellier University, Montpellier, France
| | - Jacques Reynes
- Montpellier University, Montpellier, France
- Infectious and Tropical Diseases Department, Montpellier University Hospital, Montpellier, France
| | - Clotilde Allavena
- Service de Maladies Infectieuses, CHU de Nantes, Université de Nantes, Nantes, UE, 1413, France
| | - Laurence Meyer
- INSERM CESP U1018, Le Kremlin Bicêtre, France
- Department of Public Health and Epidemiology, Bicêtre Hospital, AP-HP, Paris-Saclay University, Le Kremlin-Bicêtre, France
| | - Alain Makinson
- Montpellier University, Montpellier, France
- Infectious and Tropical Diseases Department, Montpellier University Hospital, Montpellier, France
| | - Pierre Corbeau
- Institute of Human Genetics, CNRS-Montpellier University UMR9002, 141 rue de la Cardonille, Montpellier, 34396, France.
- Immunology Department, Nîmes University Hospital, Place du Pr Debré, Nîmes, 30029, France.
- Montpellier University, Montpellier, France.
| |
Collapse
|
21
|
Kiyak C, Ijezie OA, Ackah JA, Armstrong M, Cowen J, Cetinkaya D, Burianová H, Akudjedu TN. Topographical Distribution of Neuroanatomical Abnormalities Following COVID-19 Invasion : A Systematic Literature Review. Clin Neuroradiol 2024; 34:13-31. [PMID: 37697012 PMCID: PMC10881816 DOI: 10.1007/s00062-023-01344-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 08/14/2023] [Indexed: 09/13/2023]
Abstract
PURPOSE This systematic review is aimed at synthesising the literature base to date on the frequency and topographical distribution of neuroanatomical changes seen on imaging following COVID-19 invasion with a focus on both the acute and chronic phases of the disease. METHODS In this study, 8 databases were systematically searched to identify relevant articles published from December 2019 to March 2022 and supplemented with a manual reference search. Data were extracted from the included studies and narrative synthesis was employed to integrate the findings. RESULTS A total of 110 studies met the inclusion criteria and comprised 119,307 participants (including 31,073 acute and 143 long COVID-19 patients manifesting neurological alterations) and controls. Considerable variability in both the localisation and nature of neuroanatomical abnormalities are noted along the continuum with a wide range of neuropathologies relating to the cerebrovascular/neurovascular system, (sub)cortical structures (including deep grey and white matter structures), brainstem, and predominant regional and/or global alterations in the cerebellum with varying degrees of spinal involvement. CONCLUSION Structural regional alterations on neuroimaging are frequently demonstrated in both the acute and chronic phases of SARS-CoV‑2 infection, particularly prevalent across subcortical, prefrontal/frontal and cortico-limbic brain areas as well as the cerebrovascular/neurovascular system. These findings contribute to our understanding of the acute and chronic effects of the virus on the nervous system and has the potential to provide information on acute and long-term treatment and neurorehabilitation decisions.
Collapse
Affiliation(s)
- Ceyda Kiyak
- Faculty of Science and Technology, Bournemouth University, Bournemouth, UK
- School of Psychology, University of East Anglia, Norwich, UK
| | | | - Joseph A Ackah
- Institute of Medical Imaging and Visualisation, Faculty of Health and Social Sciences, Bournemouth University, 8 8GP, Bournemouth, UK
| | - Matthew Armstrong
- Department of Rehabilitation & Sports Sciences, Faculty of Health and Social Sciences, Bournemouth University, Bournemouth, UK
| | - Jake Cowen
- Department of Radiology, Queen Alexandra Hospital, Portsmouth Hospitals University NHS Trust, Portsmouth, UK
| | - Deniz Cetinkaya
- Faculty of Science and Technology, Bournemouth University, Bournemouth, UK
| | - Hana Burianová
- Faculty of Science and Technology, Bournemouth University, Bournemouth, UK
| | - Theophilus N Akudjedu
- Institute of Medical Imaging and Visualisation, Faculty of Health and Social Sciences, Bournemouth University, 8 8GP, Bournemouth, UK.
| |
Collapse
|
22
|
Stephan Y, Sutin AR, Luchetti M, Aschwanden D, Terracciano A. Personality and Cognition: The Mediating Role of Inflammatory Markers. J Gerontol B Psychol Sci Soc Sci 2024; 79:gbad152. [PMID: 37813576 PMCID: PMC10745263 DOI: 10.1093/geronb/gbad152] [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: 04/19/2023] [Indexed: 10/17/2023] Open
Abstract
OBJECTIVES Five-Factor Model personality traits are associated consistently with cognition. Inflammation has been hypothesized as a biological pathway in this association, but this assumption has yet to be tested. The present study tested inflammatory markers as mediators between personality traits and cognition. METHODS Participants were from the Health and Retirement Study (HRS; N = 4,364; 60% women; mean age = 64.48 years, standard deviation = 8.79). Personality traits and demographic factors were assessed in 2010/2012. Data on inflammatory markers (high-sensitivity C-reactive protein [hsCRP], interleukin-6 [IL-6], soluble tumor necrosis factor 1 (sTNFR1), interleukin-10 [IL-10], interleukin-1 receptor antagonist [IL-1Ra], and transforming growth factor [TGF]-β1) were obtained in 2016 from the HRS Venuous Blood Study. Cognition was assessed in 2020 using the modified Telephone Interview for Cognitive Status. RESULTS Higher neuroticism was related to lower cognition at follow-up, whereas higher extraversion, openness, agreeableness, and conscientiousness were associated with better cognition. Higher extraversion and higher conscientiousness were related to lower hsCRP, IL-6, IL-10, IL-1Ra, and sTNFR1, and higher openness was associated with lower IL-10, IL-1Ra, and sTNFR1 and to higher soluble TGF-β1. Lower sTNFR1 partially mediated the associations between conscientiousness, extraversion, and openness and cognition at follow-up, explaining an estimated 4%-12% of these associations. The mediating role of sTNFR1 persisted when physical activity and depressive symptoms were included as additional mediators. DISCUSSION The present study provides new evidence on personality and inflammatory markers. Consistent with the inflammation hypothesis, the sTNFR1 finding supports a potential biological pathway between personality and cognition.
Collapse
Affiliation(s)
| | - Angelina R Sutin
- Department of Behavioral Sciences and Social Medicine, College of Medicine, Florida State University, Tallahassee, Florida, USA
| | - Martina Luchetti
- Department of Behavioral Sciences and Social Medicine, College of Medicine, Florida State University, Tallahassee, Florida, USA
| | - Damaris Aschwanden
- Center for the Interdisciplinary Study of Gerontology and Vulnerability, University of Geneva, Geneva, Switzerland
- Department of Geriatrics, College of Medicine, Florida State University, Tallahassee, Florida, USA
| | - Antonio Terracciano
- Department of Geriatrics, College of Medicine, Florida State University, Tallahassee, Florida, USA
| |
Collapse
|
23
|
Hochuli N, Kadyan S, Park G, Patoine C, Nagpal R. Pathways linking microbiota-gut-brain axis with neuroinflammatory mechanisms in Alzheimer's pathophysiology. MICROBIOME RESEARCH REPORTS 2023; 3:9. [PMID: 38455083 PMCID: PMC10917618 DOI: 10.20517/mrr.2023.39] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 11/03/2023] [Accepted: 11/30/2023] [Indexed: 03/09/2024]
Abstract
Disturbances in the local and peripheral immune systems are closely linked to a wide range of diseases. In the context of neurodegenerative disorders such as Alzheimer's disease (AD), inflammation plays a crucial role, often appearing as a common manifestation despite the variability in the occurrence of other pathophysiological hallmarks. Thus, combating neuroinflammation holds promise in treating complex pathophysiological diseases like AD. Growing evidence suggests the gut microbiome's crucial role in shaping the pathogenesis of AD by influencing inflammatory mediators. Gut dysbiosis can potentially activate neuroinflammatory pathways through bidirectional signaling of the gut-brain axis; however, the precise mechanisms of this complex interweaved network remain largely unclear. In these milieus, this review attempts to summarize the contributing role of gut microbiome-mediated neuroinflammatory signals in AD pathophysiology, while also pondering potential mechanisms through which commensal and pathogenic gut microbes affect neuroinflammation. While certain taxa such as Roseburia and Escherichia have been strongly correlated with AD, other clades such as Bacteroides and Faecalibacterium exhibit variations at the species and strain levels. In order to disentangle the inflammatory aspects of neurodegeneration attributed to the gut microbiome, it is imperative that future mechanistic studies investigate the species/strain-level dependency of commensals, opportunistic, and pathogenic gut microbes that consistently show correlations with AD patients across multiple associative studies.
Collapse
Affiliation(s)
| | | | | | | | - Ravinder Nagpal
- Department of Health, Nutrition, and Food Sciences, College of Education, Health, and Human Sciences, Florida State University, Tallahassee, FL 32306, USA
| |
Collapse
|
24
|
Negrey JD, Frye BM, Johnson CSC, Kim J, Barcus RA, Lockhart SN, Whitlow CT, Sutphen C, Chiou KL, Snyder-Mackler N, Montine TJ, Craft S, Shively CA, Register TC. Mediterranean Diet Protects Against a Neuroinflammatory Cortical Transcriptome: Associations with Brain Volumetrics, Peripheral Inflammation, Social Isolation and Anxiety. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.01.565068. [PMID: 37961556 PMCID: PMC10635044 DOI: 10.1101/2023.11.01.565068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
INTRODUCTION Mediterranean diets may be neuroprotective and prevent cognitive decline relative to Western diets, however the underlying biology is poorly understood. METHODS We assessed the effects of Western vs. Mediterranean-like diets on RNAseq generated transcriptional profiles in temporal cortex and their relationships with changes in MRI neuroimaging phenotypes, circulating monocyte gene expression, and observations of social isolation and anxiety in 38 socially-housed, middle-aged female cynomolgus macaques. RESULTS Diet resulted in differential expression of seven transcripts (FDR<0.05). Cyclin dependent kinase 14 ( CDK14 ), a proinflammatory regulator, was lower in the Mediterranean group. The remaining six transcripts [i.e., "lunatic fringe" ( LFNG ), mannose receptor C type 2 ( MRC2 ), solute carrier family 3 member 2 ( SLCA32 ), butyrophilin subfamily 2 member A1 ( BTN2A1 ), katanin regulatory subunit B1 ( KATNB1 ), and transmembrane protein 268 ( TMEM268 )] were higher in cortex of the Mediterranean group and generally associated with anti-inflammatory/neuroprotective pathways. KATNB1 encodes a subcomponent of katanin, important in maintaining microtubule homeostasis. BTN2A1 is involved in immunomodulation of γδ T-cells which have anti-neuroinflammatory and neuroprotective effects. CDK14 , LFNG , MRC2, and SLCA32 are associated with inflammatory pathways. The latter four differentially expressed cortex transcripts were associated with monocyte transcript levels, changes in AD-relevant brain volumes determined by MRI over the course of the study, and social isolation and anxiety. CDK14 was positively correlated with monocyte inflammatory transcripts, changes in total brain, gray matter, cortical gray matter volumes, and time alone and anxious behavior, and negatively correlated with changes in total white matter and cerebrospinal fluid (CSF) volumes. In contrast, LFNG , MRC2 , and SLCA32 were negatively correlated with monocyte inflammatory transcripts and changes in total gray matter volume, and positively correlated with CSF volume changes, and SLCA32 was negatively correlated with time alone. DISCUSSION Collectively, our results suggest that relative to Western diets, Mediterranean diets confer protection against peripheral and central inflammation which is reflected in preserved brain structure and behavior.
Collapse
|
25
|
Midlife Systemic Inflammatory Markers Are Associated With Late-Life Brain Volume: The ARIC Study. Neurology 2023; 101:375. [PMID: 37604669 PMCID: PMC10449445 DOI: 10.1212/wnl.0000000000207420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2023] Open
|
26
|
Shi Y, Lin F, Li Y, Wang Y, Chen X, Meng F, Ye Q, Cai G. Association of pro-inflammatory diet with increased risk of all-cause dementia and Alzheimer's dementia: a prospective study of 166,377 UK Biobank participants. BMC Med 2023; 21:266. [PMID: 37480061 PMCID: PMC10362711 DOI: 10.1186/s12916-023-02940-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 06/13/2023] [Indexed: 07/23/2023] Open
Abstract
BACKGROUND Increasing evidence suggests an association between pro-inflammatory diets and cognitive function. However, only a few studies based on small sample sizes have explored the association between pro-inflammatory diets and dementia using the dietary inflammatory index (DII). Additionally, the relationship between DII and different subtypes of dementia, such as Alzheimer's dementia and vascular dementia, remains largely unexplored. Given the changes in brain structure already observed in patients with dementia, we also investigated the association between DII and magnetic resonance imaging (MRI) measures of brain structure to provide some hints to elucidate the potential mechanisms between pro-inflammatory diet and cognitive decline. METHODS A total of 166,377 UK Biobank participants without dementia at baseline were analyzed. DII calculations were based on the information collected by the 24-h recall questionnaire. Brain structural anatomy and tissue-specific volumes were measured using brain MRI. Cox proportional hazards models, competing risk models, and restricted cubic spline were applied to assess the longitudinal associations. The generalized linear model was used to assess the association between DII and MRI measurements. RESULTS During a median follow-up time of 9.46 years, a total of 1372 participants developed dementia. The incidence of all-cause dementia increased by 4.6% for each additional unit of DII [hazard ratio (HR): 1.046]. Besides, DII displayed a "J-shaped" non-linear association with Alzheimer's dementia (Pnonlinear = 0.003). When DII was above 1.30, an increase in DII was significantly associated with an increased risk of Alzheimer's dementia (HR: 1.391, 95%CI: 1.085-1.784, P = 0.009). For brain MRI, the total volume of white matter hyperintensities increased with an increase in DII, whereas the volume of gray matter in the hippocampus decreased. CONCLUSIONS In this cohort study, higher DII was associated with a higher risk of all-cause dementia and Alzheimer's dementia. However, our findings suggested that the association with DII and vascular and frontotemporal dementia was not significant.
Collapse
Affiliation(s)
- Yisen Shi
- Department of Neurology, Center for Cognitive Neurology, Institute of Clinical Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, China
- Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 88 Jiaotong Road, Fuzhou, 350001, China
| | - Fabin Lin
- Department of Neurology, Center for Cognitive Neurology, Institute of Clinical Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, China
- Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 88 Jiaotong Road, Fuzhou, 350001, China
- Department of Neurosurgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Yueping Li
- Department of Neurology, Center for Cognitive Neurology, Institute of Clinical Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, China
- Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 88 Jiaotong Road, Fuzhou, 350001, China
| | - Yingqing Wang
- Department of Neurology, Center for Cognitive Neurology, Institute of Clinical Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, China
- Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 88 Jiaotong Road, Fuzhou, 350001, China
| | - Xiaochun Chen
- Department of Neurology, Center for Cognitive Neurology, Institute of Clinical Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, China
- Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 88 Jiaotong Road, Fuzhou, 350001, China
| | - Fangang Meng
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100050, China.
| | - Qinyong Ye
- Department of Neurology, Center for Cognitive Neurology, Institute of Clinical Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, China.
- Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, China.
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 88 Jiaotong Road, Fuzhou, 350001, China.
| | - Guoen Cai
- Department of Neurology, Center for Cognitive Neurology, Institute of Clinical Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, China.
- Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, China.
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 88 Jiaotong Road, Fuzhou, 350001, China.
| |
Collapse
|
27
|
Zhang W, Xiao D, Mao Q, Xia H. Role of neuroinflammation in neurodegeneration development. Signal Transduct Target Ther 2023; 8:267. [PMID: 37433768 PMCID: PMC10336149 DOI: 10.1038/s41392-023-01486-5] [Citation(s) in RCA: 410] [Impact Index Per Article: 205.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 03/22/2023] [Accepted: 05/07/2023] [Indexed: 07/13/2023] Open
Abstract
Studies in neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease and Amyotrophic lateral sclerosis, Huntington's disease, and so on, have suggested that inflammation is not only a result of neurodegeneration but also a crucial player in this process. Protein aggregates which are very common pathological phenomenon in neurodegeneration can induce neuroinflammation which further aggravates protein aggregation and neurodegeneration. Actually, inflammation even happens earlier than protein aggregation. Neuroinflammation induced by genetic variations in CNS cells or by peripheral immune cells may induce protein deposition in some susceptible population. Numerous signaling pathways and a range of CNS cells have been suggested to be involved in the pathogenesis of neurodegeneration, although they are still far from being completely understood. Due to the limited success of traditional treatment methods, blocking or enhancing inflammatory signaling pathways involved in neurodegeneration are considered to be promising strategies for the therapy of neurodegenerative diseases, and many of them have got exciting results in animal models or clinical trials. Some of them, although very few, have been approved by FDA for clinical usage. Here we comprehensively review the factors affecting neuroinflammation and the major inflammatory signaling pathways involved in the pathogenicity of neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, and Amyotrophic lateral sclerosis. We also summarize the current strategies, both in animal models and in the clinic, for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Weifeng Zhang
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, 199 South Chang'an Road, Xi'an, 710062, P.R. China
| | - Dan Xiao
- The State Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Air Force Medical University, No. 169 Changle West Road, Xi'an, 710032, P.R. China
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Air Force Medical University, No. 169 Changle West Road, Xi'an, 710032, China
| | - Qinwen Mao
- Department of Pathology, University of Utah, Huntsman Cancer Institute, 2000 Circle of Hope Drive, Salt Lake City, UT, 84112, USA
| | - Haibin Xia
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, 199 South Chang'an Road, Xi'an, 710062, P.R. China.
| |
Collapse
|
28
|
França DCH, Fujimori M, de Queiroz AA, Borges MD, Magalhães Neto AM, de Camargos PJV, Ribeiro EB, França EL, Honorio-França AC, Fagundes-Triches DLG. Melatonin and Cytokines Modulate Daily Instrumental Activities of Elderly People with SARS-CoV-2 Infection. Int J Mol Sci 2023; 24:ijms24108647. [PMID: 37239991 DOI: 10.3390/ijms24108647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/03/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
The Comprehensive Geriatric Assessment analyzes the health and quality of life of the elderly. Basic and instrumental daily activities may be compromised due to neuroimmunoendocrine changes, and studies suggest that possible immunological changes occur during infections in the elderly. Thus, this study aimed to analyze cytokine and melatonin levels in serum and correlate the Comprehensive Geriatric Assessment in elderly patients with SARS-CoV-2 infection. The sample consisted of 73 elderly individuals, 43 of whom were without infection and 30 of whom had positive diagnoses of COVID-19. Blood samples were collected to quantify cytokines by flow cytometry and melatonin by ELISA. In addition, structured and validated questionnaires were applied to assess basic (Katz) and instrumental (Lawton and Brody) activities. There was an increase in IL-6, IL-17, and melatonin in the group of elderly individuals with infection. In addition, a positive correlation was observed between melatonin and IL-6 and IL-17 in elderly patients with SARS-CoV-2 infection. Furthermore, there was a reduction in the score of the Lawton and Brody Scale in the infected elderly. These data suggest that the melatonin hormone and inflammatory cytokines are altered in the serum of the elderly with SARS-CoV-2 infection. In addition, there is a degree of dependence, mainly regarding the performance of daily instrumental activities, in the elderly. The considerable impact on the elderly person's ability to perform everyday tasks necessary for independent living is an extremely important result, and changes in cytokines and melatonin probably are associated with alterations in these daily activities of the elderly.
Collapse
Affiliation(s)
| | - Mahmi Fujimori
- Biological and Health Sciences Institute, Federal University of Mato Grosso, Barra do Garças 78605-091, Mato Grosso, Brazil
| | - Adriele Ataídes de Queiroz
- Biological and Health Sciences Institute, Federal University of Mato Grosso, Barra do Garças 78605-091, Mato Grosso, Brazil
| | - Maraísa Delmut Borges
- Biological and Health Sciences Institute, Federal University of Mato Grosso, Barra do Garças 78605-091, Mato Grosso, Brazil
| | - Aníbal Monteiro Magalhães Neto
- Biological and Health Sciences Institute, Federal University of Mato Grosso, Barra do Garças 78605-091, Mato Grosso, Brazil
| | | | - Elton Brito Ribeiro
- Health Sciences Institute, Federal University of Mato Grosso, Sinop 78557-287, Mato Grosso, Brazil
| | - Eduardo Luzía França
- Biological and Health Sciences Institute, Federal University of Mato Grosso, Barra do Garças 78605-091, Mato Grosso, Brazil
| | | | | |
Collapse
|
29
|
Qi X, Ng TKS, Wu B. Sex differences in the mediating role of chronic inflammation on the association between social isolation and cognitive functioning among older adults in the United States. Psychoneuroendocrinology 2023; 149:106023. [PMID: 36603408 PMCID: PMC10105626 DOI: 10.1016/j.psyneuen.2023.106023] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/25/2022] [Accepted: 01/01/2023] [Indexed: 01/04/2023]
Abstract
BACKGROUND Previous research has reported the association between social isolation and cognitive impairment. However, biological mechanisms underlying this association are understudied. It is also unclear whether there are sex differences in these biological mechanisms. OBJECTIVES To examine whether chronic inflammation biomarkers are potential mediators of the association between social isolation and cognitive functioning among older men and women. METHODS Data were the National Health and Nutrition Examination Survey 1999-2002. A total of 2535 older adults aged 60 and older were included. Chronic inflammation was measured by C-reactive protein (CRP), plasma fibrinogen, and serum albumin. Cognitive functioning was assessed by the Digit Symbol Substitution Test (DSST). Social isolation was defined using a 4-point composite index of items pertaining to the strength of social network and support. Linear regression models and formal mediation analysis were applied. RESULTS Social isolation was associated with lower DSST scores [β (SE) = -2.445 (1.180), p < 0.01 for men; β (SE) = -5.478 (1.167), p < 0.001 for women]. For older men, social isolation was associated with higher levels of CRP (β [SE] = 0.226 (0.110), p < 0.05) and fibrinogen (β [SE] = 0.058 (0.026), p < 0.05). In mediation analyses, among older men, CRP mediated 6.1% and fibrinogen mediated 12.0% of the association of social isolation with DSST. CONCLUSION Social isolation was associated with poorer cognitive functioning partially via heightened inflammatory responses in older men. Defining these associations' mechanisms in sex-specific contexts could inform preventive and therapeutic strategies for cognitive impairment in older adults.
Collapse
Affiliation(s)
- Xiang Qi
- Rory Meyers College of Nursing, New York University, New York, NY, United States
| | - Ted Kheng Siang Ng
- Edson College of Nursing and Health Innovation, Arizona State University, Phoenix, AZ, United States
| | - Bei Wu
- Rory Meyers College of Nursing, New York University, New York, NY, United States.
| |
Collapse
|
30
|
Cano A, Esteban-de-Antonio E, Bernuz M, Puerta R, García-González P, de Rojas I, Olivé C, Pérez-Cordón A, Montrreal L, Núñez-Llaves R, Sotolongo-Grau Ó, Alarcón-Martín E, Valero S, Alegret M, Martín E, Martino-Adami PV, Ettcheto M, Camins A, Vivas A, Gomez-Chiari M, Tejero MÁ, Orellana A, Tárraga L, Marquié M, Ramírez A, Martí M, Pividori MI, Boada M, Ruíz A. Plasma extracellular vesicles reveal early molecular differences in amyloid positive patients with early-onset mild cognitive impairment. J Nanobiotechnology 2023; 21:54. [PMID: 36788617 PMCID: PMC9930227 DOI: 10.1186/s12951-023-01793-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 01/24/2023] [Indexed: 02/16/2023] Open
Abstract
In the clinical course of Alzheimer's disease (AD) development, the dementia phase is commonly preceded by a prodromal AD phase, which is mainly characterized by reaching the highest levels of Aβ and p-tau-mediated neuronal injury and a mild cognitive impairment (MCI) clinical status. Because of that, most AD cases are diagnosed when neuronal damage is already established and irreversible. Therefore, a differential diagnosis of MCI causes in these prodromal stages is one of the greatest challenges for clinicians. Blood biomarkers are emerging as desirable tools for pre-screening purposes, but the current results are still being analyzed and much more data is needed to be implemented in clinical practice. Because of that, plasma extracellular vesicles (pEVs) are gaining popularity as a new source of biomarkers for the early stages of AD development. To identify an exosome proteomics signature linked to prodromal AD, we performed a cross-sectional study in a cohort of early-onset MCI (EOMCI) patients in which 184 biomarkers were measured in pEVs, cerebrospinal fluid (CSF), and plasma samples using multiplex PEA technology of Olink© proteomics. The obtained results showed that proteins measured in pEVs from EOMCI patients with established amyloidosis correlated with CSF p-tau181 levels, brain ventricle volume changes, brain hyperintensities, and MMSE scores. In addition, the correlations of pEVs proteins with different parameters distinguished between EOMCI Aβ( +) and Aβ(-) patients, whereas the CSF or plasma proteome did not. In conclusion, our findings suggest that pEVs may be able to provide information regarding the initial amyloidotic changes of AD. Circulating exosomes may acquire a pathological protein signature of AD before raw plasma, becoming potential biomarkers for identifying subjects at the earliest stages of AD development.
Collapse
Affiliation(s)
- Amanda Cano
- Ace Alzheimer Center Barcelona - International University of Catalunya (UIC), C/Marquès de Sentmenat, 57, 08029, Barcelona, Spain.
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.
| | - Ester Esteban-de-Antonio
- Ace Alzheimer Center Barcelona - International University of Catalunya (UIC), C/Marquès de Sentmenat, 57, 08029, Barcelona, Spain
| | - Mireia Bernuz
- Grup de Sensors I Biosensors, Departament de Química, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Raquel Puerta
- Ace Alzheimer Center Barcelona - International University of Catalunya (UIC), C/Marquès de Sentmenat, 57, 08029, Barcelona, Spain
| | - Pablo García-González
- Ace Alzheimer Center Barcelona - International University of Catalunya (UIC), C/Marquès de Sentmenat, 57, 08029, Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Itziar de Rojas
- Ace Alzheimer Center Barcelona - International University of Catalunya (UIC), C/Marquès de Sentmenat, 57, 08029, Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Claudia Olivé
- Ace Alzheimer Center Barcelona - International University of Catalunya (UIC), C/Marquès de Sentmenat, 57, 08029, Barcelona, Spain
| | - Alba Pérez-Cordón
- Ace Alzheimer Center Barcelona - International University of Catalunya (UIC), C/Marquès de Sentmenat, 57, 08029, Barcelona, Spain
| | - Laura Montrreal
- Ace Alzheimer Center Barcelona - International University of Catalunya (UIC), C/Marquès de Sentmenat, 57, 08029, Barcelona, Spain
| | - Raúl Núñez-Llaves
- Ace Alzheimer Center Barcelona - International University of Catalunya (UIC), C/Marquès de Sentmenat, 57, 08029, Barcelona, Spain
| | - Óscar Sotolongo-Grau
- Ace Alzheimer Center Barcelona - International University of Catalunya (UIC), C/Marquès de Sentmenat, 57, 08029, Barcelona, Spain
| | - Emilio Alarcón-Martín
- Ace Alzheimer Center Barcelona - International University of Catalunya (UIC), C/Marquès de Sentmenat, 57, 08029, Barcelona, Spain
| | - Sergi Valero
- Ace Alzheimer Center Barcelona - International University of Catalunya (UIC), C/Marquès de Sentmenat, 57, 08029, Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Montserrat Alegret
- Ace Alzheimer Center Barcelona - International University of Catalunya (UIC), C/Marquès de Sentmenat, 57, 08029, Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Elvira Martín
- Ace Alzheimer Center Barcelona - International University of Catalunya (UIC), C/Marquès de Sentmenat, 57, 08029, Barcelona, Spain
| | - Pamela V Martino-Adami
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937, Cologne, Germany
| | - Miren Ettcheto
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028, Barcelona, Spain
- Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| | - Antonio Camins
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028, Barcelona, Spain
- Institute of Neurosciences, University of Barcelona, Barcelona, Spain
| | - Assumpta Vivas
- Departament de Diagnòstic Per La Imatge, Clínica Corachan, Barcelona, Spain
| | - Marta Gomez-Chiari
- Departament de Diagnòstic Per La Imatge, Clínica Corachan, Barcelona, Spain
| | | | - Adelina Orellana
- Ace Alzheimer Center Barcelona - International University of Catalunya (UIC), C/Marquès de Sentmenat, 57, 08029, Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Lluís Tárraga
- Ace Alzheimer Center Barcelona - International University of Catalunya (UIC), C/Marquès de Sentmenat, 57, 08029, Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Marta Marquié
- Ace Alzheimer Center Barcelona - International University of Catalunya (UIC), C/Marquès de Sentmenat, 57, 08029, Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Alfredo Ramírez
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937, Cologne, Germany
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Medical Faculty, 53127, Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), 53127, Bonn, Germany
- Department of Psychiatry and Glenn, Biggs Institute for Alzheimer's and Neurodegenerative Diseases, San Antonio, TX, 78229, USA
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931, Cologne, Germany
| | - Mercè Martí
- Grup de Sensors I Biosensors, Departament de Química, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - María Isabel Pividori
- Grup de Sensors I Biosensors, Departament de Química, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
- Biosensing and Bioanalysis Group, Institut de Biotecnologia I de Biomedicina (IBB-UAB), Mòdul B Parc de Recerca UAB, Campus Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Mercè Boada
- Ace Alzheimer Center Barcelona - International University of Catalunya (UIC), C/Marquès de Sentmenat, 57, 08029, Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Agustín Ruíz
- Ace Alzheimer Center Barcelona - International University of Catalunya (UIC), C/Marquès de Sentmenat, 57, 08029, Barcelona, Spain.
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.
| |
Collapse
|
31
|
Farina MP, Kim JK, Hayward MD, Crimmins EM. Links between inflammation and immune functioning with cognitive status among older Americans in the Health and Retirement Study. Brain Behav Immun Health 2022; 26:100559. [PMID: 36439057 PMCID: PMC9694056 DOI: 10.1016/j.bbih.2022.100559] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 11/11/2022] [Indexed: 11/15/2022] Open
Abstract
Elevated inflammation and poor immune functioning are tied to worse cognitive health. Both processes are fundamental to aging and are strongly implicated in the development of age-related health outcomes, including cognitive status. However, results from prior studies evaluating links between indicators of inflammation and immune function and cognitive impairment have been inconsistent due to biomarker selection, sample selection, and cognitive outcome. Using the Health and Retirement Study (HRS), a nationally representative study of older adults in the United States, we assessed how indicators of inflammation (neutrophil-lymphocyte ratio (NLR), albumin, CRP, IL6, IL10, IL-1Ra, sTNFR1, and TGFβ1) and immune functioning (CMV, CD4+ TN/TM, and CD8+ TN/TM) are associated with cognitive status. First, to examine the association between each biomarker and cognitive status, we tested whether markers of inflammation and immune functioning varied across cognitive status categories. We found that dementia and cognitive impairment without dementia (CIND) were associated with elevated inflammation and poorer immune functioning across biomarkers except for CD4+ TN/TM. Next, we estimated multinomial logistic regression models to assess which biomarkers would continue to be associated with dementia and CIND, net of each other. In these models, albumin, cytokines, CMV, CD4+ TN/TM, and CD8+ TN/TM are associated with cognitive status. Because poor immune functioning and increased inflammation are associated with cognitive impairment, improving immune functioning and reducing inflammation may provide a mechanism for reducing ADRD risk in the population.
Collapse
Affiliation(s)
- Mateo P. Farina
- Leonard Davis School of Gerontology, University of Southern California, USA
| | - Jung Ki Kim
- Leonard Davis School of Gerontology, University of Southern California, USA
| | - Mark D. Hayward
- Population Research Center and Department of Sociology, University of Texas at Austin, USA
| | - Eileen M. Crimmins
- Leonard Davis School of Gerontology, University of Southern California, USA
| |
Collapse
|
32
|
Xi X, Han L. Exploring the relationship between novel Coronavirus pneumonia and Parkinson's disease. Medicine (Baltimore) 2022; 101:e31813. [PMID: 36401405 PMCID: PMC9678520 DOI: 10.1097/md.0000000000031813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
The hypothesis is that there is 0a relationship between Parkinson's disease and coronavirus disease 2019 (COVID-19). By summarizing the pathogenesis of Parkinson's disease and COVID-19 and the impact of COVID-19 on the central nervous system, the relationship between Parkinson's disease and COVID-19 was analyzed, including whether Parkinson's disease is a predisposition factor for COVID-19 and whether COVID-19 causes the occurrence of Parkinson's disease. Discuss the impact of COVID-19 on patients with Parkinson's disease, including symptoms and life impact. To summarize the principles, goals and methods of home rehabilitation for Parkinson's disease patients during COVID-19. Through the analysis of this paper, it is believed that COVID-19 may cause Parkinson's disease. Parkinson's disease has the condition of susceptibility to COVID-19, but this conclusion is still controversial.
Collapse
Affiliation(s)
- Xiaoming Xi
- Rehabilitation Center,Beijing Rehabilitation Hospital Affiliated to Capital Medical University, Beijing
- * Correspondence: Xiaoming Xi, Beijing Rehabilitation Hospital Affiliated to Capital Medical University, No.15, Badachu Xixizhuang, Shijingshan District, Beijing (e-mail: )
| | - Liang Han
- Shandong University of Traditional Chinese Medicine
| |
Collapse
|
33
|
Bouazzaoui W, Xiao P, Couve‐Bonnaire S, Bouillon J, Mulengi JK. Chronic Inflammation and Chronic Diseases: Potential Healing with Glutathione‐Inspired Fragments. ChemistrySelect 2022. [DOI: 10.1002/slct.202203051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Wafaa Bouazzaoui
- Laboratory of Organic Chemistry Natural Products and Analysis University of Tlemcen P.O. BOX 117 Tlemcen 13 000 Algeria
| | - Pan Xiao
- Normandie Université COBRA, UMR 6014 et FR 3038 INSA Rouen, CNRS Université de Rouen 1, Rue Tesnière 76821 Mont Saint-Aignan Cedex France
| | - Samuel Couve‐Bonnaire
- Normandie Université COBRA, UMR 6014 et FR 3038 INSA Rouen, CNRS Université de Rouen 1, Rue Tesnière 76821 Mont Saint-Aignan Cedex France
| | - Jean‐Philippe Bouillon
- Normandie Université COBRA, UMR 6014 et FR 3038 INSA Rouen, CNRS Université de Rouen 1, Rue Tesnière 76821 Mont Saint-Aignan Cedex France
| | - Joseph Kajima Mulengi
- Department of Chemistry Faculty of Sciences Faculty of Sciences University of Tlemcen P.O. Box 119 13000 Tlemcen Algeria
| |
Collapse
|
34
|
De Vlieger L, Vandenbroucke RE, Van Hoecke L. Recent insights into viral infections as a trigger and accelerator in alzheimer's disease. Drug Discov Today 2022; 27:103340. [PMID: 35987492 PMCID: PMC9385395 DOI: 10.1016/j.drudis.2022.103340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 07/08/2022] [Accepted: 08/12/2022] [Indexed: 11/16/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder for which only symptomatic medication is available, except for the recently FDA-approved aducanumab. This lack of effective treatment urges us to investigate alternative paths that might contribute to disease development. In light of the recent SARS-CoV-2 pandemic and the disturbing neurological complications seen in some patients, it is desirable to (re)investigate the viability of the viral infection theory claiming that a microbe could affect AD initiation and/or progression. Here, we review the most important evidence for this theory with a special focus on two viruses, namely HSV-1 and SARS-CoV-2. Moreover, we discuss the possible involvement of extracellular vesicles (EVs). This overview will contribute to a more rational approach of potential treatment strategies for AD patients.
Collapse
Affiliation(s)
- Lize De Vlieger
- Barriers in Inflammation Lab, VIB Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Roosmarijn E Vandenbroucke
- Barriers in Inflammation Lab, VIB Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
| | - Lien Van Hoecke
- Barriers in Inflammation Lab, VIB Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| |
Collapse
|
35
|
Sun Y, Geng J, Chen X, Chen H, Wang X, Chen J, Li X, Hesketh T. Association Between Inflammatory Bowel Disease and Dementia: A Longitudinal Cohort Study. Inflamm Bowel Dis 2022; 28:1520-1526. [PMID: 34849925 PMCID: PMC9527613 DOI: 10.1093/ibd/izab300] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND The association between inflammatory bowel disease (IBD) and dementia remains uncertain. We aim to investigate whether IBD is associated with higher dementia risk. METHODS Using multivariable Cox regression models, we analyzed the onset of all-cause dementia among 497,775 participants, including 5778 IBD patients in the UK Biobank as primary analysis. In secondary analysis, we further examined the difference in brain structure and cognitive function changes between IBD and non-IBD individuals. The diagnosis of IBD and dementia was confirmed with combination of primary care data, hospital inpatient data, death registry, and self-report data. Brain structure was measured by brain MRI as anatomic and tissue-specific volumes; cognitive function was tested in terms of reaction, visual episodic memory, verbal-numerical reasoning, and prospective memory. RESULTS During a mean follow-up of 11.58 years, 100 and 6709 incident all-cause dementia with or without IBD were documented, respectively. In multivariable Cox regression model, hazard ratio for incident dementia among IBD patients was 1.14 (95% confidence interval [CI], 0.94-1.39; P=.182) comparing with non-IBD participants; no statistically significant difference was observed in their brain MRI measures of anatomic and tissue-specific volumes, whereas IBD patients had a significantly increased reaction time (β=12.32; 95% CI, 1.97, 22.67; P = .020). Results of subgroup and sensitivity analyses were consistent with the main analysis. CONCLUSIONS Our study does not support a significant association between IBD and dementia. Further studies with better design and longer follow-up are needed to elucidate the association.
Collapse
Affiliation(s)
- Yuhao Sun
- Center for Global Health, Zhejiang University, Hangzhou, China
| | - Jiawei Geng
- Center for Global Health, Zhejiang University, Hangzhou, China
| | - Xuejie Chen
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Hui Chen
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoyan Wang
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Jie Chen
- Center for Global Health, Zhejiang University, Hangzhou, China
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Xue Li
- School of Public Health and the Second Affiliated Hospital, Zhejiang University, Hangzhou, China
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Therese Hesketh
- Center for Global Health, Zhejiang University, Hangzhou, China
- Institute for Global Health, University College London, London, UK
| |
Collapse
|
36
|
Magyari A, Ye M, Margolis DJ, McCulloch CE, Cummings SR, Yaffe K, Langan SM, Abuabara K. Adult atopic eczema and the risk of dementia: A population-based cohort study. J Am Acad Dermatol 2022; 87:314-322. [PMID: 35367295 DOI: 10.1016/j.jaad.2022.03.049] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 03/03/2022] [Accepted: 03/09/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Chronic inflammatory conditions have been linked to dementia, but little is known about the role of atopic eczema, an inflammatory condition recently recognized to be common among older adults. OBJECTIVE To determine whether active atopic eczema is associated with incident dementia. METHODS A longitudinal cohort study of 1,767,667 individuals aged 60 to 99 years registered with The Health Improvement Network, a primary care cohort in the United Kingdom. The diagnoses of atopic eczema and dementia were identified using medical record codes. RESULTS The incidence of dementia was 57 per 10,000 person-years among those with atopic eczema during follow-up (12.1% of the population) compared with 44 per 10,000 person-years in the control group. This translated to a 27% increased risk of dementia (hazard ratio, 1.27; 95% CI, 1.23-1.30) in adjusted Cox proportional hazard models. Similar associations were observed in subgroup analyses of vascular dementia and Alzheimer's disease. The association persisted after additionally adjusting for the use of systemic corticosteroids (hazard ratio, 1.29; 95% CI, 1.26-1.33) and potential mediators (hazard ratio, 1.19; 95% CI, 1.16-1.22). More severe eczema was associated with a higher risk of dementia. LIMITATIONS Lack of detailed data on severity. CONCLUSION Atopic eczema was associated with a small but increased risk of incident dementia. The association increased with the severity of atopic eczema.
Collapse
Affiliation(s)
- Alexa Magyari
- UC Berkeley School of Public Health, Berkeley, California
| | - Morgan Ye
- Department of Dermatology, University of California, San Francisco School of Medicine (UCSF), San Francisco, California
| | - David J Margolis
- Department of Dermatology and Center for Epidemiology and Biostatistics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Charles E McCulloch
- Division of Biostatistics, University of California, San Francisco School of Medicine (UCSF), San Francisco, California
| | - Steven R Cummings
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, California; Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California
| | - Kristine Yaffe
- Center for Population Brain Health, University of California, San Francisco School of Medicine (UCSF), San Francisco, California
| | - Sinéad M Langan
- Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, Keppel Street, London, United Kingdom
| | - Katrina Abuabara
- UC Berkeley School of Public Health, Berkeley, California; Department of Dermatology, University of California, San Francisco School of Medicine (UCSF), San Francisco, California.
| |
Collapse
|
37
|
Wang Y, Li Y, Liu K, Han X, Dong Y, Wang X, Wang M, Cong L, Zhang Q, Tang S, Hou T, Liu C, Song L, Han X, Vetrano DL, Du Y, Qiu C. Non-alcoholic fatty liver disease, serum cytokines, and dementia among rural-dwelling older adults in China: a population-based study. Eur J Neurol 2022; 29:2612-2621. [PMID: 35608965 DOI: 10.1111/ene.15416] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/13/2022] [Accepted: 05/19/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Little is known about whether non-alcoholic fatty liver disease (NAFLD) is associated with dementia as well as the role of serum pro-inflammatory cytokines in the association. We aimed to investigate the interrelationships of NAFLD, serum cytokines, and dementias among rural-dwelling older adults. METHODS This population-based cross-sectional study included 5,129 participants (age ≥60 years; 61.79% women) who were living in rural communities and examined in March-September 2018. NAFLD was defined through transabdominal ultrasound examination in the absence of hepatitis B or excessive alcohol consumption. Serum cytokines were measured in a subsample (n=1,686). Dementia, Alzheimer's disease (AD), and vascular dementia (VaD) were diagnosed following international criteria. Data were analyzed with logistic regression and mediation models. RESULTS Of the 5,129 participants, 455 (8.87%) were detected with moderate-to-severe NAFLD and 292 (5.69%) were diagnosed with dementia (188 with AD and 96 with VaD). The multivariable-adjusted odds ratios (ORs) associated with moderate-to-severe (vs. no-to-mild) NAFLD were 2.22 (95% confidence interval, 1.41-3.49) for all-cause dementia, 1.88 (1.01-3.50) for AD, and 2.62 (1.33-5.17) for VaD. In the cytokine subsample, controlling for multiple potential confounders, moderate-to-severe NAFLD was significantly associated with higher levels of serum monocyte chemotactic protein-1 (MCP-1), interleukin-17A (IL-17A), interleukin-6 (IL-6), interleukin-8 (IL-8), and tumor necrosis factor-α (TNF-α) (P<0.05). The mediation analysis showed that IL-6 mediated 12.56% of the association between NAFLD and VaD. CONCLUSIONS Moderate-to-severe NAFLD is associated with dementia and AD, especially with VaD, among rural-dwelling Chinese older adults, in which the association with VaD is partly mediated by serum inflammatory cytokines.
Collapse
Affiliation(s)
- Yongxiang Wang
- Department of Neurology, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China.,Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, P.R. China.,Shandong Provincial Clinical Research Center for Neurological Diseases, Jinan, Shandong, P.R. China
| | - Yuanjing Li
- Aging Research Center, Department of NVS, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Keke Liu
- Department of Neurology, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China.,Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, P.R. China.,Shandong Provincial Clinical Research Center for Neurological Diseases, Jinan, Shandong, P.R. China
| | - Xiaolei Han
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, P.R. China.,Shandong Provincial Clinical Research Center for Neurological Diseases, Jinan, Shandong, P.R. China
| | - Yi Dong
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, P.R. China.,Shandong Provincial Clinical Research Center for Neurological Diseases, Jinan, Shandong, P.R. China
| | - Xiaojie Wang
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, P.R. China.,Shandong Provincial Clinical Research Center for Neurological Diseases, Jinan, Shandong, P.R. China
| | - Mingqi Wang
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, P.R. China.,Shandong Provincial Clinical Research Center for Neurological Diseases, Jinan, Shandong, P.R. China
| | - Lin Cong
- Department of Neurology, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China.,Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, P.R. China.,Shandong Provincial Clinical Research Center for Neurological Diseases, Jinan, Shandong, P.R. China
| | - Qinghua Zhang
- Department of Neurology, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China.,Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, P.R. China.,Shandong Provincial Clinical Research Center for Neurological Diseases, Jinan, Shandong, P.R. China
| | - Shi Tang
- Department of Neurology, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China.,Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, P.R. China.,Shandong Provincial Clinical Research Center for Neurological Diseases, Jinan, Shandong, P.R. China
| | - Tingting Hou
- Department of Neurology, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China.,Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, P.R. China.,Shandong Provincial Clinical Research Center for Neurological Diseases, Jinan, Shandong, P.R. China
| | - Cuicui Liu
- Department of Neurology, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China.,Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, P.R. China.,Shandong Provincial Clinical Research Center for Neurological Diseases, Jinan, Shandong, P.R. China
| | - Lin Song
- Department of Neurology, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China.,Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, P.R. China.,Shandong Provincial Clinical Research Center for Neurological Diseases, Jinan, Shandong, P.R. China
| | - Xiaojuan Han
- Department of Neurology, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China.,Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, P.R. China.,Shandong Provincial Clinical Research Center for Neurological Diseases, Jinan, Shandong, P.R. China
| | - Davide L Vetrano
- Aging Research Center, Department of NVS, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Yifeng Du
- Department of Neurology, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China.,Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, P.R. China.,Shandong Provincial Clinical Research Center for Neurological Diseases, Jinan, Shandong, P.R. China
| | - Chengxuan Qiu
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, P.R. China.,Aging Research Center, Department of NVS, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| |
Collapse
|
38
|
Liu M, Li D, Hong X, Sun Z. Increased Risk for Dementia in Patients With Inflammatory Bowel Disease: A Systematic Review and Meta-Analysis of Population-Based Studies. Front Neurol 2022; 13:813266. [PMID: 35645979 PMCID: PMC9135967 DOI: 10.3389/fneur.2022.813266] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 04/13/2022] [Indexed: 12/19/2022] Open
Abstract
Background Mounting evidence suggests that there may be a causal relationship or common pathogenic pathway between inflammatory bowel disease (IBD) and dementia. However, inconsistent results have emerged from epidemiological studies. We therefore conducted this review to clarify the relationship between IBD and dementia. Methods We systematically searched PubMed, Web of Science, Embase, and Cochrane library to identify all studies exploring the relationship between IBD and dementia published as of September 2021. Risk estimates were pooled using both fixed and random-effects models. Results Six studies involving 2,334,472 subjects were included. Pooled results suggested that the risk of developing dementia significantly increased after IBD diagnosis (HR = 1.27, 95% CI: 1.10-1.47, P = 0.001), which did not vary by age, gender, dementia subtype, or IBD subtype. Whereas, the dementia incidence before IBD diagnosis and the comorbidity rate of dementia in IBD patients were similar to those without IBD (HR = 0.92, 95% CI: 0.68-1.25; 0.82, 95% CI: 0.64-1.06, respectively). However, current evidence was insufficient to establish a causal relationship. Conclusion This study shows an unidirectional association between IBD and dementia; patients with IBD have an increased risk of dementia, and it may be beneficial to develop individualized dementia screening strategies for this population. Future research needs to further investigate whether effective therapies of IBD can reduce this risk and pathophysiological mechanisms of the association.
Collapse
Affiliation(s)
- Mengsi Liu
- Hengyang Medical School, University of South China, Hengyang, China
| | - Dongxiu Li
- Department of Nursing, Fujian Health College, Fuzhou, China
| | - Xia Hong
- Department of Nursing, Fujian Health College, Fuzhou, China
| | - Zhen Sun
- Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
39
|
Li S, An N, Chen N, Wang Y, Yang L, Wang Y, Yao Z, Hu B. The impact of Alzheimer's disease susceptibility loci on lateral ventricular surface morphology in older adults. Brain Struct Funct 2022; 227:913-924. [PMID: 35028746 DOI: 10.1007/s00429-021-02429-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 11/13/2021] [Indexed: 11/25/2022]
Abstract
The enlargement of ventricular volume is a general trend in the elderly, especially in patients with Alzheimer's disease (AD). Multiple susceptibility loci have been reported to have an increased risk for AD and the morphology of brain structures are affected by the variations in the risk loci. Therefore, we hypothesized that genes contributed significantly to the ventricular surface, and the changes of ventricular surface were associated with the impairment of cognitive functions. After the quality controls (QC) and genotyping, a lateral ventricular segmentation method was employed to obtain the surface features of lateral ventricle. We evaluated the influence of 18 selected AD susceptibility loci on both volume and surface morphology across 410 subjects from Alzheimer's Disease Neuroimaging Initiative (ADNI). Correlations were conducted between radial distance (RD) and Montreal Cognitive Assessment (MoCA) subscales. Only the C allele at the rs744373 loci in BIN1 gene significantly accelerated the atrophy of lateral ventricle, including the anterior horn, body, and temporal horn of left lateral ventricle. No significant effect on lateral ventricle was found at other loci. Our results revealed that most regions of the bilateral ventricular surface were significantly negatively correlated with cognitive scores, particularly in delayed recall. Besides, small areas of surface were negatively correlated with language, orientation, and visuospatial scores. Together, our results indicated that the genetic variation affected the localized areas of lateral ventricular surface, and supported that lateral ventricle was an important brain structure associated with cognition in the elderly.
Collapse
Affiliation(s)
- Shan Li
- Gansu Provincial Key Laboratory of Wearable Computing, School of Information Science and Engineering, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, 730000, Gansu Province, People's Republic of China
| | - Na An
- Gansu Provincial Key Laboratory of Wearable Computing, School of Information Science and Engineering, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, 730000, Gansu Province, People's Republic of China
| | - Nan Chen
- Gansu Provincial Key Laboratory of Wearable Computing, School of Information Science and Engineering, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, 730000, Gansu Province, People's Republic of China
| | - Yin Wang
- Gansu Provincial Key Laboratory of Wearable Computing, School of Information Science and Engineering, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, 730000, Gansu Province, People's Republic of China
| | - Lin Yang
- Gansu Provincial Key Laboratory of Wearable Computing, School of Information Science and Engineering, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, 730000, Gansu Province, People's Republic of China
| | - Yalin Wang
- School of Computing, Informatics, and Decision Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Zhijun Yao
- Gansu Provincial Key Laboratory of Wearable Computing, School of Information Science and Engineering, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, 730000, Gansu Province, People's Republic of China.
| | - Bin Hu
- Gansu Provincial Key Laboratory of Wearable Computing, School of Information Science and Engineering, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, 730000, Gansu Province, People's Republic of China.
- CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, ShangHai, China.
- Joint Research Center for Cognitive Neurosensor Technology of Lanzhou University and Institute of Semiconductors, Chinese Academy of Sciences, LanZhou, China.
- Engineering Research Center of Open Source Software and Real-Time System, Ministry of Education, Lanzhou University, Lanzhou, China.
| |
Collapse
|
40
|
de Asteasu MLS, Cuevas-Lara C, García-Hermoso A, Ramírez-Vélez R, Martínez-Velilla N, Zambom-Ferraresi F, Cadore EL, Izquierdo M. Effects of Physical Exercise on the Incidence of Delirium and Cognitive Function in Acutely Hospitalized Older Adults: A Systematic Review with Meta-Analysis. J Alzheimers Dis 2022; 87:503-517. [PMID: 35275553 DOI: 10.3233/jad-220103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Acute care hospitalization increases the likelihood of developing cognitive impairment and delirium in older adults. OBJECTIVE To summarize evidence about the effectiveness of exercise and physical rehabilitation interventions on the incidence of delirium and cognitive impairment in acutely hospitalized older patients. METHODS Relevant articles were systematically searched (PubMed, Web of Science, and CINHAL databases) until 26 August 2021. Randomized and nonrandomized controlled trials of in-hospital physical exercise interventions and rehabilitation programs compared to usual care performed for older patients (> 65 years) hospitalized for an acute medical condition were selected. The primary endpoints were changes in the incidence of delirium and cognition during acute hospitalization. The secondary endpoints included functional independence, psychological measures, well-being status, length of hospital stay, transfer after discharge, fall occurrence, hospital readmissions, and mortality rate. The endpoints were evaluated at different time points (at admission, at discharge, and after discharge). RESULTS Eleven studies from 8 trials (n = 3,646) were included. The methodological quality of the studies was mostly high. None of the studies reported any adverse events related to the intervention. Early rehabilitation improved cognitive function at 3 months postdischarge (Hedge's g = 0.33, 95% confidence interval [CI] 0.19 to 0.46, p < 0.001). No between-group differences were found for incident delirium and cognitive impairment during hospitalization (all p > 0.05). CONCLUSION In-hospital physical exercise and early rehabilitation programs seem to be safe and effective interventions for enhancing cognitive function after discharge in older patients hospitalized for an acute medical condition. However, no potential benefits were obtained over usual hospital care for the incidence of delirium.
Collapse
Affiliation(s)
- Mikel L Sáez de Asteasu
- Navarrabiomed, Hospital Universitario de Navarra (HUN)-Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain.,CIBER of Frailty and Healthy Aging (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Cesar Cuevas-Lara
- Navarrabiomed, Hospital Universitario de Navarra (HUN)-Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain
| | - Antonio García-Hermoso
- Navarrabiomed, Hospital Universitario de Navarra (HUN)-Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain
| | - Robinson Ramírez-Vélez
- Navarrabiomed, Hospital Universitario de Navarra (HUN)-Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain
| | - Nicolás Martínez-Velilla
- Navarrabiomed, Hospital Universitario de Navarra (HUN)-Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain.,CIBER of Frailty and Healthy Aging (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain.,Geriatric Department, Hospital Universitario de Navarra (HUN), Pamplona, Navarra, Spain
| | - Fabricio Zambom-Ferraresi
- Navarrabiomed, Hospital Universitario de Navarra (HUN)-Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain.,CIBER of Frailty and Healthy Aging (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Mikel Izquierdo
- Navarrabiomed, Hospital Universitario de Navarra (HUN)-Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain.,CIBER of Frailty and Healthy Aging (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
41
|
Connolly MG, Bruce SR, Kohman RA. Exercise duration differentially effects age-related neuroinflammation and hippocampal neurogenesis. Neuroscience 2022; 490:275-286. [PMID: 35331843 PMCID: PMC9038708 DOI: 10.1016/j.neuroscience.2022.03.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 03/10/2022] [Accepted: 03/16/2022] [Indexed: 12/14/2022]
Abstract
The physiological effects of exercise vary as a function of frequency and length. However, research on the duration-dependent effects of exercise has focused primarily on young adults and less is known about the influence of exercise duration in the aged. The current study compared the effects of short-term and long-term running wheel access on hippocampal neurogenesis and neuroimmune markers in aged (19-23 months) male C57BL/6J mice. Aged mice were given 24-hour access to a running wheel for 14 days (short-term) or 51 days (long-term). Groups of non-running aged and young (5 months) mice served as comparison groups to detect age-related differences and effects of exercise. Long-term, but not short-term, exercise increased hippocampal neurogenesis as assessed by number of doublecortin (DCX) positive cells in the granular cell layer. Assessment of cytokines, receptors, and glial-activation markers showed the expected age-related increase compared to young controls. In the aged, exercise as a function of duration regulated select aspects of the neuroimmune profile. For instance, hippocampal expression of interleukin (IL)-10 was increased only following long-term exercise. While in contrast brain levels of IL-6 were reduced by both short- and long-term exercise. Additional findings showed that exercise does not modulate all aspects of age-related neuroinflammation and/or may have differential effects in hippocampal compared to brain samples. Overall, the data indicate that increasing exercise duration produces more robust effects on immune modulation and hippocampal neurogenesis.
Collapse
Affiliation(s)
- Meghan G Connolly
- University of Illinois Urbana-Champaign, Department of Animal Sciences, Champaign, IL, USA.
| | - Spencer R Bruce
- University of North Carolina Wilmington, Department of Psychology, Wilmington, NC, USA.
| | - Rachel A Kohman
- University of North Carolina Wilmington, Department of Psychology, Wilmington, NC, USA.
| |
Collapse
|
42
|
Wu-Chung EL, Leal SL, Denny BT, Cheng SL, Fagundes CP. Spousal caregiving, widowhood, and cognition: A systematic review and a biopsychosocial framework for understanding the relationship between interpersonal losses and dementia risk in older adulthood. Neurosci Biobehav Rev 2022; 134:104487. [PMID: 34971701 PMCID: PMC8925984 DOI: 10.1016/j.neubiorev.2021.12.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 11/23/2021] [Accepted: 12/06/2021] [Indexed: 01/18/2023]
Abstract
Accumulating research suggests that stressful life events, especially those that threaten close intimate bonds, are associated with an increased risk of dementia. Grieving the loss of a spouse, whether in the form of caregiving or after the death, ranks among 'life's most significant stressors', evoking intense psychological and physiological distress. Despite numerous studies reporting elevated dementia risk or poorer cognition among spousal caregivers and widow(er)s compared to controls, no review has summarized findings across cognitive outcomes (i.e., dementia incidence, cognitive impairment rates, cognitive performance) or proposed a theoretical model for understanding the links between partner loss and abnormal cognitive decline. The current systematic review summarizes findings across 64 empirical studies. Overall, both cross-sectional and longitudinal studies revealed an adverse association between partner loss and cognitive outcomes. In turn, we propose a biopsychosocial model of cognitive decline that explains how caregiving and bereavement may position some to develop cognitive impairment or Alzheimer's disease and related dementias. More longitudinal studies that focus on the biopsychosocial context of caregivers and widow(er)s are needed.
Collapse
Affiliation(s)
- E Lydia Wu-Chung
- Department of Psychological Sciences, Rice University, Houston, TX, United States.
| | - Stephanie L Leal
- Department of Psychological Sciences, Rice University, Houston, TX, United States
| | - Bryan T Denny
- Department of Psychological Sciences, Rice University, Houston, TX, United States
| | - Samantha L Cheng
- Department of Psychological Sciences, Rice University, Houston, TX, United States
| | - Christopher P Fagundes
- Department of Psychological Sciences, Rice University, Houston, TX, United States; Department of Behavioral Science, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; Department of Psychiatry & Behavioral Sciences, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
43
|
Goikolea J, Gerenu G, Daniilidou M, Mangialasche F, Mecocci P, Ngandu T, Rinne J, Solomon A, Kivipelto M, Cedazo-Minguez A, Sandebring-Matton A, Maioli S. Serum Thioredoxin-80 is associated with age, ApoE4, and neuropathological biomarkers in Alzheimer's disease: a potential early sign of AD. Alzheimers Res Ther 2022; 14:37. [PMID: 35209952 PMCID: PMC8876266 DOI: 10.1186/s13195-022-00979-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 02/16/2022] [Indexed: 12/12/2022]
Abstract
Background Thioredoxin-80 (Trx80) is a cleavage product from the redox-active protein Thioredoxin-1 and has been previously described as a pro-inflammatory cytokine secreted by immune cells. Previous studies in our group reported that Trx80 levels are depleted in Alzheimer’s disease (AD) brains. However, no studies so far have investigated peripheral Trx80 levels in the context of AD pathology and whether could be associated with the main known AD risk factors and biomarkers. Methods Trx80 was measured in serum samples from participants from two different cohorts: the observational memory clinic biobank (GEDOC) (N = 99) with AD CSF biomarker data was available and the population-based lifestyle multidomain intervention trial Finnish Geriatric Intervention Study to Prevent Cognitive Impairment and Disability (FINGER) (N = 47), with neuroimaging data and blood markers of inflammation available. The GEDOC cohort consists of participants diagnosed with subjective cognitive impairment (SCI), mild cognitive impairment (MCI), and AD, whereas the FINGER participants are older adults at-risk of dementia, but without substantial cognitive impairment. One-way ANOVA and multiple comparison tests were used to assess the levels of Trx80 between groups. Linear regression models were used to explore associations of Trx80 with cognition, AD CSF biomarkers (Aβ42, t-tau, p-tau and p-tau/t-tau ratio), inflammatory cytokines, and neuroimaging markers. Results In the GEDOC cohort, Trx80 was associated to p-tau/t-tau ratio in the MCI group. In the FINGER cohort, serum Trx80 levels correlated with lower hippocampal volume and higher pro-inflammatory cytokine levels. In both GEDOC and FINGER cohorts, ApoE4 carriers had significantly higher serum Trx80 levels compared to non-ApoE4 carriers. However, Trx80 levels in the brain were further decreased in AD patients with ApoE4 genotype. Conclusion We report that serum Trx80 levels are associated to AD disease stage as well as to several risk factors for AD such as age and ApoE4 genotype, which suggests that Trx80 could have potential as serum AD biomarker. Increased serum Trx80 and decreased brain Trx80 levels was particularly seen in ApoE4 carriers. Whether this could contribute to the mechanism by which ApoE4 show increased vulnerability to develop AD would need to be further investigated. Trial registration ClinicalTrials.govNCT01041989. Registered on 4 January 2010—retrospectively registered Supplementary Information The online version contains supplementary material available at 10.1186/s13195-022-00979-9.
Collapse
Affiliation(s)
- Julen Goikolea
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden.
| | - Gorka Gerenu
- Biodonostia Health Research Institute, Neuroscience Area, 20014, Donostia-San Sebastián, Gipuzkoa, Spain.,CIBERNED (Network Center for Biomedical Research in Neurodegenerative Diseases), Carlos III Institute, Madrid, Spain.,Department of Physiology, Medicine and Nursing School, University of Basque Country UPV/EHU, Leioa, Spain
| | - Makrina Daniilidou
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden.,Department of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Francesca Mangialasche
- Department of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Patrizia Mecocci
- Department of Medicine and Surgery, Santa Maria della Misericordia Hospital, Section of Gerontology and Geriatrics, University of Perugia, Perugia, Italy
| | - Tiia Ngandu
- Department of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden.,Department of Public Health Solutions, Public Health Promotion Unit, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Juha Rinne
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland
| | - Alina Solomon
- Department of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden.,Institute of Clinical Medicine/Neurology, University of Eastern Finland, Kuopio, Finland.,Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, London, UK
| | - Miia Kivipelto
- Department of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden.,Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, London, UK.,Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland.,Theme Aging, Karolinska University Hospital, Stockholm, Sweden
| | - Angel Cedazo-Minguez
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Anna Sandebring-Matton
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden.,Department of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden.,Institute of Clinical Medicine/Neurology, University of Eastern Finland, Kuopio, Finland
| | - Silvia Maioli
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
44
|
Wittfeld K, Raman MR, Conner SC, Aslam A, Teumer A, Nauck M, Hosten N, Habes M, DeCarli C, Vasan RS, Beiser AS, Himali JJ, Seshadri S, Grabe HJ, Satizabal CL. Insulin-Like Growth Factor, Inflammation, and MRI Markers of Alzheimer's Disease in Predominantly Middle-Aged Adults. J Alzheimers Dis 2022; 88:311-322. [PMID: 35599493 PMCID: PMC9472289 DOI: 10.3233/jad-220356] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Insulin-like growth factor 1 (IGF-1) signaling has been implicated in Alzheimer's disease pathogenesis, and further evidence suggests inflammation can be a moderator of this association. However, most research to date has been conducted on older adults. OBJECTIVE To investigate the association of serum IGF-1 and IGF binding protein 3 (IGFBP-3) concentrations with MRI markers of Alzheimer's disease in predominantly middle-aged adults, and further assess moderation by chronic inflammation. METHODS We included participants from the Framingham Heart Study (n = 1,852, mean age 46±8, 46% men) and the Study of Health in Pomerania (n = 674, mean age 50±13, 42% men) with available serum IGF-1, IFGBP-3, as well as brain MRI. IGF-1 and IFGBP-3 were related to MRI outcomes (i.e., total brain, cortical gray matter, white matter, white matter hyperintensities (WMH), and hippocampal volumes) using multivariable regression models adjusting for potential confounders. Subgroup analyses by C-reactive protein (CRP) concentrations were also performed. Cohort-specific summary statistics were meta-analyzed using random-effects models and corrected for multiple comparisons. RESULTS Meta-analysis results revealed that higher IGF-1 concentrations were associated with lower WMH (estimate [β] [95% CI], -0.05 [-0.09, -0.02], p = 0.006) and larger hippocampal volumes (0.07 [0.02, 0.12], p = 0.01), independent of vascular risk factors. These associations occurred predominantly in individuals with CRP concentrations < 75th percentile. We did not observe associations between IGFBP-3 and MRI outcomes. CONCLUSION Our findings suggest that IGF-1-related signaling may be implicated in brain health as early as midlife.
Collapse
Affiliation(s)
- Katharina Wittfeld
- German Center for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, Germany
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Mekala R Raman
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Framingham Heart Study, Framingham, MA, USA
| | - Sarah C Conner
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Asra Aslam
- Long School of Medicine, UT Health San Antonio, San Antonio, TX, USA
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Bialystok, Bialystok, Poland
| | - Matthias Nauck
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Norbert Hosten
- Institute of Diagnostic Radiology and Neuroradiology, University Medicine Greifswald, Greifswald, Germany
| | - Mohamad Habes
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX, USA
- Department of Radiology, Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurology, Penn Memory Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Charles DeCarli
- Department of Neurology, University of California, Davis School of Medicine, Sacramento, CA, USA
| | - Ramachandran S Vasan
- Framingham Heart Study, Framingham, MA, USA
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston MA, USA
| | - Alexa S Beiser
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Framingham Heart Study, Framingham, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Jayandra J Himali
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Framingham Heart Study, Framingham, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX, USA
| | - Sudha Seshadri
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Framingham Heart Study, Framingham, MA, USA
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX, USA
| | - Hans J Grabe
- German Center for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, Germany
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Claudia L Satizabal
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Framingham Heart Study, Framingham, MA, USA
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX, USA
| |
Collapse
|
45
|
Kim REY, Abbott RD, Kim S, Thomas RJ, Yun CH, Kim H, Johnson H, Shin C. Sleep Duration, Sleep Apnea, and Gray Matter Volume. J Geriatr Psychiatry Neurol 2022; 35:47-56. [PMID: 33511901 DOI: 10.1177/0891988720988918] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
This study aimed to evaluate the effect of sleep duration on brain structures in the presence versus absence of sleep apnea in middle-aged and older individuals. The study investigated a population-based sample of 2,560 individuals, aged 49-80 years. The presence of sleep apnea and self-reported sleep duration were examined in relation to gray matter volume (GMV) in total and lobar brain regions. We identified ranges of sleep duration associated with maximal GMV using quadratic regression and bootstrap sampling. A significant quadratic association between sleep duration and GMV was observed in total and lobar brain regions of men with sleep apnea. In the fully adjusted model, optimal sleep durations associated with peak GMV between brain regions ranged from 6.7 to 7.0 hours. Shorter and longer sleep durations were associated with lower GMV in total and 4 sub-regions of the brain in men with sleep apnea.
Collapse
Affiliation(s)
- Regina Eun Young Kim
- Institute of Human Genomic Study, Korea University College of Medicine, Ansan City, South Korea.,Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Robert Douglas Abbott
- Institute of Human Genomic Study, Korea University College of Medicine, Ansan City, South Korea
| | - Soriul Kim
- Institute of Human Genomic Study, Korea University College of Medicine, Ansan City, South Korea
| | - Robert Joseph Thomas
- Department of Neurology, Seoul National University Bundang Hospital, Seongnam-si Gyeonggi-do, South Korea
| | - Chang-Ho Yun
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, Beth Israel Deaconess Medical Center and the Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA
| | - Hyun Kim
- Department of Clinical Psychology, Boston University, Boston, MA, USA
| | - Hans Johnson
- Department of Electrical and Computer Engineering, University of Iowa, Iowa City, IA, USA
| | - Chol Shin
- Institute of Human Genomic Study, Korea University College of Medicine, Ansan City, South Korea
| |
Collapse
|
46
|
Akbarialiabad H, Taghrir MH, Abdollahi A, Ghahramani N, Kumar M, Paydar S, Razani B, Mwangi J, Asadi-Pooya AA, Malekmakan L, Bastani B. Long COVID, a comprehensive systematic scoping review. Infection 2021; 49:1163-1186. [PMID: 34319569 PMCID: PMC8317481 DOI: 10.1007/s15010-021-01666-x] [Citation(s) in RCA: 199] [Impact Index Per Article: 49.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 07/10/2021] [Indexed: 02/07/2023]
Abstract
PURPOSE To find out what is known from literature about Long COVID until January 30, 2021. METHODS We undertook a four-step search with no language restriction. A preliminary search was made to identify the keywords. A search strategy of all electronic databases resulted in 66 eligible studies. A forward and backward search of the references and citations resulted in additional 54 publications. Non-English language articles were translated using Google Translate. We conducted our scoping review based on the PRISMA-ScR Checklist. RESULTS Of 120 papers, we found only one randomized clinical trial. Of the 67 original studies, 22 were cohort, and 28 were cross-sectional studies. Of the total 120 publications, 49.1% focused on signs and symptoms, 23.3% on management, and 10.8% on pathophysiology. Ten publications focused on imaging studies. The results are also presented extensively in a narrative synthesis in separated sections (nomenclature, diagnosis, pathophysiology, risk factors, signs/symptoms, management). CONCLUSIONS The controversies in its definition have impaired proper recognition and management. The predominant symptoms were: fatigue, breathlessness, arthralgia, sleep difficulties, and chest pain. Recent reports also point to the risk of long-term sequela with cutaneous, respiratory, cardiovascular, musculoskeletal, mental health, neurologic, and renal involvement in those who survive the acute phase of the illness.
Collapse
Affiliation(s)
- Hossein Akbarialiabad
- Student Research Committee, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Hossein Taghrir
- Student Research Committee, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ashkan Abdollahi
- Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nasrollah Ghahramani
- Division of Nephrology, Department of Medicine, Penn State University College of Medicine, Hershey, PA, 17033, USA
| | - Manasi Kumar
- Department of Psychiatry, University of Nairobi, Nairobi, Kenya
- Department of Clinical, Educational and Health Psychology, University College London, London, UK
| | - Shahram Paydar
- Trauma Research Center, Shahid Rajaee (Emtiaz) Trauma Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Babak Razani
- Cardiology Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
- John Cochran Division, Veterans Affairs St. Louis Healthcare System, St. Louis, MO, 63106, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - John Mwangi
- Pulmonary and Critical Care Medicine, Saint Louis University School of Medicine, Saint Louis, MO, USA
| | - Ali A Asadi-Pooya
- Epilepsy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Neurology, Jefferson Comprehensive Epilepsy Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Leila Malekmakan
- Shiraz Nephro-Urology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bahar Bastani
- Professor of Medicine-Nephrology, Saint Louis University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
47
|
Machhi J, Yeapuri P, Lu Y, Foster E, Chikhale R, Herskovitz J, Namminga KL, Olson KE, Abdelmoaty MM, Gao J, Quadros RM, Kiyota T, Jingjing L, Kevadiya BD, Wang X, Liu Y, Poluektova LY, Gurumurthy CB, Mosley RL, Gendelman HE. CD4+ effector T cells accelerate Alzheimer's disease in mice. J Neuroinflammation 2021; 18:272. [PMID: 34798897 PMCID: PMC8603581 DOI: 10.1186/s12974-021-02308-7] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 10/28/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by pathological deposition of misfolded self-protein amyloid beta (Aβ) which in kind facilitates tau aggregation and neurodegeneration. Neuroinflammation is accepted as a key disease driver caused by innate microglia activation. Recently, adaptive immune alterations have been uncovered that begin early and persist throughout the disease. How these occur and whether they can be harnessed to halt disease progress is unclear. We propose that self-antigens would induct autoreactive effector T cells (Teffs) that drive pro-inflammatory and neurodestructive immunity leading to cognitive impairments. Here, we investigated the role of effector immunity and how it could affect cellular-level disease pathobiology in an AD animal model. METHODS In this report, we developed and characterized cloned lines of amyloid beta (Aβ) reactive type 1 T helper (Th1) and type 17 Th (Th17) cells to study their role in AD pathogenesis. The cellular phenotype and antigen-specificity of Aβ-specific Th1 and Th17 clones were confirmed using flow cytometry, immunoblot staining and Aβ T cell epitope loaded haplotype-matched major histocompatibility complex II IAb (MHCII-IAb-KLVFFAEDVGSNKGA) tetramer binding. Aβ-Th1 and Aβ-Th17 clones were adoptively transferred into APP/PS1 double-transgenic mice expressing chimeric mouse/human amyloid precursor protein and mutant human presenilin 1, and the mice were assessed for memory impairments. Finally, blood, spleen, lymph nodes and brain were harvested for immunological, biochemical, and histological analyses. RESULTS The propagated Aβ-Th1 and Aβ-Th17 clones were confirmed stable and long-lived. Treatment of APP/PS1 mice with Aβ reactive Teffs accelerated memory impairment and systemic inflammation, increased amyloid burden, elevated microglia activation, and exacerbated neuroinflammation. Both Th1 and Th17 Aβ-reactive Teffs progressed AD pathology by downregulating anti-inflammatory and immunosuppressive regulatory T cells (Tregs) as recorded in the periphery and within the central nervous system. CONCLUSIONS These results underscore an important pathological role for CD4+ Teffs in AD progression. We posit that aberrant disease-associated effector T cell immune responses can be controlled. One solution is by Aβ reactive Tregs.
Collapse
Affiliation(s)
- Jatin Machhi
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Pravin Yeapuri
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Yaman Lu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Emma Foster
- Department of Biological Sciences, Northern Kentucky University, Highland Heights, KY 41099 USA
| | - Rupesh Chikhale
- University College London School of Pharmacy, Bloomsbury, London, WC1E 6DE UK
| | - Jonathan Herskovitz
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Krista L. Namminga
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Katherine E. Olson
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Mai Mohamed Abdelmoaty
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198 USA
- Therapeutic Chemistry Department, National Research Centre, Giza, Egypt
| | - Ju Gao
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Rolen M. Quadros
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198 USA
- Mouse Genome Engineering Core Facility, University of Nebraska Medical Center, Omaha, NE USA
| | - Tomomi Kiyota
- Department of Safety Assessment, Genentech Inc., South San Francisco, CA 94080 USA
| | - Liang Jingjing
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Bhavesh D. Kevadiya
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Xinglong Wang
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Yutong Liu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198 USA
- Department of Radiology, University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Larisa Y. Poluektova
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Channabasavaiah B. Gurumurthy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198 USA
- Mouse Genome Engineering Core Facility, University of Nebraska Medical Center, Omaha, NE USA
| | - R. Lee Mosley
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198 USA
| | - Howard E. Gendelman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198 USA
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198 USA
| |
Collapse
|
48
|
Vemuri P, Davey C, Johansen KL, Zuk SM, Reid RI, Thostenson KB, Reddy AL, Jack CR, Knopman DS, Murray AM. Chronic Kidney Disease Associated with Worsening White Matter Disease and Ventricular Enlargement. J Alzheimers Dis 2021; 83:1729-1740. [PMID: 34459402 PMCID: PMC8609691 DOI: 10.3233/jad-210604] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Chronic kidney disease (CKD), a growing public health issue in the elderly, is associated with increased risk of cognitive impairment. OBJECTIVE To investigate the mechanisms through which CKD impacts brain health using longitudinal imaging. METHODS We identified 97 participants (74 CKD and 23 non-CKD) from the BRINK (BRain IN Kidney Disease), a longitudinal study of CKD with two MRI scans (baseline and 3-year follow-up). We measured the associations between baseline and change in kidney disease biomarkers of estimated glomerular filtration rate (eGFR) and urinary albumin to creatinine ratio (UACR), considered a measure of microvascular inflammation, and imaging outcomes of cortical thickness and ventricular volume from structural MRI, white matter hyperintensities (WMH) volume from FLAIR images, and fractional anisotropy of the corpus callosum (FACC). RESULTS There were white matter-specific changes as observed by increased WMH volume and decreased FACC in CKD participants, as well as ventricular volume increase in both CKD and non-CKD groups reflective of aging-related changes. Decline in eGFR was associated with decrease in the FACC, suggesting that subtle early white matter changes due to kidney disease can be captured using DTI. An increase in UACR was associated with increase in ventricular volume. CONCLUSION Our results support the role of eGFR as a measure of kidney microvascular disease which is associated with concurrent white matter damage in CKD. Future work is needed to investigate the possible link between endothelial microvascular inflammation (as measured by an increased UACR) and ventricular volume increase.
Collapse
Affiliation(s)
| | - Cynthia Davey
- Biostatistical Design and Analysis Center, University of Minnesota Clinical and Translational Science Institute, Minneapolis, MN, USA
| | - Kirsten L Johansen
- Department of Internal Medicine, Nephrology Division, Hennepin Healthcare, Minneapolis, MN, USA.,United States Renal Data Systems, NIDDK, Bethesda, MD, USA
| | - Samantha M Zuk
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Robert I Reid
- Department of Information Technology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | - Anne M Murray
- Berman Center for Clinical Research and Outcomes, Hennepin Healthcare Research Institute, Minneapolis, MN, USA.,Department of Internal Medicine, Geriatrics Division, Hennepin Healthcare, Minneapolis, MN, USA.,Departments of Medicine and Neurology, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
49
|
Muzambi R, Bhaskaran K, Smeeth L, Brayne C, Chaturvedi N, Warren-Gash C. Assessment of common infections and incident dementia using UK primary and secondary care data: a historical cohort study. THE LANCET. HEALTHY LONGEVITY 2021; 2:e426-e435. [PMID: 34240064 PMCID: PMC8245326 DOI: 10.1016/s2666-7568(21)00118-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Common infections have been associated with dementia risk; however, evidence is scarce. We aimed to investigate the association between common infections and dementia in adults (≥65 years) in a UK population-based cohort study. METHODS We did a historical cohort study of individuals who were 65 years and older with no history of dementia or cognitive impairment using the Clinical Practice Research Datalink linked to Hospital Episode Statistics between Jan 1, 2004, and Dec 31, 2018. Multivariable Cox proportional hazard regression models were used to estimate the association between time-updated previous common infections (sepsis, pneumonia, other lower respiratory tract infections, urinary tract infections, and skin and soft tissue infections) and incident dementia diagnosis. We also tested for effect modification by diabetes since it is an independent risk factor for dementia and co-occurs with infection. FINDINGS Between Jan 1, 2004, and Dec 31, 2018, our study included 989 800 individuals (median age 68·6 years [IQR 65·0-77·0]; 537 602 [54·3%] women) of whom 402 204 (40·6%) were diagnosed with at least one infection and 56 802 (5·7%) had incident dementia during a median follow-up of 5·2 years (IQR 2·3-9·0). Dementia risk increased in those with any infection (adjusted hazard ratio [HR] 1·53 [95% CI 1·50-1·55]) compared with those without infection. HRs were highest for sepsis (HR 2·08 [1·89-2·29]) and pneumonia (HR 1·88 [1·77-1·99]) and for infections leading to hospital admission (1·99 [1·94-2·04]). HRs were also higher in individuals with diabetes compared with those without diabetes. INTERPRETATION Common infections, particularly those resulting in hospitalisation, were associated with an increased risk of dementia persisting over the long term. Whether reducing infections lowers the risk of subsequent dementia warrants evaluation. FUNDING Alzheimer's Society, Wellcome Trust, and the Royal Society.
Collapse
Affiliation(s)
- Rutendo Muzambi
- Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK
| | - Krishnan Bhaskaran
- Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK
| | - Liam Smeeth
- Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK
| | - Carol Brayne
- Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK
| | - Nish Chaturvedi
- Medical Research Council Unit for Lifelong Health and Ageing at University College London, Institute of Cardiovascular Science, University College London, London, UK
| | - Charlotte Warren-Gash
- Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK
| |
Collapse
|
50
|
Alexaki VI. The Impact of Obesity on Microglial Function: Immune, Metabolic and Endocrine Perspectives. Cells 2021; 10:cells10071584. [PMID: 34201844 PMCID: PMC8307603 DOI: 10.3390/cells10071584] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 06/18/2021] [Accepted: 06/22/2021] [Indexed: 02/06/2023] Open
Abstract
Increased life expectancy in combination with modern life style and high prevalence of obesity are important risk factors for development of neurodegenerative diseases. Neuroinflammation is a feature of neurodegenerative diseases, and microglia, the innate immune cells of the brain, are central players in it. The present review discusses the effects of obesity, chronic peripheral inflammation and obesity-associated metabolic and endocrine perturbations, including insulin resistance, dyslipidemia and increased glucocorticoid levels, on microglial function.
Collapse
Affiliation(s)
- Vasileia Ismini Alexaki
- Institute for Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, TU Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
| |
Collapse
|