1
|
Bell TR, Franz CE, Thomas KR, Williams ME, Eyler LT, Lerman I, Fennema-Notestine C, Puckett OK, Dorros SM, Panizzon MS, Pearce RC, Hagler DJ, Lyons MJ, Elman JA, Kremen WS. Elevated C-Reactive Protein in Older Men With Chronic Pain: Association With Plasma Amyloid Levels and Hippocampal Volume. J Gerontol A Biol Sci Med Sci 2024; 79:glae206. [PMID: 39169831 PMCID: PMC11439493 DOI: 10.1093/gerona/glae206] [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: 03/25/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND Chronic pain leads to tau accumulation and hippocampal atrophy, which may be moderated through inflammation. In older men, we examined associations of chronic pain with Alzheimer's disease (AD)-related plasma biomarkers and hippocampal volume as moderated by systemic inflammation. METHODS Participants were men without dementia. Chronic pain was defined as moderate-to-severe pain in 2+ study waves at average ages 56, 62, and 68. At age 68, we measured plasma amyloid-beta (Aβ42, n = 871), Aβ40 (n = 887), total tau (t-tau, n = 841), and neurofilament light chain (NfL, n = 915), and serum high-sensitivity C-reactive protein (hs-CRP, n = 968), a marker of systemic inflammation. A subgroup underwent structural MRI to measure hippocampal volume (n = 385). Analyses adjusted for medical morbidities, depressive symptoms, and opioid use. RESULTS Chronic pain was related to higher Aβ40 (β = 0.25, p = .009), but hs-CRP was unrelated to AD-related biomarkers (ps > .05). There was a significant interaction such that older men with both chronic pain and higher levels of hs-CRP had higher levels of Aβ42 (β = 0.36, p = .001) and Aβ40 (β = 0.29, p = .003). Chronic pain and hs-CRP did not interact to predict levels of Aβ42/Aβ40, t-tau, or NfL. Furthermore, there were significant interactions such that Aβ42 and Aβ40 were associated with lower hippocampal volume, particularly when levels of hs-CRP were elevated (hs-CRP × Aβ42: β = -0.19, p = .002; hs-CRP × Aβ40: β = -0.21, p = .001), regardless of chronic pain status. CONCLUSIONS Chronic pain was associated with higher plasma Aβ, especially when hs-CRP was also elevated. Higher hs-CRP and Aβ levels were both related to smaller hippocampal volumes. Chronic pain, when accompanied by systemic inflammation, may elevate the risk of neurodegeneration in AD-vulnerable regions.
Collapse
Affiliation(s)
- Tyler R Bell
- Department of Psychiatry, University of California, San Diego, La Jolla, California, USA
- Center for Behavior Genetics of Aging, University of California, San Diego, La Jolla, California, USA
| | - Carol E Franz
- Department of Psychiatry, University of California, San Diego, La Jolla, California, USA
- Center for Behavior Genetics of Aging, University of California, San Diego, La Jolla, California, USA
| | - Kelsey R Thomas
- Department of Psychiatry, University of California, San Diego, La Jolla, California, USA
- Research Service, VA San Diego Healthcare System, San Diego, California, USA
| | - McKenna E Williams
- Department of Psychiatry, University of California, San Diego, La Jolla, California, USA
| | - Lisa T Eyler
- Department of Psychiatry, University of California, San Diego, La Jolla, California, USA
| | - Imanuel Lerman
- Department of Anesthesiology, University of California, San Diego, La Jolla, California, USA
| | - Christine Fennema-Notestine
- Department of Psychiatry, University of California, San Diego, La Jolla, California, USA
- Department of Radiology, University of California, San Diego, La Jolla, California, USA
| | - Olivia K Puckett
- Department of Psychiatry, University of California, San Diego, La Jolla, California, USA
- Center for Behavior Genetics of Aging, University of California, San Diego, La Jolla, California, USA
| | - Stephen M Dorros
- Department of Radiology, University of California, San Diego, La Jolla, California, USA
| | - Matthew S Panizzon
- Department of Psychiatry, University of California, San Diego, La Jolla, California, USA
- Center for Behavior Genetics of Aging, University of California, San Diego, La Jolla, California, USA
| | - Rahul C Pearce
- Department of Psychiatry, University of California, San Diego, La Jolla, California, USA
- Center for Behavior Genetics of Aging, University of California, San Diego, La Jolla, California, USA
| | - Donald J Hagler
- Department of Anesthesiology, University of California, San Diego, La Jolla, California, USA
- Department of Radiology, University of California, San Diego, La Jolla, California, USA
| | - Michael J Lyons
- Department of Psychology, Boston University, Boston, Massachusetts, USA
| | - Jeremy A Elman
- Department of Psychiatry, University of California, San Diego, La Jolla, California, USA
- Center for Behavior Genetics of Aging, University of California, San Diego, La Jolla, California, USA
| | - William S Kremen
- Department of Psychiatry, University of California, San Diego, La Jolla, California, USA
- Center for Behavior Genetics of Aging, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
2
|
Duggan MR, Gomez GT, Joynes CM, Bilgel M, Chen J, Fattorelli N, Hohman TJ, Mancuso R, Cordon J, Castellano T, Koran MEI, Candia J, Lewis A, Moghekar A, Ashton NJ, Kac PR, Karikari TK, Blennow K, Zetterberg H, Martinez-Muriana A, De Strooper B, Thambisetty M, Ferrucci L, Gottesman RF, Coresh J, Resnick SM, Walker KA. Proteome-wide analysis identifies plasma immune regulators of amyloid-beta progression. Brain Behav Immun 2024; 120:604-619. [PMID: 38977137 DOI: 10.1016/j.bbi.2024.07.002] [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: 04/04/2024] [Revised: 06/07/2024] [Accepted: 07/04/2024] [Indexed: 07/10/2024] Open
Abstract
While immune function is known to play a mechanistic role in Alzheimer's disease (AD), whether immune proteins in peripheral circulation influence the rate of amyloid-β (Aβ) progression - a central feature of AD - remains unknown. In the Baltimore Longitudinal Study of Aging, we quantified 942 immunological proteins in plasma and identified 32 (including CAT [catalase], CD36 [CD36 antigen], and KRT19 [keratin 19]) associated with rates of cortical Aβ accumulation measured with positron emission tomography (PET). Longitudinal changes in a subset of candidate proteins also predicted Aβ progression, and the mid- to late-life (20-year) trajectory of one protein, CAT, was associated with late-life Aβ-positive status in the Atherosclerosis Risk in Communities (ARIC) study. Genetic variation that influenced plasma levels of CAT, CD36 and KRT19 predicted rates of Aβ accumulation, including causal relationships with Aβ PET levels identified with two-sample Mendelian randomization. In addition to associations with tau PET and plasma AD biomarker changes, as well as expression patterns in human microglia subtypes and neurovascular cells in AD brain tissue, we showed that 31 % of candidate proteins were related to mid-life (20-year) or late-life (8-year) dementia risk in ARIC. Our findings reveal plasma proteins associated with longitudinal Aβ accumulation, and identify specific peripheral immune mediators that may contribute to the progression of AD pathophysiology.
Collapse
Affiliation(s)
- Michael R Duggan
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Gabriela T Gomez
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Cassandra M Joynes
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Murat Bilgel
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Jingsha Chen
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Nicola Fattorelli
- VIB Center for Brain and Disease Research, Flanders Institute for Biotechnology, Leuven, Belgium; Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - 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
| | - Renzo Mancuso
- Microglia and Inflammation in Neurological Disorders Laboratory, Center for Molecular Neurology, Flanders Institute for Biotechnology, Antwerp, Belgium; Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Jenifer Cordon
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Tonnar Castellano
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mary Ellen I Koran
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Julián Candia
- Translational Gerontology Branch, 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
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden; Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK; NIHR Biomedical Research Center for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK; Center for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
| | - Przemysław R Kac
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden; Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; ICM Institute, Pitié-Salpêtrière University Hospital, Sorbonne University, Paris, France; First Affiliated Hospital, University of Science and Technology of China, Anhui, PR China
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease, University College London Institute of Neurology, London, UK; UK Dementia Research Institute, University College London, London, UK; Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong Special Administrative Region; Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Anna Martinez-Muriana
- VIB Center for Brain and Disease Research, Flanders Institute for Biotechnology, Leuven, Belgium; Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Bart De Strooper
- VIB Center for Brain and Disease Research, Flanders Institute for Biotechnology, Leuven, Belgium; Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium; UK Dementia Research Institute, University College London, London, UK
| | - Madhav Thambisetty
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Rebecca F Gottesman
- Stroke Branch, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Josef Coresh
- Departments of Population Health and Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Susan M Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Keenan A Walker
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA.
| |
Collapse
|
3
|
Toribio-Fernandez R, Ceron C, Tristão-Pereira C, Fernandez-Nueda I, Perez-Castillo A, Fernandez-Ferro J, Moro MA, Ibañez B, Fuster V, Cortes-Canteli M. Oral anticoagulants: A plausible new treatment for Alzheimer's disease? Br J Pharmacol 2024; 181:760-776. [PMID: 36633908 DOI: 10.1111/bph.16032] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/02/2023] [Accepted: 01/06/2023] [Indexed: 01/13/2023] Open
Abstract
Alzheimer's disease (AD) and cardiovascular disease (CVD) are strongly associated. Both are multifactorial disorders with long asymptomatic phases and similar risk factors. Indeed, CVD signatures such as cerebral microbleeds, micro-infarcts, atherosclerosis, cerebral amyloid angiopathy and a procoagulant state are highly associated with AD. However, AD and CVD co-development and the molecular mechanisms underlying such associations are not understood. Here, we review the evidence regarding the vascular component of AD and clinical studies using anticoagulants that specifically evaluated the development of AD and other dementias. Most studies reported a markedly decreased incidence of composite dementia in anticoagulated patients with atrial fibrillation, with the highest benefit for direct oral anticoagulants. However, sub-analyses by differential dementia diagnosis were scarce and inconclusive. We finally discuss whether anticoagulation could be a plausible preventive/therapeutic approach for AD and, if so, which would be the best drug and strategy to maximize clinical benefit and minimize potential risks. LINKED ARTICLES: This article is part of a themed issue From Alzheimer's Disease to Vascular Dementia: Different Roads Leading to Cognitive Decline. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.6/issuetoc.
Collapse
Affiliation(s)
- Raquel Toribio-Fernandez
- Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Carlos Ceron
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | | | - Irene Fernandez-Nueda
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Ana Perez-Castillo
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Madrid, Spain
| | - Jose Fernandez-Ferro
- Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Stroke Unit, Neurology Service, Hospital Universitario Rey Juan Carlos (HURJC), Madrid, Spain
| | - Maria Angeles Moro
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Borja Ibañez
- Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- CIBER de enfermedades cardiovasculares (CIBERCV), ISCIII, Madrid, Spain
| | - Valentin Fuster
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Marta Cortes-Canteli
- Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
4
|
Reyes-Reyes EM, Brown J, Trial MD, Chinnasamy D, Wiegand JP, Bradford D, Brinton RD, Rodgers KE. Vivaria housing conditions expose sex differences in brain oxidation, microglial activation, and immune system states in aged hAPOE4 mice. Exp Brain Res 2024; 242:543-557. [PMID: 38206365 PMCID: PMC10894770 DOI: 10.1007/s00221-023-06763-x] [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: 08/17/2023] [Accepted: 12/04/2023] [Indexed: 01/12/2024]
Abstract
Apolipoprotein E ε4 allele (APOE4) is the predominant genetic risk factor for late-onset Alzheimer's disease (AD). APOE4 mouse models have provided advances in the understanding of disease pathogenesis, but unaccounted variables like rodent housing status may hinder translational outcomes. Non-sterile aspects like food and bedding can be major sources of changes in rodent microflora. Alterations in intestinal microbial ecology can cause mucosal barrier impairment and increase pro-inflammatory signals. The present study examined the role of sterile and non-sterile food and housing on redox indicators and the immune status of humanized-APOE4 knock-in mice (hAPOe4). hAPOE4 mice were housed under sterile conditions until 22 months of age, followed by the transfer of a cohort of mice to non-sterile housing for 2 months. At 24 months of age, the redox/immunologic status was evaluated by flow cytometry/ELISA. hAPOE4 females housed under non-sterile conditions exhibited: (1) higher neuronal and microglial oxygen radical production and (2) lower CD68+ microglia (brain) and CD8+ T cells (periphery) compared to sterile-housed mice. In contrast, hAPOE4 males in non-sterile housing exhibited: (1) higher MHCII+ microglia and CD11b+CD4+ T cells (brain) and (2) higher CD11b+CD4+ T cells and levels of lipopolysaccharide-binding protein and inflammatory cytokines in the periphery relative to sterile-housed mice. This study demonstrated that sterile vs. non-sterile housing conditions are associated with the activation of redox and immune responses in the brain and periphery in a sex-dependent manner. Therefore, housing status may contribute to variable outcomes in both the brain and periphery.
Collapse
Affiliation(s)
- E M Reyes-Reyes
- Center for Innovation in Brain Science, University of Arizona, 1230 N. Cherry Ave, PO Box 210242, Tucson, AZ, 85721-0242, USA
| | - J Brown
- Center for Innovation in Brain Science, University of Arizona, 1230 N. Cherry Ave, PO Box 210242, Tucson, AZ, 85721-0242, USA
| | - M D Trial
- Center for Innovation in Brain Science, University of Arizona, 1230 N. Cherry Ave, PO Box 210242, Tucson, AZ, 85721-0242, USA
| | - D Chinnasamy
- Center for Innovation in Brain Science, University of Arizona, 1230 N. Cherry Ave, PO Box 210242, Tucson, AZ, 85721-0242, USA
| | - J P Wiegand
- Center for Innovation in Brain Science, University of Arizona, 1230 N. Cherry Ave, PO Box 210242, Tucson, AZ, 85721-0242, USA
| | - D Bradford
- Center for Innovation in Brain Science, University of Arizona, 1230 N. Cherry Ave, PO Box 210242, Tucson, AZ, 85721-0242, USA
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - R D Brinton
- Center for Innovation in Brain Science, University of Arizona, 1230 N. Cherry Ave, PO Box 210242, Tucson, AZ, 85721-0242, USA
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - K E Rodgers
- Center for Innovation in Brain Science, University of Arizona, 1230 N. Cherry Ave, PO Box 210242, Tucson, AZ, 85721-0242, USA.
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
5
|
Okine DN, Knopman DS, Mosley TH, Wong DF, Johansen MC, Walker KA, Jack CR, Kantarci K, Pike JR, Graff-Radford J, Gottesman RF. Cerebral Microbleed Patterns and Cortical Amyloid-β: The ARIC-PET Study. Stroke 2023; 54:2613-2620. [PMID: 37638398 PMCID: PMC10877560 DOI: 10.1161/strokeaha.123.042835] [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: 02/16/2023] [Revised: 07/10/2023] [Accepted: 08/02/2023] [Indexed: 08/29/2023]
Abstract
BACKGROUND Cerebral microbleeds (CMBs) are associated with cognitive decline, but their importance outside of cerebral amyloid angiopathy and the mechanisms of their impact on cognition are poorly understood. We evaluated the cross-sectional association between CMB patterns and cerebral Aβ (amyloid-β) deposition, by florbetapir positron emission tomography. METHODS The longitudinal ARIC study (Atherosclerosis Risk in Communities) recruited individuals from 4 US communities from 1987 to 1989. From 2012 to 2014, the ARIC-PET (Atherosclerosis Risk in Communities - Positron Emission Tomography) ancillary recruited 322 nondemented ARIC participants who completed 3T brain magnetic resonance imaging with T2*GRE as part of ARIC visit 5 to undergo florbetapir positron emission tomography imaging. Magnetic resonance imaging images were read for CMBs and superficial siderosis; on positron emission tomography, global cortical standardized uptake value ratio >1.2 was considered a positive Aβ scan. Multivariable logistic regression models evaluated CMB characteristics in association with Aβ positivity. Effect modification by sex, race, APOE status, and cognition was evaluated. RESULTS CMBs were present in 24% of ARIC-PET participants. No significant associations were found between CMBs and Aβ positivity, but a pattern of isolated lobar CMBs or superficial siderosis was associated with over 4-fold higher odds of elevated Aβ when compared with those with no CMBs (odds ratio, 4.72 [95% CI, 1.16-19.16]). A similar elevated risk was not observed in those with isolated subcortical or mixed subcortical and either lobar CMBs or superficial siderosis. Although no significant interactions were found, effect estimates for elevated Aβ were nonsignificantly lower (P>0.10, odds ratio, 0.4-0.6) for a mixed CMB pattern, and odds ratios were nonsignificantly higher for lobar-only CMBs for 4 subgroups: women (versus men); Black participants (versus White participants), APOE ε4 noncarriers (versus carriers), and cognitively normal (versus mild cognitive impairment). CONCLUSIONS In this community-based cohort of nondemented adults, lobar-only pattern of CMBs or superficial siderosis is most strongly associated with brain Aβ, with no elevated risk for a mixed CMB pattern. Further studies are needed to understand differences in CMB patterns and their meaning across subgroups.
Collapse
Affiliation(s)
- Derrick N. Okine
- National Institute of Neurological Disorders and Stroke Intramural Research Program, NIH, Bethesda, MD
| | | | - Thomas H. Mosley
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS
| | - Dean F. Wong
- Department of Radiology, Washington University, St. Louis, MO
| | - Michelle C. Johansen
- Department of Neurology, The John Hopkins University School of Medicine, Baltimore, MD
| | - Keenan A. Walker
- National Institute on Aging Intramural Program, NIH, Baltimore, MD
| | | | | | - James R. Pike
- Gillings School of Global Public Health, University of North Carolina
| | | | - Rebecca F. Gottesman
- National Institute of Neurological Disorders and Stroke Intramural Research Program, NIH, Bethesda, MD
| |
Collapse
|
6
|
Feng V, Lanctot K, Herrmann N, Kiss A, Fischer CE, Flint AJ, Mah L, Mulsant BH, Pollock BG, Rajji TK, Tumati S, Verhoeff NP, Graff-Guerrero A, Gallagher D. Lipopolysaccharide, Immune Biomarkers and Cerebral Amyloid-Beta Deposition in Older Adults With Mild Cognitive Impairment & Major Depressive Disorder. Am J Geriatr Psychiatry 2023; 31:786-795. [PMID: 37211499 DOI: 10.1016/j.jagp.2023.04.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 05/23/2023]
Abstract
OBJECTIVE Inflammatory activation and increased immune response to lipopolysaccharide occur in both depression and cognitive decline and may link these two conditions. We investigated whether lipopolysaccharide (LPS), LPS binding protein (LBP) and peripheral biomarkers of immune response were associated with increased cerebral deposition of amyloid-beta (Abeta) in older adults with mild cognitive impairment (MCI) and remitted major depressive disorder (rMDD). DESIGN Cross-sectional analysis. SETTING Five academic health centers in Toronto. PARTICIPANTS Older adults with MCI with/without rMDD. MEASUREMENTS We investigated the associations among serum LPS, LBP, biomarkers of inflammatory activation - Interleukin-6 (IL-6), C-reactive protein (CRP), monocyte chemoattractant protein-1 (MCP-1), and cerebral Abeta deposition quantified by positron emission tomography. RESULTS Among 133 study participants (82 with MCI and 51 with MCI+rMDD) there was no association between LPS (beta - 0.17, p = 0.8) or LBP (beta - 0.11, p = 0.12) and global deposition of Abeta following adjustment for age, gender, and APOE genotype in multivariable regression analyses. LBP was positively correlated with CRP (r = 0.5, p <0.001) and IL-6 (r = 0.2, p = 0.02) but no inflammatory biomarker was associated with Abeta deposition; rMDD was not associated with deposition of Abeta (beta -0.09, p = 0.22). CONCLUSION In this cross-sectional analysis, we did not find an association among LPS/LBP, immune biomarkers or rMDD and global deposition of Abeta. Future analyses should assess the longitudinal relationships between peripheral and central biomarkers of immune activation, depression and cerebral Abeta deposition.
Collapse
Affiliation(s)
- Vivian Feng
- Neuropsychopharmacology Research Group (VF, KL, NH, AK, ST, DG), Sunnybrook Research Institute, Toronto, Canada
| | - Krista Lanctot
- Neuropsychopharmacology Research Group (VF, KL, NH, AK, ST, DG), Sunnybrook Research Institute, Toronto, Canada; Department of Psychiatry (KL, NH, CEF, AJF, LM, BHM, BGP, TKR, NPV, AG-G, DG), Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Nathan Herrmann
- Neuropsychopharmacology Research Group (VF, KL, NH, AK, ST, DG), Sunnybrook Research Institute, Toronto, Canada; Department of Psychiatry (KL, NH, CEF, AJF, LM, BHM, BGP, TKR, NPV, AG-G, DG), Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Alex Kiss
- Neuropsychopharmacology Research Group (VF, KL, NH, AK, ST, DG), Sunnybrook Research Institute, Toronto, Canada; Department of Health Policy, Management and Evaluation (AK), University of Toronto, Toronto, Canada
| | - Corinne E Fischer
- Department of Psychiatry (KL, NH, CEF, AJF, LM, BHM, BGP, TKR, NPV, AG-G, DG), Temerty Faculty of Medicine, University of Toronto, Toronto, Canada; Keenan Research Centre for Biomedical Science (CEF), St. Michael's Hospital, Toronto, Canada
| | - Alastair J Flint
- Department of Psychiatry (KL, NH, CEF, AJF, LM, BHM, BGP, TKR, NPV, AG-G, DG), Temerty Faculty of Medicine, University of Toronto, Toronto, Canada; Centre for Mental Health (AJF), University Health Network and University of Toronto, Toronto, Canada
| | - Linda Mah
- Department of Psychiatry (KL, NH, CEF, AJF, LM, BHM, BGP, TKR, NPV, AG-G, DG), Temerty Faculty of Medicine, University of Toronto, Toronto, Canada; Department of Psychiatry (LM, NPV), Baycrest Health Sciences Centre, Toronto, Canada; Rotman Research Institute (LM), Baycrest Health Sciences Centre, Toronto, Canada
| | - Benoit H Mulsant
- Department of Psychiatry (KL, NH, CEF, AJF, LM, BHM, BGP, TKR, NPV, AG-G, DG), Temerty Faculty of Medicine, University of Toronto, Toronto, Canada; Adult Neurodevelopment and Geriatric Psychiatry Division (BHM, BGP, TKR, AG-G), Centre for Addiction and Mental Health, Toronto, Canada; Toronto Dementia Research Alliance (BMH, BGP, TKR, NPV), University of Toronto, Toronto, Canada
| | - Bruce G Pollock
- Department of Psychiatry (KL, NH, CEF, AJF, LM, BHM, BGP, TKR, NPV, AG-G, DG), Temerty Faculty of Medicine, University of Toronto, Toronto, Canada; Adult Neurodevelopment and Geriatric Psychiatry Division (BHM, BGP, TKR, AG-G), Centre for Addiction and Mental Health, Toronto, Canada; Toronto Dementia Research Alliance (BMH, BGP, TKR, NPV), University of Toronto, Toronto, Canada
| | - Tarek K Rajji
- Department of Psychiatry (KL, NH, CEF, AJF, LM, BHM, BGP, TKR, NPV, AG-G, DG), Temerty Faculty of Medicine, University of Toronto, Toronto, Canada; Adult Neurodevelopment and Geriatric Psychiatry Division (BHM, BGP, TKR, AG-G), Centre for Addiction and Mental Health, Toronto, Canada; Toronto Dementia Research Alliance (BMH, BGP, TKR, NPV), University of Toronto, Toronto, Canada
| | - Shankar Tumati
- Neuropsychopharmacology Research Group (VF, KL, NH, AK, ST, DG), Sunnybrook Research Institute, Toronto, Canada
| | - Nicolaas Paul Verhoeff
- Department of Psychiatry (KL, NH, CEF, AJF, LM, BHM, BGP, TKR, NPV, AG-G, DG), Temerty Faculty of Medicine, University of Toronto, Toronto, Canada; Department of Psychiatry (LM, NPV), Baycrest Health Sciences Centre, Toronto, Canada; Toronto Dementia Research Alliance (BMH, BGP, TKR, NPV), University of Toronto, Toronto, Canada
| | - Ariel Graff-Guerrero
- Department of Psychiatry (KL, NH, CEF, AJF, LM, BHM, BGP, TKR, NPV, AG-G, DG), Temerty Faculty of Medicine, University of Toronto, Toronto, Canada; Adult Neurodevelopment and Geriatric Psychiatry Division (BHM, BGP, TKR, AG-G), Centre for Addiction and Mental Health, Toronto, Canada
| | - Damien Gallagher
- Neuropsychopharmacology Research Group (VF, KL, NH, AK, ST, DG), Sunnybrook Research Institute, Toronto, Canada; Department of Psychiatry (KL, NH, CEF, AJF, LM, BHM, BGP, TKR, NPV, AG-G, DG), Temerty Faculty of Medicine, University of Toronto, Toronto, Canada.
| |
Collapse
|
7
|
Ebright B, Assante I, Poblete RA, Wang S, Duro MV, Bennett DA, Arvanitakis Z, Louie SG, Yassine HN. Eicosanoid lipidome activation in post-mortem brain tissues of individuals with APOE4 and Alzheimer's dementia. Alzheimers Res Ther 2022; 14:152. [PMID: 36217192 PMCID: PMC9552454 DOI: 10.1186/s13195-022-01084-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 09/16/2022] [Indexed: 01/18/2023]
Abstract
BACKGROUND Chronic neuroinflammation is one of the hallmarks of late-onset Alzheimer's disease (AD) dementia pathogenesis. Carrying the apolipoprotein ε4 (APOE4) allele has been associated with an accentuated response to brain inflammation and increases the risk of AD dementia progression. Among inflammation signaling pathways, aberrant eicosanoid activation plays a prominent role in neurodegeneration. METHODS Using brains from the Religious Order Study (ROS), this study compared measures of brain eicosanoid lipidome in older persons with AD dementia to age-matched controls with no cognitive impairment (NCI), stratified by APOE genotype. RESULTS Lipidomic analysis of the dorsolateral prefrontal cortex demonstrated lower levels of omega-3 fatty acids eicosapentaenoic acid (EPA), docosapentaenoic acid (DPA), and DHA-derived neuroprotectin D1 (NPD-1) in persons with AD dementia, all of which associated with lower measures of cognitive function. A significant interaction was observed between carrying the APOE4 allele and higher levels of both pro-inflammatory lipids and pro-resolving eicosanoid lipids on measures of cognitive performance and on neuritic plaque burden. Furthermore, analysis of lipid metabolism pathways implicated activation of calcium-dependent phospholipase A2 (cPLA2), 5-lipoxygenase (5-LOX), and soluble epoxide hydrolase (sEH) enzymes. CONCLUSION These findings implicate activation of the eicosanoid lipidome in the chronic unresolved state of inflammation in AD dementia, which is increased in carriers of the APOE4 allele, and identify potential therapeutic targets for resolving this chronic inflammatory state.
Collapse
Affiliation(s)
- Brandon Ebright
- School of Pharmacy, University of Southern California, Los Angeles, USA
| | - Isaac Assante
- School of Pharmacy, University of Southern California, Los Angeles, USA
- Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Roy A Poblete
- Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Shaowei Wang
- Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Marlon V Duro
- Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Zoe Arvanitakis
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Stan G Louie
- School of Pharmacy, University of Southern California, Los Angeles, USA.
- Keck School of Medicine, University of Southern California, Los Angeles, USA.
| | - Hussein N Yassine
- Keck School of Medicine, University of Southern California, Los Angeles, USA.
| |
Collapse
|
8
|
Oliver C, Adams D, Holland AJ, Brown SSG, Ball S, Dodd K, Carr J. Acquired mild cognitive impairment in adults with Down syndrome: Age-related prevalence derived from single point assessment data normed by degree of intellectual disability. Int J Geriatr Psychiatry 2022; 37:10.1002/gps.5674. [PMID: 34994494 PMCID: PMC9306607 DOI: 10.1002/gps.5674] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 12/21/2021] [Indexed: 12/23/2022]
Abstract
BACKGROUND Individuals with Down syndrome (DS) are at significant risk for early onset Alzheimer's disease (AD), likely due to the triplication of genes on chromosome 21 that facilitate AD neuropathology. To aid the effective early diagnosis of dementia in DS, we demonstrate the strategy of using single point assessment of cognitive performance with scoring normed for degree of intellectual disability to generate age related prevalence data for acquired mild cognitive impairment (AMCI). METHODS Four hundred and twelve adults with DS were assessed using the Neuropsychological Assessment of dementia in adults with Intellectual Disability. Normative data, banded by degree of intellectual disability, allowed identification of AMCI by atypical deviation from expected performance. RESULTS AMCI was evident in approximately 20% of adults with DS aged 40 and under, 40% aged 41-50 and 45% aged 51 and over. Relative risk increased significantly in those aged 46 and over. Analysis of prevalence by 5-year age bands revealed two peaks for higher prevalence of AMCI. CONCLUSIONS Psychometric data indicate single point assessment of AMCI is possible for the majority of adults with DS. Two peaks for age-related prevalence of AMCI suggest the risk for onset of AD conferred by trisomy of chromosome 21 is moderated by another factor, possibly ApoE status.
Collapse
Affiliation(s)
- Chris Oliver
- School of PsychologyUniversity of BirminghamBirminghamUK
| | - Dawn Adams
- Autism Centre of ExcellenceGriffith UniversityBrisbaneAustralia
| | - Anthony J. Holland
- Section of Developmental PsychiatryDepartment of PsychiatryUniversity of CambridgeCambridgeUK
| | - Stephanie S. G. Brown
- Section of Developmental PsychiatryDepartment of PsychiatryUniversity of CambridgeCambridgeUK
| | - Sarah Ball
- Section of Developmental PsychiatryDepartment of PsychiatryUniversity of CambridgeCambridgeUK
| | - Karen Dodd
- Psychology DepartmentSurrey and Borders Partnership NHS Foundation TrustLeatherheadUK
| | | |
Collapse
|
9
|
Weinstein G, O’Donnell A, Davis-Plourde K, Zelber-Sagi S, Ghosh S, DeCarli CS, Thibault EG, Sperling RA, Johnson KA, Beiser AS, Seshadri S. Non-Alcoholic Fatty Liver Disease, Liver Fibrosis, and Regional Amyloid-β and Tau Pathology in Middle-Aged Adults: The Framingham Study. J Alzheimers Dis 2022; 86:1371-1383. [PMID: 35213373 PMCID: PMC11323287 DOI: 10.3233/jad-215409] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Liver steatosis and fibrosis are emerging as risk factors for multiple extrahepatic health conditions; however, their relationship with Alzheimer's disease pathology is unclear. OBJECTIVE To examine whether non-alcoholic fatty liver disease (NAFLD) and FIB-4, a non-invasive index of advanced fibrosis, are associated with brain amyloid-β (Aβ) and tau pathology. METHODS The study sample included Framingham Study participants from the Offspring and Third generation cohorts who attended exams 9 (2011-2014) and 2 (2008-2011), respectively. Participants underwent 11C-Pittsburgh Compound-B amyloid and 18F-Flortaucipir tau positron emission tomography (PET) imaging and abdomen computed tomography, or had information on all components of the FIB-4 index. Linear regression models were used to assess the relationship of NAFLD and FIB-4 with regional tau and Aβ, adjusting for potential confounders and multiple comparisons. RESULTS Of the subsample with NAFLD information (N = 169; mean age 52±9 y; 57% males), 57 (34%) had NAFLD. Of the subsample with information on liver fibrosis (N = 177; mean age 50±10 y; 51% males), 34 (19%) had advanced fibrosis (FIB-4 > 1.3). Prevalent NAFLD was not associated with Aβ or tau PET. However, FIB-4 index was significantly associated with increased rhinal tau (β= 1.03±0.33, p = 0.002). Among individuals with prevalent NAFLD, FIB-4 was related to inferior temporal, parahippocampal gyrus, entorhinal and rhinal tau (β= 2.01±0.47, p < 0.001; β= 1.60±0.53, p = 0.007, and β= 1.59±0.47, p = 0.003 and β= 1.60±0.42, p = 0.001, respectively) and to Aβ deposition overall and in the inferior temporal and parahippocampal regions (β= 1.93±0.47, p < 0.001; β= 1.59±0.38, p < 0.001, and β= 1.52±0.54, p = 0.008, respectively). CONCLUSION This study suggests a possible association between liver fibrosis and early Alzheimer's disease pathology, independently of cardio-metabolic risk factors.
Collapse
Affiliation(s)
| | - Adrienne O’Donnell
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- The Framingham Study, Framingham, MA, USA
| | - Kendra Davis-Plourde
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- The Framingham Study, Framingham, MA, USA
| | - Shira Zelber-Sagi
- School of Public Health, University of Haifa, Haifa, Israel
- Liver Unit, Department of Gastroenterology, Tel-Aviv Medical Center, Tel-Aviv, Israel
| | - Saptaparni Ghosh
- The Framingham Study, Framingham, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Charles S. DeCarli
- Department of Neurology, School of Medicine & Imaging of Dementia and Aging Laboratory, Center for Neuroscience, University of California Davis, Davis, CA, USA
| | - Emma G. Thibault
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Reisa A. Sperling
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Keith A. Johnson
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Alexa S. Beiser
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- The Framingham Study, Framingham, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Sudha Seshadri
- The Framingham Study, Framingham, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, USA
| |
Collapse
|
10
|
Cohen AJ, Teramoto K, Claggett B, Buckley L, Solomon S, Ballantyne C, Selvin E, Shah AM. Mid- to Late-Life Inflammation and Risk of Cardiac Dysfunction, HFpEF and HFrEF in Late Life. J Card Fail 2021; 27:1382-1392. [PMID: 34314823 PMCID: PMC8823406 DOI: 10.1016/j.cardfail.2021.07.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 07/06/2021] [Accepted: 07/08/2021] [Indexed: 12/28/2022]
Abstract
BACKGROUND Epidemiologic data supporting the association of accumulated inflammation from mid- to late life with late-life risk of cardiac dysfunction and heart failure (HF) is limited. METHODS AND RESULTS Among 4011 participants in the Atherosclerosis Risk in Communities study who were free of prevalent cardiovascular disease at study Visit 5, accumulated inflammation was defined as time-averaged high-sensitivity c-reactive protein (hsCRP) over 3 visits spanning 1990 to 2013. Associations with left ventricular (LV) function at Visit 5 and with incident adjudicated HF post Visit 5 were assessed using linear and Cox regression, adjusting for demographics and comorbidities. Higher accumulated hsCRP was associated with greater LV mass index, lower e', higher E/e', and higher adjusting for demographics (all P ≤0.01), but only with higher pulmonary artery systolic pressure after adjustment for comorbidities (P = 0.024). At 5.3 ± 1.2 year follow-up, higher accumulated hsCRP was associated with greater risk of incident HF (HR 1.31 [95% CI 1.18-1.47], P < 0.001), HFrEF (1.26 [1.05-1.52], P = 0.01), and HFpEF (1.30 [1.11-1.53], P = 0.001) in demographic-adjusted models, but not after adjustment for comorbidities (all P > 0.10). Only Visit 5 hsCRP remained associated with incident HF (1.12 [1.02-1.24], P = 0.02) after full adjustment. CONCLUSIONS Greater accumulated inflammation is associated with worse LV function and heightened HF risk in late-life. These relationships are attenuated after adjusting for HF risk factors.
Collapse
Affiliation(s)
- Aaron J Cohen
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA.
| | - Kanako Teramoto
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA; Division of Cardiology, St. Marianna University School of Medicine Hospital, Kanagawa, Japan
| | - Brian Claggett
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Leo Buckley
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Scott Solomon
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | | | - Elizabeth Selvin
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Amil M Shah
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA.
| |
Collapse
|
11
|
Vore AS, Deak T. Alcohol, inflammation, and blood-brain barrier function in health and disease across development. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2021; 161:209-249. [PMID: 34801170 DOI: 10.1016/bs.irn.2021.06.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Alcohol is the most commonly used drug of abuse in the world and binge drinking is especially harmful to the brain, though the mechanisms by which alcohol compromises overall brain health remain somewhat elusive. A number of brain diseases and pathological states are accompanied by perturbations in Blood-Brain Barrier (BBB) function, ultimately exacerbating disease progression. The BBB is critical for coordinating activity between the peripheral immune system and the brain. Importantly, BBB integrity is responsive to circulating cytokines and other immune-related signaling molecules, which are powerfully modulated by alcohol exposure. This review will highlight key cellular components of the BBB; discuss mechanisms by which permeability is achieved; offer insight into methodological approaches for assessing BBB integrity; and forecast how alcohol-induced changes in the peripheral and central immune systems might influence BBB function in individuals with a history of binge drinking and ultimately Alcohol Use Disorders (AUD).
Collapse
Affiliation(s)
- A S Vore
- Behavioral Neuroscience Program, Department of Psychology, Developmental Exposure Alcohol Research Center, Binghamton, NY, United States
| | - T Deak
- Behavioral Neuroscience Program, Department of Psychology, Developmental Exposure Alcohol Research Center, Binghamton, NY, United States.
| |
Collapse
|
12
|
Bettcher BM, Tansey MG, Dorothée G, Heneka MT. Peripheral and central immune system crosstalk in Alzheimer disease - a research prospectus. Nat Rev Neurol 2021; 17:689-701. [PMID: 34522039 PMCID: PMC8439173 DOI: 10.1038/s41582-021-00549-x] [Citation(s) in RCA: 178] [Impact Index Per Article: 59.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/30/2021] [Indexed: 02/08/2023]
Abstract
Dysregulation of the immune system is a cardinal feature of Alzheimer disease (AD), and a considerable body of evidence indicates pathological alterations in central and peripheral immune responses that change over time. Considering AD as a systemic immune process raises important questions about how communication between the peripheral and central compartments occurs and whether this crosstalk represents a therapeutic target. We established a whitepaper workgroup to delineate the current status of the field and to outline a research prospectus for advancing our understanding of peripheral-central immune crosstalk in AD. To guide the prospectus, we begin with an overview of seminal clinical observations that suggest a role for peripheral immune dysregulation and peripheral-central immune communication in AD, followed by formative animal data that provide insights into possible mechanisms for these clinical findings. We then present a roadmap that defines important next steps needed to overcome conceptual and methodological challenges, opportunities for future interdisciplinary research, and suggestions for translating promising mechanistic studies into therapeutic interventions.
Collapse
Affiliation(s)
- Brianne M Bettcher
- Behavioral Neurology Section, Department of Neurology, University of Colorado Alzheimer's and Cognition Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Malú G Tansey
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine, Gainesville, FL, USA
| | - Guillaume Dorothée
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, Team "Immune System and Neuroinflammation", Hôpital Saint-Antoine, Paris, France
| | - Michael T Heneka
- Department of Neurodegenerative Diseases & Geropsychiatry/Neurology, University of Bonn Medical Center, Bonn, Germany
- Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA, USA
| |
Collapse
|
13
|
Qian XH, Song XX, Liu XL, Chen SD, Tang HD. Inflammatory pathways in Alzheimer's disease mediated by gut microbiota. Ageing Res Rev 2021; 68:101317. [PMID: 33711509 DOI: 10.1016/j.arr.2021.101317] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 02/25/2021] [Accepted: 03/08/2021] [Indexed: 12/14/2022]
Abstract
In the past decade, numerous studies have demonstrated the close relationship between gut microbiota and the occurrence and development of Alzheimer's disease (AD). However, the specific mechanism is still unclear. Both the neuroinflammation and systemic inflammation serve as the key hubs to accelerate the process of AD by promoting pathology and damaging neuron. What's more, the gut microbiota is also crucial for the regulation of inflammation. Therefore, this review focused on the role of gut microbiota in AD through inflammatory pathways. Firstly, this review summarized the relationship and interaction among gut microbiota, inflammation, and AD. Secondly, the direct and indirect regulatory effects of gut microbiota on AD through inflammatory pathways were described. These effects were mainly mediated by the component of the gut microbiota (lipopolysaccharides (LPS) and amyloid peptides), the metabolites of bacteria (short-chain fatty acids, branched amino acids, and neurotransmitters) and functional by-products (bile acids). In addition, potential treatments (fecal microbiota transplantation, antibiotics, probiotics, prebiotics, and dietary interventions) for AD were also discussed through these mechanisms. Finally, according to the current research status, the key problems to be solved in the future studies were proposed.
Collapse
Affiliation(s)
- Xiao-Hang Qian
- Department of Neurology and Institute of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Xiao-Xuan Song
- Department of Neurology and Institute of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Xiao-Li Liu
- Department of Neurology, Shanghai Fengxian District Central Hospital, Shanghai Jiao Tong University Affiliated Sixth People's Hospital South Campus, Shanghai, 201406, China.
| | - Sheng-di Chen
- Department of Neurology and Institute of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Hui-Dong Tang
- Department of Neurology and Institute of Neurology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
14
|
Oberlin LE, Erickson KI, Mackey R, Klunk WE, Aizenstein H, Lopresti BJ, Kuller LH, Lopez OL, Snitz BE. Peripheral inflammatory biomarkers predict the deposition and progression of amyloid-β in cognitively unimpaired older adults. Brain Behav Immun 2021; 95:178-189. [PMID: 33737171 PMCID: PMC8647033 DOI: 10.1016/j.bbi.2021.03.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 02/23/2021] [Accepted: 03/10/2021] [Indexed: 12/28/2022] Open
Abstract
INTRODUCTION Systemic inflammation has been increasingly implicated in the pathogenesis of Alzheimer's disease (AD), yet the mechanistic and temporal specificity of this relationship is poorly understood. We aimed to characterize the cross-sectional and longitudinal associations between peripheral inflammatory biomarkers, cognition, and Aβ deposition in oldest-old cognitively unimpaired (CU) adults. METHODS A large sample of 139 CU older adults (mean age (range) = 85.4 (82-95)) underwent neuropsychological testing, Pittsburgh compound-B (PiB)-PET imaging and structural MRI. Hierarchical regression models examined associations between circulating inflammatory biomarkers (Interleukin-6 (IL-6), soluble Tumor Necrosis Factor receptors 1 and 2 (sTNFr1 and sTNFr2), soluble cluster of differentiation 14 (sCD14), C-reactive protein (CRP)), cognition, and global and regional Aβ deposition at baseline and over follow-up. Indices of preclinical disease, including pathologic Aβ status and hippocampal volume, were incorporated to assess conditional associations. RESULTS At baseline evaluation, higher concentrations of IL-6 and sTNFr2 were associated with greater global Aβ burden in those with lower hippocampal volume. In longitudinal models, IL-6 predicted subsequent conversion to MCI and both IL-6 and CRP predicted greater change in global and regional Aβ deposition specifically among participants PiB-positive at baseline. These relationships withstood adjustment for demographic factors, anti-hypertensive medication use, history of diabetes, heart disease, APOE ε4 carrier status, and white matter lesions. DISCUSSION In a large prospective sample of CU adults aged 80 and over, peripheral inflammatory biomarkers were associated with and predictive of the progression of Aβ deposition. This was specific to those with biomarker evidence of preclinical AD at baseline, supporting recent evidence of disease-state-dependent differences in inflammatory expression profiles. Chronic, low-level systemic inflammation may exacerbate the deposition of Aβ pathology among those with emerging disease processes, and place individuals at a higher risk of developing clinically significant cognitive impairment.
Collapse
Affiliation(s)
| | - Kirk I. Erickson
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA,Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA,College of Science, Health, Engineering and Education, Murdoch University, Perth, Australia
| | - Rachel Mackey
- Premier Applied Sciences, Premier Inc., Charlotte, North Carolina,Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA
| | - William E. Klunk
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA
| | | | | | - Lewis H. Kuller
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA
| | - Oscar L. Lopez
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA,Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA
| | - Beth E. Snitz
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
15
|
Walker KA, Chawla S, Nogueras-Ortiz C, Coresh J, Sharrett AR, Wong DF, Jack CR, Spychalla AJ, Gottesman RF, Kapogiannis D. Neuronal insulin signaling and brain structure in nondemented older adults: the Atherosclerosis Risk in Communities Study. Neurobiol Aging 2021; 97:65-72. [PMID: 33160263 PMCID: PMC7736127 DOI: 10.1016/j.neurobiolaging.2020.09.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 09/10/2020] [Accepted: 09/21/2020] [Indexed: 12/28/2022]
Abstract
We used plasma neuronal extracellular vesicles to examine how neuronal insulin signaling proteins relate cross-sectionally to brain structure in nondemented older adults with varying levels of cortical amyloid. Extracellular vesicles enriched for neuronal origin by anti-L1CAM immunoabsorption were isolated from plasma of Atherosclerosis Risk in Communities-Positron Emission Tomography study participants (n = 88; mean age: 77 years [standard deviation: 6]). Neuronal extracellular vesicle levels of phosphorylated insulin signaling cascade proteins were quantified. Brain volume and white matter hyperintensity (WMH) volume were assessed using 3T magnetic resonance imaging. After adjusting for demographic variables and extracellular vesicle marker Alix, higher levels of a neuronal insulin signaling composite measure were associated with lower WMH and greater temporal lobe volume. Secondary analyses found the levels of downstream protein kinases involved in cell survival (p70S6K) and tau phosphorylation/neuroinflammation (GSK-3β) to be most strongly associated with WMH and temporal lobe volume, respectively. Associations between neuronal insulin signaling and lower WMH volume were attenuated in participants with elevated cortical amyloid. These results suggest that enhanced neuronal proximal insulin signaling is associated with preserved brain structure in nondemented older adults.
Collapse
Affiliation(s)
- Keenan A Walker
- Department of Neurology, Johns Hopkins University, Baltimore, MD, USA.
| | - Sahil Chawla
- Laboratory of Clinical Investigation, National Institute on Aging, Intramural Research Program, Baltimore, MD, USA
| | - Carlos Nogueras-Ortiz
- Laboratory of Clinical Investigation, National Institute on Aging, Intramural Research Program, Baltimore, MD, USA
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins University, Baltimore, MD, USA
| | - A Richey Sharrett
- Department of Epidemiology, Johns Hopkins University, Baltimore, MD, USA
| | - Dean F Wong
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MS, USA
| | | | | | - Rebecca F Gottesman
- Department of Neurology, Johns Hopkins University, Baltimore, MD, USA; Laboratory of Clinical Investigation, National Institute on Aging, Intramural Research Program, Baltimore, MD, USA
| | - Dimitrios Kapogiannis
- Laboratory of Clinical Investigation, National Institute on Aging, Intramural Research Program, Baltimore, MD, USA; Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
16
|
Duarte-Guterman P, Albert AY, Inkster AM, Barha CK, Galea LAM. Inflammation in Alzheimer's Disease: Do Sex and APOE Matter? J Alzheimers Dis 2020; 78:627-641. [PMID: 33016923 DOI: 10.3233/jad-200982] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) disproportionately affects females with steeper cognitive decline and more neuropathology compared to males, which is exacerbated in females carrying the APOEɛ4 allele. The risk of developing AD is also higher in female APOEɛ4 carriers in earlier age groups (aged 65-75), and the progression from cognitively normal to mild cognitive impairment (MCI) and to AD may be influenced by sex. Inflammation is observed in AD and is related to aging, stress, and neuroplasticity, and although studies are scarce, sex differences are noted in inflammation. OBJECTIVE The objective of this study was to investigate underlying physiological inflammatory mechanisms that may help explain why there are sex differences in AD and APOEɛ4 carriers. METHODS We investigated, using the ADNI database, the effect of sex and APOE genotype (non-carriers or carriers of 1 and 2 APOEɛ4 alleles) and sex and diagnosis (cognitively normal (CN), MCI, AD) on CSF (N = 279) and plasma (N = 527) markers of stress and inflammation. RESULTS We found CSF IL-16 and IL-8 levels differed by sex and APOE genotype, as IL-16 was higher in female APOEɛ4 carriers compared to non-carriers, while the opposite pattern was observed in males with IL-8. Furthermore, females had on average higher levels of plasma CRP and ICAM1 but lower levels of CSF ICAM1, IL-8, IL-16, and IgA than males. Carrying APOEɛ4 alleles and diagnosis (MCI and AD) decreased plasma CRP in both sexes. CONCLUSION Sex and APOE genotype differences in CSF and plasma inflammatory biomarkers support that the underlying physiological changes during aging differ by sex and tissue origin.
Collapse
Affiliation(s)
- Paula Duarte-Guterman
- Djavad Mowafaghian Centre for Brain Health and Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - Arianne Y Albert
- Women's Health Research Institute of British Columbia, Vancouver, BC, Canada
| | - Amy M Inkster
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada.,BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Cindy K Barha
- Djavad Mowafaghian Centre for Brain Health and Department of Physical Therapy, University of British Columbia, Vancouver, BC, Canada
| | - Liisa A M Galea
- Djavad Mowafaghian Centre for Brain Health and Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | | |
Collapse
|
17
|
Razquin C, Menéndez-Acebal C, Cervantes S, Martínez-González MA, Vázquez-Ruiz Z, Martínez-González J, Guillén-Grima F, Toledo E. Hypertension and changes in cognitive function in a Mediterranean population. Nutr Neurosci 2020; 25:612-620. [PMID: 32635835 DOI: 10.1080/1028415x.2020.1788773] [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: 10/23/2022]
Abstract
Background: Severe cognitive decline is one of the major public health problems in developed countries. Finding modifiable risk factors could become essential to develop strategies to prevent or delay dementia progression and stop its rising incidence.Objective: Our aim was to investigate the association between hypertension and cognitive function and to assess whether better adherence to the Mediterranean diet may modify this association.Methods: A subsample of 764 participants from the 'Seguimiento Universidad de Navarra' (SUN) cohort older than 55 years was evaluated with the Spanish Telephone Interview for Cognitive Status (TICS-m) at two-time points, separated by 6 years. Multivariable-adjusted linear regression models were used to prospectively assess the association between hypertension -also according to adherence to the Mediterranean diet- and 6-y changes in cognitive function.Results: The adjusted between-group difference in the 6-year change of the TICS-m score between hypertensive participants and their non-hypertensive counterparts was -0.36 (95% CI -0.70, -0.02). This association was stronger among participants with a lower adherence to the Mediterranean diet [-0.62 (95% CI: -1.09, -0.15)] but the differences between hypertensive and non-hypertensive participants were no longer significant among participants with a higher baseline adherence to the Mediterranean diet.Conclusion: In this Mediterranean cohort, hypertension was inversely associated with cognitive function, but an attenuation of this detrimental association by a moderate/high adherence to the Mediterranean diet was suggested.
Collapse
Affiliation(s)
- Cristina Razquin
- Department of Preventive Medicine and Public Health, University of Navarra, Pamplona, Spain.,Centro de Investigación Biomédica en Red Área de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Madrid, Spain.,IdiSNA, Pamplona, Spain
| | | | - Sebastián Cervantes
- Complejo Hospitalario de Navarra, Osasunbidea-Servicio Navarro de Salud, Pamplona, Spain
| | - Miguel A Martínez-González
- Department of Preventive Medicine and Public Health, University of Navarra, Pamplona, Spain.,Centro de Investigación Biomédica en Red Área de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Madrid, Spain.,IdiSNA, Pamplona, Spain.,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Zenaida Vázquez-Ruiz
- Department of Preventive Medicine and Public Health, University of Navarra, Pamplona, Spain.,IdiSNA, Pamplona, Spain
| | | | - Francisco Guillén-Grima
- Centro de Investigación Biomédica en Red Área de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Madrid, Spain.,IdiSNA, Pamplona, Spain.,Department of Preventive Medicine, Clinica Universidad de Navarra, Pamplona, Spain
| | - Estefania Toledo
- Department of Preventive Medicine and Public Health, University of Navarra, Pamplona, Spain.,Centro de Investigación Biomédica en Red Área de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Madrid, Spain.,IdiSNA, Pamplona, Spain
| |
Collapse
|
18
|
West NA, Kullo IJ, Morris MC, Mosley TH. Sex-specific associations of inflammation markers with cognitive decline. Exp Gerontol 2020; 138:110986. [PMID: 32497552 DOI: 10.1016/j.exger.2020.110986] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 04/16/2020] [Accepted: 05/26/2020] [Indexed: 10/24/2022]
Abstract
BACKGROUND/OBJECTIVE Inflammation is implicated in cognitive decline; however, there is a paucity of data for African American populations and for sex-specific associations. DESIGN Prospective cohort study. SETTING Genetic Epidemiology Network of Arteriopathy/Genetics of Microangiopathic Brain Injury studies. PARTICIPANTS African-American sibships (N = 1010). MEASUREMENTS Neurocognitive tests assessed global cognition and four cognitive domains: processing speed, memory, language, and executive function at two time points over seven years. Circulating levels of C-reactive protein (CRP), interleukin-6 (IL-6), and tumor necrosis factor receptor (TNFR)-1 and TNFR2 were measured at study baseline. Linear mixed models were used to investigate the association between inflammation markers and cognitive decline. RESULTS Among men, a one SD increase in CRP was associated with an increased rate of decline over 7 years in global cognitive Z-score (adjusted difference in slopes = -0.31, p = 0.006) and in processing speed Z-score (adjusted difference in slopes = -0.10, p = 0.02), but not declines in memory, language, or executive function Z-scores. Also among men, a one SD increase in IL-6 was associated with an increased decline rate in global cognitive Z-score (adjusted difference in slopes = -0.33, p = 0.002) and in processing speed Z-score (adjusted difference in slopes = -0.12, p = 0.007). There was no difference in decline rates by CRP or IL-6 level in adjusted analyses among women for any cognitive scores. Among men and women combined, a one SD increase in baseline sTNFR1 was associated with a faster rate of decline in memory Z-score (adjusted difference in slopes = -0.09, p = 0.02). Baseline sTNFR2 levels did not significantly predict rate of cognitive decline in any cognitive domains. CONCLUSIONS Circulating markers of CRP and IL-6 may be differential risk factors for men and women in relation to cognitive decline. A novel inflammation marker, sTNFR1, may be a useful predictor of memory decline in older adults.
Collapse
Affiliation(s)
- Nancy A West
- Department of Internal Medicine, Division of Epidemiology, University of Utah, Salt Lake City, UT, United States of America.
| | - Iftikhar J Kullo
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States of America
| | - M Caroline Morris
- School of Medicine, University of Mississippi Medical Center, Jackson, MS, United States of America
| | - Thomas H Mosley
- Department of Medicine, Division of Geriatrics, University of Mississippi Medical Center, Jackson, MS, United States of America
| |
Collapse
|
19
|
Walker KA, Gottesman RF, Coresh J, Sharrett AR, Knopman DS, Mosley TH, Alonso A, Zhou Y, Wong DF, Brown CH. Association of Surgical Hospitalization with Brain Amyloid Deposition: The Atherosclerosis Risk in Communities-Positron Emission Tomography (ARIC-PET) Study. Anesthesiology 2020; 132:1407-1418. [PMID: 32412719 PMCID: PMC7540736 DOI: 10.1097/aln.0000000000003255] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND As more older adults undergo surgery, it is critical to understand the long-term effects of surgery on brain health, particularly in relation to the development of Alzheimer's disease. This study examined the association of surgical hospitalization with subsequent brain β-amyloid deposition in nondemented older adults. METHODS The Atherosclerosis Risk in Communities-Positron Emission Tomography (ARIC-PET) study is a prospective cohort study of 346 participants without dementia who underwent florbetapir PET imaging. Active surveillance of local hospitals and annual participant contact were used to gather hospitalization and surgical information (International Classification of Disease, Ninth Revision, Clinical Modification codes) over the preceding 24-yr period. Brain amyloid measured using florbetapir PET imaging was the primary outcome. Elevated amyloid was defined as a standardized uptake value ratio of more than 1.2. RESULTS Of the 313 participants included in this analysis (age at PET: 76.0 [SD 5.4]; 56% female), 72% had a prior hospitalization, and 50% had a prior surgical hospitalization. Elevated amyloid occurred in 87 of 156 (56%) participants with previous surgical hospitalization, compared with 45 of 87 (52%) participants who had no previous hospitalization. Participants with previous surgical hospitalizations did not show an increased odds of elevated brain amyloid (odds ratio, 1.32; 95% CI, 0.72 to 2.40; P = 0.370) after adjusting for confounders (primary analysis). Results were similar using the reference group of all participants without previous surgery (hospitalized and nonhospitalized; odds ratio, 1.58; 95% CI, 0.96 to 2.58; P = 0.070). In a prespecified secondary analysis, participants with previous surgical hospitalization did demonstrate increased odds of elevated amyloid when compared with participants hospitalized without surgery (odds ratio, 2.10; 95% CI, 1.09 to 4.05; P = 0.026). However, these results were attenuated and nonsignificant when alternative thresholds for amyloid-positive status were used. CONCLUSIONS The results do not support an association between surgical hospitalization and elevated brain amyloid.
Collapse
Affiliation(s)
- Keenan A Walker
- From the Departments of Neurology (K.A.W., R.F.G.) Radiology (D.F.W.) Anesthesiology (C.H.B.), Johns Hopkins School of Medicine, Baltimore, Maryland the Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland (R.F.G., J.C., A.R.S.) the Department of Neurology, Mayo Clinic, Rochester, Minnesota (D.S.K.) Department of Medicine, Division of Geriatrics, University of Mississippi Medical Center, Jackson, Mississippi (T.H.M.) the Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia (A.A.) the Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri (Y.Z.)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Lee EE, Winston-Gray C, Barlow JW, Rissman RA, Jeste DV. Plasma Levels of Neuron- and Astrocyte-Derived Exosomal Amyloid Beta1-42, Amyloid Beta1-40, and Phosphorylated Tau Levels in Schizophrenia Patients and Non-psychiatric Comparison Subjects: Relationships With Cognitive Functioning and Psychopathology. Front Psychiatry 2020; 11:532624. [PMID: 33762974 PMCID: PMC7982803 DOI: 10.3389/fpsyt.2020.532624] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 10/13/2020] [Indexed: 12/17/2022] Open
Abstract
Introduction: Cognitive deficits in people with schizophrenia (PWS) are a major predictor of disability and functioning, yet the underlying pathophysiology remains unclear. A possible role of amyloid and tau biomarkers (hallmarks of Alzheimer's disease) is still speculative in schizophrenia. Exosomes or extracellular vesicles, involved with cell-to-cell communication and waste removal, can be used to assay brain-based proteins from peripheral blood. To our knowledge, this is the first study of exosomal amyloid and tau protein levels in PWS. Methods: This cross-sectional study included 60 PWS and 60 age- and sex-comparable non-psychiatric comparison subjects (NCs), age range 26-65 years. Assessments of global cognitive screening, executive functioning, psychopathology, and physical measures were conducted. Exosomes were extracted and precipitated from fasting plasma and identified as neuron-derived exosomes (NDEs) or astrocyte-derived exosomes (ADEs). Human-specific ELISAs were used to assay levels of amyloid-beta 1-42 (Aβ42), amyloid-beta 1-40 (Aβ40), and phosphorylated T181 tau (P-T181-tau). Plasma assays for aging biomarkers (C-reactive protein and F2-isoprostanes) were also performed. Results: ADE-Aβ42 levels were higher in PWS compared to NCs, though the other exosomal markers were similar between the two groups. Higher ADE-P-T181-tau levels were associated with worse executive functioning. Among PWS, higher ADE-P-T181-tau levels were associated with less severe negative symptoms and increased F2-isoprostane levels. Astrocyte-derived Aβ marker levels were sensitive and specific in differentiating between diagnostic groups. Among PWS, Aβ40 levels differed most by exosomal origin. Discussion: Exosomal markers may provide novel insights into brain-based processes (e.g., aging, oxidative stress) from peripheral blood samples.
Collapse
Affiliation(s)
- Ellen E Lee
- Department of Psychiatry, University of California San Diego, San Diego, CA, United States.,Veterans Affairs San Diego Healthcare System, La Jolla, CA, United States
| | - Charisse Winston-Gray
- Department of Neuroscience, University of California San Diego, San Diego, CA, United States
| | - James W Barlow
- Department of Neuroscience, University of California San Diego, San Diego, CA, United States
| | - Robert A Rissman
- Department of Neuroscience, University of California San Diego, San Diego, CA, United States
| | - Dilip V Jeste
- Department of Psychiatry and Neurosciences, University of California San Diego, San Diego, CA, United States
| |
Collapse
|
21
|
Modulation of Innate Immunity by Amyloidogenic Peptides. Trends Immunol 2019; 40:762-780. [PMID: 31320280 DOI: 10.1016/j.it.2019.06.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 06/11/2019] [Accepted: 06/12/2019] [Indexed: 12/11/2022]
Abstract
Amyloid formation contributes to the development of progressive metabolic and neurodegenerative diseases, while also serving functional roles in host defense. Emerging evidence suggests that as amyloidogenic peptides populate distinct aggregation states, they interact with different combinations of pattern recognition receptors (PRRs) to direct the phenotype and function of tissue-resident and infiltrating innate immune cells. We review recent evidence of innate immunomodulation by distinct forms of amyloidogenic peptides produced by mammals (humans, non-human primates), bacteria, and fungi, as well as the corresponding cell-surface and intracellular PRRs in these interactions, in human and mouse models. Our emerging understanding of peptide aggregate-innate immune cell interactions, and the factors regulating the balance between amyloid function and pathogenicity, might aid the development of anti-amyloid and immunomodulating therapies.
Collapse
|