1
|
Hammerschlag BL, Butts B, Likos KDS, Verble DD, Nimmagadda N, Virani R, Ramanathan S, Wharton W. Pilot: Salivary Lactoferrin as a Biomarker of Alzheimer's Disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.11.22.24317405. [PMID: 39606384 PMCID: PMC11601767 DOI: 10.1101/2024.11.22.24317405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Background Alzheimer's disease (AD) research has focused on developing accessible biomarkers that accurately detect disease pathology and progression before symptoms present. Lactoferrin (Lf) is an iron-binding antimicrobial glycoprotein found in all biological fluids, and its concentration in saliva has been correlated with AD symptoms. This pilot project aimed to determine whether salivary lactoferrin (sLF) has potential as a biomarker for AD. Methods Participants were middle to older-aged non-Hispanic white (NHW) and Black Americans (BA) at risk for AD due to parental history. We collected saliva samples after an 8-hour fast and administered a cognitive battery assessing executive function, memory, visuospatial ability, attention, and verbal fluency. We examined the relationship between sLF and cognitive performance and evaluated protein concentration across races. Results Seventeen middle-to-older-aged (age = 60.29 ± 9.7 years) BA and NHWs were enrolled. After controlling for age, sex, race, and years of education, we found a significant r between sLF and Digit Span Memory Test (DSMT) scores (P = 0.013) and a modest correlation with Mental Rotation Test scores (P = 0.194). We found no difference in average concentration across races. Conclusions Memory concerns and a worsening in visuospatial ability are early signs of cognitive decline in AD patients, and this pilot suggests a correlation of these symptoms with sLF. Bigger-scale longitudinal studies to examine the relationship between sLF and established AD biomarkers in diverse populations are needed to assess its clinical usefulness as an early biomarker for AD.
Collapse
Affiliation(s)
| | - B Butts
- Emory University, Atlanta, Georgia, USA
| | - KDS Likos
- Emory University, Atlanta, Georgia, USA
| | - DD Verble
- Emory University, Atlanta, Georgia, USA
| | | | - R Virani
- Emory University, Atlanta, Georgia, USA
| | | | - W Wharton
- Emory University, Atlanta, Georgia, USA
| |
Collapse
|
2
|
Zeng X, Lafferty TK, Sehrawat A, Chen Y, Ferreira PCL, Bellaver B, Povala G, Kamboh MI, Klunk WE, Cohen AD, Lopez OL, Ikonomovic MD, Pascoal TA, Ganguli M, Villemagne VL, Snitz BE, Karikari TK. Multi-analyte proteomic analysis identifies blood-based neuroinflammation, cerebrovascular and synaptic biomarkers in preclinical Alzheimer's disease. Mol Neurodegener 2024; 19:68. [PMID: 39385222 PMCID: PMC11465638 DOI: 10.1186/s13024-024-00753-5] [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: 04/23/2024] [Accepted: 09/04/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Blood-based biomarkers are gaining grounds for the detection of Alzheimer's disease (AD) and related disorders (ADRDs). However, two key obstacles remain: the lack of methods for multi-analyte assessments and the need for biomarkers for related pathophysiological processes like neuroinflammation, vascular, and synaptic dysfunction. A novel proteomic method for pre-selected analytes, based on proximity extension technology, was recently introduced. Referred to as the NULISAseq CNS disease panel, the assay simultaneously measures ~ 120 analytes related to neurodegenerative diseases, including those linked to both core (i.e., tau and amyloid-beta (Aβ)) and non-core AD processes. This study aimed to evaluate the technical and clinical performance of this novel targeted proteomic panel. METHODS The NULISAseq CNS disease panel was applied to 176 plasma samples from 113 individuals in the MYHAT-NI cohort of predominantly cognitively normal participants from an economically underserved region in southwestern Pennsylvania, USA. Classical AD biomarkers, including p-tau181, p-tau217, p-tau231, GFAP, NEFL, Aβ40, and Aβ42, were independently measured using Single Molecule Array (Simoa) and correlations and diagnostic performances compared. Aβ pathology, tau pathology, and neurodegeneration (AT(N) statuses) were evaluated with [11C] PiB PET, [18F]AV-1451 PET, and an MRI-based AD-signature composite cortical thickness index, respectively. Linear mixed models were used to examine cross-sectional and Wilcoxon rank sum tests for longitudinal associations between NULISA and neuroimaging-determined AT(N) biomarkers. RESULTS NULISA concurrently measured 116 plasma biomarkers with good technical performance (97.2 ± 13.9% targets gave signals above assay limits of detection), and significant correlation with Simoa assays for the classical biomarkers. Cross-sectionally, p-tau217 was the top hit to identify Aβ pathology, with age, sex, and APOE genotype-adjusted AUC of 0.930 (95%CI: 0.878-0.983). Fourteen markers were significantly decreased in Aβ-PET + participants, including TIMP3, BDNF, MDH1, and several cytokines. Longitudinally, FGF2, IL4, and IL9 exhibited Aβ PET-dependent yearly increases in Aβ-PET + participants. Novel plasma biomarkers with tau PET-dependent longitudinal changes included proteins associated with neuroinflammation, synaptic function, and cerebrovascular integrity, such as CHIT1, CHI3L1, NPTX1, PGF, PDGFRB, and VEGFA; all previously linked to AD but only reliable when measured in cerebrospinal fluid. The autophagosome cargo protein SQSTM1 exhibited significant association with neurodegeneration after adjusting age, sex, and APOE ε4 genotype. CONCLUSIONS Together, our results demonstrate the feasibility and potential of immunoassay-based multiplexing to provide a comprehensive view of AD-associated proteomic changes, consistent with the recently revised biological and diagnostic framework. Further validation of the identified inflammation, synaptic, and vascular markers will be important for establishing disease state markers in asymptomatic AD.
Collapse
Affiliation(s)
- Xuemei Zeng
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O'Hara Street, Pittsburgh, PA, 15213, USA
| | - Tara K Lafferty
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O'Hara Street, Pittsburgh, PA, 15213, USA
| | - Anuradha Sehrawat
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O'Hara Street, Pittsburgh, PA, 15213, USA
| | - Yijun Chen
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Pamela C L Ferreira
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O'Hara Street, Pittsburgh, PA, 15213, USA
| | - Bruna Bellaver
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O'Hara Street, Pittsburgh, PA, 15213, USA
| | - Guilherme Povala
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O'Hara Street, Pittsburgh, PA, 15213, USA
| | - M Ilyas Kamboh
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - William E Klunk
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O'Hara Street, Pittsburgh, PA, 15213, USA
| | - Ann D Cohen
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O'Hara Street, Pittsburgh, PA, 15213, USA
| | - Oscar L Lopez
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Milos D Ikonomovic
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O'Hara Street, Pittsburgh, PA, 15213, USA
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Geriatric Research Education and Clinical Center, VA Pittsburgh HS, Pittsburgh, PA, USA
| | - Tharick A Pascoal
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O'Hara Street, Pittsburgh, PA, 15213, USA
| | - Mary Ganguli
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O'Hara Street, Pittsburgh, PA, 15213, USA
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Victor L Villemagne
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O'Hara Street, Pittsburgh, PA, 15213, USA
| | - Beth E Snitz
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Thomas K Karikari
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O'Hara Street, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
3
|
Zuppe H, Reed E. Common cytokine receptor gamma chain family cytokines activate MAPK, PI3K, and JAK/STAT pathways in microglia to influence Alzheimer's Disease. Front Mol Neurosci 2024; 17:1441691. [PMID: 39324116 PMCID: PMC11422389 DOI: 10.3389/fnmol.2024.1441691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 08/30/2024] [Indexed: 09/27/2024] Open
Abstract
Dementia is an umbrella term used to describe deterioration of cognitive function. It is the seventh leading cause of death and is one of the major causes of dependence among older people globally. Alzheimer's Disease (AD) contributes to approximately 60-70% of dementia cases and is characterized by the accumulation of amyloid plaques and tau tangles in the brain. Neuroinflammation is now widely accepted as another disease hallmark, playing a role in both the response to and the perpetuation of disease processes. Microglia are brain-resident immune cells that are initially effective at clearing amyloid plaques but contribute to the damaging inflammatory milieu of the brain as disease progresses. Circulating peripheral immune cells contribute to this inflammatory environment through cytokine secretion, creating a positive feedback loop with the microglia. One group of these peripherally derived cytokines acting on microglia is the common cytokine receptor γ chain family. These cytokines bind heterodimer receptors to activate three major signaling pathways: MAPK, PI3K, and JAK/STAT. This perspective will look at the mechanisms of these three pathways in microglia and highlight the future directions of this research and potential therapeutics.
Collapse
Affiliation(s)
- Hannah Zuppe
- School of Biomedical Sciences, Kent State University, Kent, OH, United States
| | - Erin Reed
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States
| |
Collapse
|
4
|
Kostic M, Zivkovic N, Cvetanovic A, Basic J, Stojanovic I. Dissecting the immune response of CD4 + T cells in Alzheimer's disease. Rev Neurosci 2024:revneuro-2024-0090. [PMID: 39238424 DOI: 10.1515/revneuro-2024-0090] [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: 07/04/2024] [Accepted: 08/18/2024] [Indexed: 09/07/2024]
Abstract
The formation of amyloid-β (Aβ) plaques is a neuropathological hallmark of Alzheimer's disease (AD), however, these pathological aggregates can also be found in the brains of cognitively unimpaired elderly population. In that context, individual variations in the Aβ-specific immune response could be key factors that determine the level of Aβ-induced neuroinflammation and thus the propensity to develop AD. CD4+ T cells are the cornerstone of the immune response that coordinate the effector functions of both adaptive and innate immunity. However, despite intensive research efforts, the precise role of these cells during AD pathogenesis is still not fully elucidated. Both pathogenic and beneficial effects have been observed in various animal models of AD, as well as in humans with AD. Although this functional duality of CD4+ T cells in AD can be simply attributed to the vast phenotype heterogeneity of this cell lineage, disease stage-specific effect have also been proposed. Therefore, in this review, we summarized the current understanding of the role of CD4+ T cells in the pathophysiology of AD, from the aspect of their antigen specificity, activation, and phenotype characteristics. Such knowledge is of practical importance as it paves the way for immunomodulation as a therapeutic option for AD treatment, given that currently available therapies have not yielded satisfactory results.
Collapse
Affiliation(s)
- Milos Kostic
- Department of Immunology, Medical Faculty of Nis, University of Nis, Blvd. dr Zorana Djindjica 81, Nis, 18000, Serbia
| | - Nikola Zivkovic
- Department of Pathology, Medical Faculty of Nis, University of Nis, Blvd. dr Zorana Djindjica 81, Nis, 18000, Serbia
| | - Ana Cvetanovic
- Department of Oncology, Medical Faculty of Nis, University of Nis, Blvd. dr Zorana Djindjica 81, Nis, 18000, Serbia
| | - Jelena Basic
- Department of Biochemistry, Medical Faculty of Nis, University of Nis, Blvd. dr Zorana Djindjica 81, Nis, 18000, Serbia
| | - Ivana Stojanovic
- Department of Biochemistry, Medical Faculty of Nis, University of Nis, Blvd. dr Zorana Djindjica 81, Nis, 18000, Serbia
| |
Collapse
|
5
|
Ho G, Lam L, Tran T, Wei J, Hashimoto M. Innate neuroimmunity across aging and neurodegeneration: a perspective from amyloidogenic evolvability. Front Cell Dev Biol 2024; 12:1430593. [PMID: 39071802 PMCID: PMC11272618 DOI: 10.3389/fcell.2024.1430593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 06/17/2024] [Indexed: 07/30/2024] Open
Abstract
In Alzheimer's Disease (AD), amyloidogenic proteins (APs), such as β-amyloid (Aβ) and tau, may act as alarmins/damage-associated molecular patterns (DAMPs) to stimulate neuroinflammation and cell death. Indeed, recent evidence suggests that brain-specific type 2 immune networks may be important in modulating amyloidogenicity and brain homeostasis. Central to this, components of innate neuroimmune signaling, particularly type 2 components, assume distinctly specialized roles in regulating immune homeostasis and brain function. Whereas balanced immune surveillance stems from normal type 2 brain immune function, appropriate microglial clearance of aggregated misfolded proteins and neurotrophic and synaptotrophic signaling, aberrant pro-inflammatory activity triggered by alarmins might disrupt this normal immune homeostasis with reduced microglial amyloid clearance, synaptic loss, and ultimately neurodegeneration. Furthermore, since increased inflammation may in turn cause neurodegeneration, it is predicted that AP aggregation and neuroinflammation could synergistically promote even more damage. The reasons for maintaining such adverse biological conditions which have not been weeded out during evolution remain unclear. Here, we discuss these issues from a viewpoint of amyloidogenic evolvability, namely, aEVO, a hypothetic view of an adaptation to environmental stress by AP aggregates. Speculatively, the interaction of AP aggregation and neuroinflammation for aEVO in reproduction, which is evolutionally beneficial, might become a co-activating relationship which promotes AD pathogenesis through antagonistic pleiotropy. If validated, simultaneously suppressing both AP aggregation and specific innate neuroinflammation could greatly increase therapeutic efficacy in AD. Overall, combining a better understanding of innate neuroimmunity in aging and disease with the aEVO hypothesis may help uncover novel mechanism of pathogenesis of AD, leading to improved diagnostics and treatments.
Collapse
Affiliation(s)
- Gilbert Ho
- PCND Neuroscience Research Institute, Poway, CA, United States
| | - Linh Lam
- PCND Neuroscience Research Institute, Poway, CA, United States
| | - Tony Tran
- PCND Neuroscience Research Institute, Poway, CA, United States
| | - Jianshe Wei
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, China
| | | |
Collapse
|
6
|
Zeng X, Lafferty TK, Sehrawat A, Chen Y, Ferreira PCL, Bellaver B, Povala G, Kamboh MI, Klunk WE, Cohen AD, Lopez OL, Ikonomovic MD, Pascoal TA, Ganguli M, Villemagne VL, Snitz BE, Karikari TK. Multi-analyte proteomic analysis identifies blood-based neuroinflammation, cerebrovascular and synaptic biomarkers in preclinical Alzheimer's disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.15.24308975. [PMID: 38947065 PMCID: PMC11213097 DOI: 10.1101/2024.06.15.24308975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Background Blood-based biomarkers are gaining grounds for Alzheimer's disease (AD) detection. However, two key obstacles need to be addressed: the lack of methods for multi-analyte assessments and the need for markers of neuroinflammation, vascular, and synaptic dysfunction. Here, we evaluated a novel multi-analyte biomarker platform, NULISAseq CNS disease panel, a multiplex NUcleic acid-linked Immuno-Sandwich Assay (NULISA) targeting ~120 analytes, including classical AD biomarkers and key proteins defining various disease hallmarks. Methods The NULISAseq panel was applied to 176 plasma samples from the MYHAT-NI cohort of cognitively normal participants from an economically underserved region in Western Pennsylvania. Classical AD biomarkers, including p-tau181 p-tau217, p-tau231, GFAP, NEFL, Aβ40, and Aβ42, were also measured using Single Molecule Array (Simoa). Amyloid pathology, tau pathology, and neurodegeneration were evaluated with [11C] PiB PET, [18F]AV-1451 PET, and MRI, respectively. Linear mixed models were used to examine cross-sectional and Wilcoxon rank sum tests for longitudinal associations between NULISA biomarkers and AD pathologies. Spearman correlations were used to compare NULISA and Simoa. Results NULISA concurrently measured 116 plasma biomarkers with good technical performance, and good correlation with Simoa measures. Cross-sectionally, p-tau217 was the top hit to identify Aβ pathology, with age, sex, and APOE genotype-adjusted AUC of 0.930 (95%CI: 0.878-0.983). Fourteen markers were significantly decreased in Aβ-PET+ participants, including TIMP3, which regulates brain Aβ production, the neurotrophic factor BDNF, the energy metabolism marker MDH1, and several cytokines. Longitudinally, FGF2, IL4, and IL9 exhibited Aβ PET-dependent yearly increases in Aβ-PET+ participants. Markers with tau PET-dependent longitudinal changes included the microglial activation marker CHIT1, the reactive astrogliosis marker CHI3L1, the synaptic protein NPTX1, and the cerebrovascular markers PGF, PDGFRB, and VEFGA; all previously linked to AD but only reliably measured in cerebrospinal fluid. SQSTM1, the autophagosome cargo protein, exhibited a significant association with neurodegeneration status after adjusting age, sex, and APOE ε4 genotype. Conclusions Together, our results demonstrate the feasibility and potential of immunoassay-based multiplexing to provide a comprehensive view of AD-associated proteomic changes. Further validation of the identified inflammation, synaptic, and vascular markers will be important for establishing disease state markers in asymptomatic AD.
Collapse
Affiliation(s)
- Xuemei Zeng
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O’Hara Street, Pittsburgh, PA 15213, USA
| | - Tara K. Lafferty
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O’Hara Street, Pittsburgh, PA 15213, USA
| | - Anuradha Sehrawat
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O’Hara Street, Pittsburgh, PA 15213, USA
| | - Yijun Chen
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Pamela C. L. Ferreira
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O’Hara Street, Pittsburgh, PA 15213, USA
| | - Bruna Bellaver
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O’Hara Street, Pittsburgh, PA 15213, USA
| | - Guilherme Povala
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O’Hara Street, Pittsburgh, PA 15213, USA
| | - M. Ilyas Kamboh
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - William E. Klunk
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O’Hara Street, Pittsburgh, PA 15213, USA
| | - Ann D. Cohen
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O’Hara Street, Pittsburgh, PA 15213, USA
| | - Oscar L. Lopez
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Milos D. Ikonomovic
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O’Hara Street, Pittsburgh, PA 15213, USA
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Geriatric Research Education and Clinical Center, VA Pittsburgh HS, Pittsburgh, PA, USA
| | - Tharick A. Pascoal
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O’Hara Street, Pittsburgh, PA 15213, USA
| | - Mary Ganguli
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O’Hara Street, Pittsburgh, PA 15213, USA
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Victor L. Villemagne
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O’Hara Street, Pittsburgh, PA 15213, USA
| | - Beth E. Snitz
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Thomas K. Karikari
- Department of Psychiatry, School of Medicine, University of Pittsburgh, 3811 O’Hara Street, Pittsburgh, PA 15213, USA
| |
Collapse
|
7
|
Ball BK, Kuhn MK, Fleeman Bechtel RM, Proctor EA, Brubaker DK. Differential responses of primary neuron-secreted MCP-1 and IL-9 to type 2 diabetes and Alzheimer's disease-associated metabolites. Sci Rep 2024; 14:12743. [PMID: 38830911 PMCID: PMC11148169 DOI: 10.1038/s41598-024-62155-3] [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: 12/01/2023] [Accepted: 05/14/2024] [Indexed: 06/05/2024] Open
Abstract
Type 2 diabetes (T2D) is implicated as a risk factor for Alzheimer's disease (AD), the most common form of dementia. In this work, we investigated neuroinflammatory responses of primary neurons to potentially circulating, blood-brain barrier (BBB) permeable metabolites associated with AD, T2D, or both. We identified nine metabolites associated with protective or detrimental properties of AD and T2D in literature (lauric acid, asparagine, fructose, arachidonic acid, aminoadipic acid, sorbitol, retinol, tryptophan, niacinamide) and stimulated primary mouse neuron cultures with each metabolite before quantifying cytokine secretion via Luminex. We employed unsupervised clustering, inferential statistics, and partial least squares discriminant analysis to identify relationships between cytokine concentration and disease-associations of metabolites. We identified MCP-1, a cytokine associated with monocyte recruitment, as differentially abundant between neurons stimulated by metabolites associated with protective and detrimental properties of AD and T2D. We also identified IL-9, a cytokine that promotes mast cell growth, to be differentially associated with T2D. Indeed, cytokines, such as MCP-1 and IL-9, released from neurons in response to BBB-permeable metabolites associated with T2D may contribute to AD development by downstream effects of neuroinflammation.
Collapse
Affiliation(s)
- Brendan K Ball
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Madison K Kuhn
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
- Department of Biomedical Engineering, Penn State University, State College, PA, USA
- Center for Neural Engineering, Penn State University, State College, PA, USA
| | - Rebecca M Fleeman Bechtel
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
| | - Elizabeth A Proctor
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
- Department of Biomedical Engineering, Penn State University, State College, PA, USA
- Center for Neural Engineering, Penn State University, State College, PA, USA
- Department of Engineering Science & Mechanics, Penn State University, State College, PA, USA
| | - Douglas K Brubaker
- Center for Global Health & Diseases, Department of Pathology, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
- Blood Heart Lung Immunology Research Center, University Hospitals, Cleveland, OH, USA.
| |
Collapse
|
8
|
Hu WT, Kaluzova M, Dawson A, Sotelo V, Papas J, Lemenze A, Shu C, Jomartin M, Nayyar A, Hussain S. Clinical and CSF single-cell profiling of post-COVID-19 cognitive impairment. Cell Rep Med 2024; 5:101561. [PMID: 38744274 PMCID: PMC11148803 DOI: 10.1016/j.xcrm.2024.101561] [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: 08/22/2023] [Revised: 02/15/2024] [Accepted: 04/17/2024] [Indexed: 05/16/2024]
Abstract
Natural history and mechanisms for persistent cognitive symptoms ("brain fog") following acute and often mild COVID-19 are unknown. In a large prospective cohort of people who underwent testing a median of 9 months after acute COVID-19 in the New York City/New Jersey area, we found that cognitive dysfunction is common; is not influenced by mood, fatigue, or sleepiness; and is correlated with MRI changes in very few people. In a subgroup that underwent cerebrospinal fluid analysis, there are no changes related to Alzheimer's disease or neurodegeneration. Single-cell gene expression analysis in the cerebrospinal fluid shows findings consistent with monocyte recruitment, chemokine signaling, cellular stress, and suppressed interferon response-especially in myeloid cells. Longitudinal analysis shows slow recovery accompanied by key alterations in inflammatory genes and increased protein levels of CXCL8, CCL3L1, and sTREM2. These findings suggest that the prognosis for brain fog following COVID-19 correlates with myeloid-related chemokine and interferon-responsive genes.
Collapse
Affiliation(s)
- William T Hu
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Center for Innovation in Health and Aging Research, Institute for Health, Health Care Policy, and Aging Research, New Brunswick, NJ, USA.
| | - Milota Kaluzova
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Alice Dawson
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Center for Innovation in Health and Aging Research, Institute for Health, Health Care Policy, and Aging Research, New Brunswick, NJ, USA
| | - Victor Sotelo
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Center for Innovation in Health and Aging Research, Institute for Health, Health Care Policy, and Aging Research, New Brunswick, NJ, USA
| | - Julia Papas
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Center for Innovation in Health and Aging Research, Institute for Health, Health Care Policy, and Aging Research, New Brunswick, NJ, USA
| | - Alexander Lemenze
- Department of Pathology and Laboratory Medicine, Rutgers-New Jersey Medical School, Newark, NJ, USA
| | - Carol Shu
- Department of Medicine-Pulmonary and Critical Care, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Mini Jomartin
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Ashima Nayyar
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Sabiha Hussain
- Department of Medicine-Pulmonary and Critical Care, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| |
Collapse
|
9
|
Maestre G, Hill C, Griffin P, Hall S, Hu W, Flatt J, Babulal G, Thorpe R, Henderson JN, Buchwald D, Manson S, Cicero E, Gilmore‐Bykovskyi A, Gamaldo A, Glover C, Barnes L, Kind A, James B, Zeki Al Hazzouri A, Wharton W, Caramelli P, Szanton S, Whitmer R, Benn Torres J, Deters K, Okonkwo O, Das R, Martinez‐Gonzalez K, Carrillo M. Promoting diverse perspectives: Addressing health disparities related to Alzheimer's and all dementias. Alzheimers Dement 2024; 20:3099-3107. [PMID: 38460119 PMCID: PMC11032522 DOI: 10.1002/alz.13752] [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: 11/10/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 03/11/2024]
Abstract
Dementia research lacks appropriate representation of diverse groups who often face substantial adversity and greater risk of dementia. Current research participants are primarily well-resourced, non-Hispanic White, cisgender adults who live close to academic medical centers where much of the research is based. Consequently, the field faces a knowledge gap about Alzheimer's-related risk factors in those other groups. The Alzheimer's Association hosted a virtual conference on June 14-16, 2021, supported in part by the National Institute on Aging (R13 AG072859-01), focused on health disparities. The conference was held entirely online and consisted of 2 days of core programming and a day of focused meetings centered on American Indian and Alaska Natives and on LGBTQIA+ populations. Over 1300 registrants attended discussions focused on the structural and systemic inequities experienced across diverse groups, as well as ways to investigate and address these inequities.
Collapse
Affiliation(s)
- Gladys Maestre
- School of MedicineAlzheimer's Disease Resource Center for Minority Aging ResearchUniversity of Texas Rio Grande ValleyBrownsvilleTexasUSA
| | - Carl Hill
- Medical & Scientific RelationsAlzheimer's AssociationChicagoIllinoisUSA
| | - Percy Griffin
- Medical & Scientific RelationsAlzheimer's AssociationChicagoIllinoisUSA
| | - Stephen Hall
- Medical & Scientific RelationsAlzheimer's AssociationChicagoIllinoisUSA
| | - William Hu
- Rutgers Robert Wood Johnson Medical School and Rutgers Institute for HealthHealth Care Policy, and Aging ResearchNew BrunswickNew JerseyUSA
| | - Jason Flatt
- Department of Social and Behavioral HealthSchool of Public HealthUniversity of Nevada Las VegasLas VegasNevadaUSA
| | - Ganesh Babulal
- Department of NeurologySchool of MedicineWashington University in St. LouisSt. LouisMissouriUSA
| | - Roland Thorpe
- Center on AgingCenter on Health Disparities SolutionsHopkins Population CenterAlzheimer's Disease Resource Center for Minority Aging ResearchJohns Hopkins University Bloomberg School of Public HealthBaltimoreMarylandUSA
| | | | - Dedra Buchwald
- Institute for Research and Education to Advance Community Health Elson S Floyd College of Medicine Washington State UniversitySeattleWashingtonUSA
| | - Spero Manson
- Centers for American Indian and Alaska Native HealthUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Ethan Cicero
- Nell Hodgson Woodruff School of NursingEmory UniversityAtlantaGeorgiaUSA
| | - Andrea Gilmore‐Bykovskyi
- BerbeeWalsh Department of Emergency MedicineUniversity of Wisconsin‐Madison School of Medicine and Public HealthMadisonWisconsinUSA
| | - Alyssa Gamaldo
- Pennsylvania State UniversityState CollegePennsylvaniaUSA
| | | | - Lisa Barnes
- Rush University Medical CenterChicagoIllinoisUSA
| | - Amy Kind
- BerbeeWalsh Department of Emergency MedicineUniversity of Wisconsin‐Madison School of Medicine and Public HealthMadisonWisconsinUSA
| | - Bryan James
- Rush University Medical CenterChicagoIllinoisUSA
| | - Adina Zeki Al Hazzouri
- Mailman School of Public HealthDepartment of EpidemiologyColumbia UniversityNew YorkNew YorkUSA
| | - Whitney Wharton
- Nell Hodgson Woodruff School of NursingEmory UniversityAtlantaGeorgiaUSA
| | - Paulo Caramelli
- Behavioral and Cognitive Neurology Unit, Faculdade de MedicinaUniversidade Federal de Minas GeraisBelo HorizonteBrazil
| | - Sarah Szanton
- Johns Hopkins University School of NursingBaltimoreMarylandUSA
| | - Rachel Whitmer
- Department of Public Health SciencesDepartment of NeurologyUniversity of California DavisDavisCaliforniaUSA
| | | | - Kacie Deters
- Department of NeurosciencesUniversity of California San Diego School of MedicineLa JollaCaliforniaUSA
| | - Ozioma Okonkwo
- Department of Medicine and the Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Rina Das
- National Institute on Minority Health and Health DisparitiesBethesdaMarylandUSA
| | | | - Maria Carrillo
- Medical & Scientific RelationsAlzheimer's AssociationChicagoIllinoisUSA
| |
Collapse
|
10
|
Ikanga J, Patrick SD, Schwinne M, Patel SS, Epenge E, Gikelekele G, Tshengele N, Kavugho I, Mampunza S, Yarasheski KE, Teunissen CE, Stringer A, Levey A, Rojas JC, Chan B, Lario Lago A, Kramer JH, Boxer AL, Jeromin A, Alonso A, Spencer RJ. Sensitivity of the African neuropsychology battery memory subtests and learning slopes in discriminating APOE 4 and amyloid pathology in adult individuals in the Democratic Republic of Congo. Front Neurol 2024; 15:1320727. [PMID: 38601333 PMCID: PMC11004441 DOI: 10.3389/fneur.2024.1320727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 03/14/2024] [Indexed: 04/12/2024] Open
Abstract
Background The current study examined the sensitivity of two memory subtests and their corresponding learning slope metrics derived from the African Neuropsychology Battery (ANB) to detect amyloid pathology and APOEε4 status in adults from Kinshasa, the Democratic Republic of the Congo. Methods 85 participants were classified for the presence of β-amyloid pathology and based on allelic presence of APOEε4 using Simoa. All participants were screened using CSID and AQ, underwent verbal and visuospatial memory testing from ANB, and provided blood samples for plasma Aβ42, Aβ40, and APOE proteotype. Pearson correlation, linear and logistic regression were conducted to compare amyloid pathology and APOEε4 status with derived learning scores, including initial learning, raw learning score, learning over trials, and learning ratio. Results Our sample included 35 amyloid positive and 44 amyloid negative individuals as well as 42 without and 39 with APOEε4. All ROC AUC ranges for the prediction of amyloid pathology based on learning scores were low, ranging between 0.56-0.70 (95% CI ranging from 0.44-0.82). The sensitivity of all the scores ranged between 54.3-88.6, with some learning metrics demonstrating good sensitivity. Regarding APOEε4 prediction, all AUC values ranged between 0.60-0.69, with all sensitivity measures ranging between 53.8-89.7. There were minimal differences in the AUC values across learning slope metrics, largely due to the lack of ceiling effects in this sample. Discussion This study demonstrates that some ANB memory subtests and learning slope metrics can discriminate those that are normal from those with amyloid pathology and those with and without APOEε4, consistent with findings reported in Western populations.
Collapse
Affiliation(s)
- Jean Ikanga
- Department of Rehabilitation Medicine, Emory University School of Medicine, Atlanta, GA, United States
- Department of Psychiatry, School of Medicine, University of Kinshasa and Catholic University of Congo, Kinshasa, Democratic Republic of Congo
| | - Sarah D. Patrick
- Veteran Affairs Ann Arbor Healthcare System, Ann Arbor, MI, United States
| | - Megan Schwinne
- Department of Biomedical Informatics, School of Medicine, Emory University, Atlanta, GA, United States
| | - Saranya Sundaram Patel
- Department of Rehabilitation Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Emmanuel Epenge
- Department of Neurology, University of Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Guy Gikelekele
- Department of Psychiatry, School of Medicine, University of Kinshasa and Catholic University of Congo, Kinshasa, Democratic Republic of Congo
| | - Nathan Tshengele
- Department of Psychiatry, School of Medicine, University of Kinshasa and Catholic University of Congo, Kinshasa, Democratic Republic of Congo
| | | | - Samuel Mampunza
- Department of Psychiatry, School of Medicine, University of Kinshasa and Catholic University of Congo, Kinshasa, Democratic Republic of Congo
| | | | - Charlotte E. Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Neurodegeneration, Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, Netherlands
| | - Anthony Stringer
- Department of Rehabilitation Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Allan Levey
- Department of Neurology, School of Medicine, Emory University, Atlanta, GA, United States
| | - Julio C. Rojas
- Department of Neurology, University of San Francisco, Memory and Aging Center, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, United States
| | - Brandon Chan
- Department of Neurology, University of San Francisco, Memory and Aging Center, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, United States
| | - Argentina Lario Lago
- Department of Neurology, University of San Francisco, Memory and Aging Center, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, United States
| | - Joel H. Kramer
- Department of Neurology, University of San Francisco, Memory and Aging Center, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, United States
| | - Adam L. Boxer
- Department of Neurology, University of San Francisco, Memory and Aging Center, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, United States
| | | | - Alvaro Alonso
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Robert J. Spencer
- Veteran Affairs Ann Arbor Healthcare System, Ann Arbor, MI, United States
| |
Collapse
|
11
|
Ball BK, Kuhn MK, Fleeman RM, Proctor EA, Brubaker DK. Differential responses of primary neuron-secreted MCP-1 and IL-9 to type 2 diabetes and Alzheimer's disease-associated metabolites. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.17.567595. [PMID: 38014333 PMCID: PMC10680853 DOI: 10.1101/2023.11.17.567595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Type 2 diabetes (T2D) is implicated as a risk factor for Alzheimer's disease (AD), the most common form of dementia. In this work, we investigated neuroinflammatory responses of primary neurons to potentially circulating, blood-brain barrier (BBB) permeable metabolites associated with AD, T2D, or both. We identified nine metabolites associated with protective or detrimental properties of AD and T2D in literature (lauric acid, asparagine, fructose, arachidonic acid, aminoadipic acid, sorbitol, retinol, tryptophan, niacinamide) and stimulated primary mouse neuron cultures with each metabolite before quantifying cytokine secretion via Luminex. We employed unsupervised clustering, inferential statistics, and partial least squares discriminant analysis to identify relationships between cytokine concentration and disease-associations of metabolites. We identified MCP-1, a cytokine associated with monocyte recruitment, as differentially abundant between neurons stimulated by metabolites associated with protective and detrimental properties of AD and T2D. We also identified IL-9, a cytokine that promotes mast cell growth, to be differentially associated with T2D. Indeed, cytokines, such as MCP-1 and IL-9, released from neurons in response to BBB-permeable metabolites associated with T2D may contribute to AD development by downstream effects of neuroinflammation.
Collapse
Affiliation(s)
- Brendan K. Ball
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Madison K. Kuhn
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
- Department of Biomedical Engineering, Penn State University, State College, PA, USA
- Center for Neural Engineering, Penn State University, State College, PA, USA
| | - Rebecca M. Fleeman
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
| | - Elizabeth A. Proctor
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
- Department of Biomedical Engineering, Penn State University, State College, PA, USA
- Center for Neural Engineering, Penn State University, State College, PA, USA
- Department of Engineering Science & Mechanics, Penn State University, State College, PA, USA
| | - Douglas K. Brubaker
- Center for Global Health & Diseases, Department of Pathology, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Blood Heart Lung Immunology Research Center, University Hospitals, Cleveland, OH, USA
| |
Collapse
|
12
|
Afsar A, Chen M, Xuan Z, Zhang L. A glance through the effects of CD4 + T cells, CD8 + T cells, and cytokines on Alzheimer's disease. Comput Struct Biotechnol J 2023; 21:5662-5675. [PMID: 38053545 PMCID: PMC10694609 DOI: 10.1016/j.csbj.2023.10.058] [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: 07/31/2023] [Revised: 10/31/2023] [Accepted: 10/31/2023] [Indexed: 12/07/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia. Unfortunately, despite numerous studies, an effective treatment for AD has not yet been established. There is remarkable evidence indicating that the innate immune mechanism and adaptive immune response play significant roles in the pathogenesis of AD. Several studies have reported changes in CD8+ and CD4+ T cells in AD patients. This mini-review article discusses the potential contribution of CD4+ and CD8+ T cells reactivity to amyloid β (Aβ) protein in individuals with AD. Moreover, this mini-review examines the potential associations between T cells, heme oxygenase (HO), and impaired mitochondria in the context of AD. While current mathematical models of AD have not extensively addressed the inclusion of CD4+ and CD8+ T cells, there exist models that can be extended to consider AD as an autoimmune disease involving these T cell types. Additionally, the mini-review covers recent research that has investigated the utilization of machine learning models, considering the impact of CD4+ and CD8+ T cells.
Collapse
Affiliation(s)
- Atefeh Afsar
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Min Chen
- Department of Mathematical Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Zhenyu Xuan
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Li Zhang
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX, USA
| |
Collapse
|
13
|
Benjamin KJM, Chen Q, Eagles NJ, Huuki-Myers LA, Collado-Torres L, Stolz JM, Pertea G, Shin JH, Paquola ACM, Hyde TM, Kleinman JE, Jaffe AE, Han S, Weinberger DR. Genetic and environmental contributions to ancestry differences in gene expression in the human brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.28.534458. [PMID: 37034760 PMCID: PMC10081196 DOI: 10.1101/2023.03.28.534458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Ancestral differences in genomic variation are determining factors in gene regulation; however, most gene expression studies have been limited to European ancestry samples or adjusted for ancestry to identify ancestry-independent associations. We instead examined the impact of genetic ancestry on gene expression and DNA methylation (DNAm) in admixed African/Black American neurotypical individuals to untangle effects of genetic and environmental factors. Ancestry-associated differentially expressed genes (DEGs), transcripts, and gene networks, while notably not implicating neurons, are enriched for genes related to immune response and vascular tissue and explain up to 26% of heritability for ischemic stroke, 27% of heritability for Parkinson's disease, and 30% of heritability for Alzhemier's disease. Ancestry-associated DEGs also show general enrichment for heritability of diverse immune-related traits but depletion for psychiatric-related traits. The cell-type enrichments and direction of effects vary by brain region. These DEGs are less evolutionarily constrained and are largely explained by genetic variations; roughly 15% are predicted by DNAm variation implicating environmental exposures. We also compared Black and White Americans, confirming most of these ancestry-associated DEGs. Our results highlight how environment and genetic background affect genetic ancestry differences in gene expression in the human brain and affect risk for brain illness.
Collapse
Affiliation(s)
- Kynon J M Benjamin
- Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Qiang Chen
- Lieber Institute for Brain Development, Baltimore, MD, USA
| | | | | | - Leonardo Collado-Torres
- Lieber Institute for Brain Development, Baltimore, MD, USA
- Center for Computational Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Joshua M Stolz
- Lieber Institute for Brain Development, Baltimore, MD, USA
| | - Geo Pertea
- Lieber Institute for Brain Development, Baltimore, MD, USA
| | - Joo Heon Shin
- Lieber Institute for Brain Development, Baltimore, MD, USA
| | - Apuã C M Paquola
- Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thomas M Hyde
- Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Joel E Kleinman
- Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Andrew E Jaffe
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Neumora Therapeutics, Watertown, MA, USA
| | - Shizhong Han
- Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Daniel R Weinberger
- Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
14
|
Lutshumba J, Wilcock DM, Monson NL, Stowe AM. Sex-based differences in effector cells of the adaptive immune system during Alzheimer's disease and related dementias. Neurobiol Dis 2023; 184:106202. [PMID: 37330146 PMCID: PMC10481581 DOI: 10.1016/j.nbd.2023.106202] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 06/12/2023] [Accepted: 06/13/2023] [Indexed: 06/19/2023] Open
Abstract
Neurological conditions such as Alzheimer's disease (AD) and related dementias (ADRD) present with many challenges due to the heterogeneity of the related disease(s), making it difficult to develop effective treatments. Additionally, the progression of ADRD-related pathologies presents differently between men and women. With two-thirds of the population affected with ADRD being women, ADRD has presented itself with a bias toward the female population. However, studies of ADRD generally do not incorporate sex-based differences in investigating the development and progression of the disease, which is detrimental to understanding and treating dementia. Additionally, recent implications for the adaptive immune system in the development of ADRD bring in new factors to be considered as part of the disease, including sex-based differences in immune response(s) during ADRD development. Here, we review the sex-based differences of pathological hallmarks of ADRD presentation and progression, sex-based differences in the adaptive immune system and how it changes with ADRD, and the importance of precision medicine in the development of a more targeted and personalized treatment for this devastating and prevalent neurodegenerative condition.
Collapse
Affiliation(s)
- Jenny Lutshumba
- Department of Neurology, College of Medicine, University of Kentucky, Lexington, KY, United States of America
| | - Donna M Wilcock
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States of America; Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, United States of America
| | - Nancy L Monson
- Department of Neurology and Immunology, University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | - Ann M Stowe
- Department of Neurology, College of Medicine, University of Kentucky, Lexington, KY, United States of America; Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, KY, United States of America.
| |
Collapse
|
15
|
Kuhn MK, Fleeman RM, Beidler LM, Snyder AM, Chan DC, Proctor EA. Amyloid-β Pathology-Specific Cytokine Secretion Suppresses Neuronal Mitochondrial Metabolism. Cell Mol Bioeng 2023; 16:405-421. [PMID: 37811007 PMCID: PMC10550897 DOI: 10.1007/s12195-023-00782-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 08/28/2023] [Indexed: 10/10/2023] Open
Abstract
Introduction Neuroinflammation and metabolic dysfunction are early alterations in Alzheimer's disease (AD) brain that are thought to contribute to disease onset and progression. Glial activation due to protein deposition results in cytokine secretion and shifts in brain metabolism, which have been observed in AD patients. However, the mechanism by which this immunometabolic feedback loop can injure neurons and cause neurodegeneration remains unclear. Methods We used Luminex XMAP technology to quantify hippocampal cytokine concentrations in the 5xFAD mouse model of AD at milestone timepoints in disease development. We used partial least squares regression to build cytokine signatures predictive of disease progression, as compared to healthy aging in wild-type littermates. We applied the disease-defining cytokine signature to wild-type primary neuron cultures and measured downstream changes in gene expression using the NanoString nCounter system and mitochondrial function using the Seahorse Extracellular Flux live-cell analyzer. Results We identified a pattern of up-regulated IFNγ, IP-10/CXCL10, and IL-9 as predictive of advanced disease. When healthy neurons were exposed to these cytokines in proportions found in diseased brain, gene expression of mitochondrial electron transport chain complexes, including ATP synthase, was suppressed. In live cells, basal and maximal mitochondrial respiration were impaired following cytokine stimulation. Conclusions We identify a pattern of cytokine secretion predictive of progressing amyloid-β pathology in the 5xFAD mouse model of AD that reduces expression of mitochondrial electron transport complexes and impairs mitochondrial respiration in healthy neurons. We establish a mechanistic link between disease-specific immune cues and impaired neuronal metabolism, potentially causing neuronal vulnerability and susceptibility to degeneration in AD. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-023-00782-y.
Collapse
Affiliation(s)
- Madison K. Kuhn
- Department of Neurosurgery, Penn State College of Medicine, Hershey, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA USA
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA USA
- Center for Neural Engineering, Pennsylvania State University, University Park, PA USA
| | - Rebecca M. Fleeman
- Department of Neurosurgery, Penn State College of Medicine, Hershey, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA USA
| | - Lynne M. Beidler
- Department of Microbiology & Immunology, Penn State College of Medicine, Hershey, PA USA
| | - Amanda M. Snyder
- Department of Neurology, Penn State College of Medicine, Hershey, PA USA
| | - Dennis C. Chan
- Department of Neurosurgery, Penn State College of Medicine, Hershey, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA USA
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA USA
- Center for Neural Engineering, Pennsylvania State University, University Park, PA USA
| | - Elizabeth A. Proctor
- Department of Neurosurgery, Penn State College of Medicine, Hershey, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA USA
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA USA
- Department of Engineering Science & Mechanics, Pennsylvania State University, University Park, PA USA
- Center for Neural Engineering, Pennsylvania State University, University Park, PA USA
| |
Collapse
|
16
|
Lim AC, Barnes LL, Weissberger GH, Lamar M, Nguyen AL, Fenton L, Herrera J, Han SD. Quantification of race/ethnicity representation in Alzheimer's disease neuroimaging research in the USA: a systematic review. COMMUNICATIONS MEDICINE 2023; 3:101. [PMID: 37491471 PMCID: PMC10368705 DOI: 10.1038/s43856-023-00333-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 07/05/2023] [Indexed: 07/27/2023] Open
Abstract
BACKGROUND Racial and ethnic minoritized groups are disproportionately at risk for Alzheimer's Disease (AD), but are not sufficiently recruited in AD neuroimaging research in the United States. This is important as sample composition impacts generalizability of findings, biomarker cutoffs, and treatment effects. No studies have quantified the breadth of race/ethnicity representation in the AD literature. METHODS This review identified median race/ethnicity composition of AD neuroimaging US-based research samples available as free full-text articles on PubMed. Two types of published studies were analyzed: studies that directly report race/ethnicity data (i.e., direct studies), and studies that do not report race/ethnicity but used data from a cohort study/database that does report this information (i.e., indirect studies). RESULTS Direct studies (n = 719) have median representation of 88.9% white or 87.4% Non-Hispanic white, 7.3% Black/African American, and 3.4% Hispanic/Latino ethnicity, with 0% Asian American, Native Hawaiian/Pacific Islander, and American Indian/Alaska Native, Multiracial, and Other Race participants. Cohort studies/databases (n = 44) from which indirect studies (n = 1745) derived are more diverse, with median representation of 84.2% white, 83.7% Non-Hispanic white, 11.6% Black/African American, 4.7% Hispanic/Latino, and 1.75% Asian American participants. Notably, 94% of indirect studies derive from just 10 cohort studies/databases. Comparisons of two time periods using a median split for publication year, 1994-2017 and 2018-2022, indicate that sample diversity has improved recently, particularly for Black/African American participants (3.39% from 1994-2017 and 8.29% from 2018-2022). CONCLUSIONS There is still underrepresentation of all minoritized groups relative to Census data, especially for Hispanic/Latino and Asian American individuals. The AD neuroimaging literature will benefit from increased representative recruitment of ethnic/racial minorities. More transparent reporting of race/ethnicity data is needed.
Collapse
Affiliation(s)
- Aaron C Lim
- Department of Family Medicine, Keck School of Medicine of USC, Alhambra, CA, USA
| | - Lisa L Barnes
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Gali H Weissberger
- The Interdisciplinary Department of Social Sciences, Bar-Ilan University, Raman Gat, Israel
| | - Melissa Lamar
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Annie L Nguyen
- Department of Family Medicine, Keck School of Medicine of USC, Alhambra, CA, USA
| | - Laura Fenton
- Department of Psychology, USC Dornsife College of Letters, Arts, and Sciences, Los Angeles, CA, USA
| | - Jennifer Herrera
- Department of Family Medicine, Keck School of Medicine of USC, Alhambra, CA, USA
| | - S Duke Han
- Department of Family Medicine, Keck School of Medicine of USC, Alhambra, CA, USA.
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA.
- Department of Psychology, USC Dornsife College of Letters, Arts, and Sciences, Los Angeles, CA, USA.
- USC School of Gerontology, Los Angeles, CA, USA.
- Department of Neurology, Keck School of Medicine of USC, Los Angeles, CA, USA.
| |
Collapse
|
17
|
Giannisis A, Al-Grety A, Carlsson H, Howell JC, Hu WT, Kultima K, Nielsen HM. Plasma apolipoprotein E levels, isoform composition, and dimer profile in relation to plasma lipids in racially diverse patients with Alzheimer's disease and mild cognitive impairment. Alzheimers Res Ther 2023; 15:119. [PMID: 37400888 PMCID: PMC10316569 DOI: 10.1186/s13195-023-01262-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 06/14/2023] [Indexed: 07/05/2023]
Abstract
BACKGROUND The APOEε4-promoted risk of Alzheimer's disease (AD) is lower in Black/African-Americans (B/AAs), compared to non-Hispanic whites (NHWs). Previous studies reported lower plasma apolipoprotein E (apoE) levels in NHW APOEε4-carriers compared to non-carriers, and low plasma apoE levels were directly associated with an increased risk of AD and all dementia. We further showed that APOEε3/ε3 AD patients exhibited reduced plasma apoE dimers compared to corresponding control subjects. Whether plasma apoE levels and apoE dimer formation differ between races/ethnicities and therefore may help explain AD risk racial disparity remains to be elucidated. METHODS Using mass spectrometry, we determined total plasma apoE and apoE isoform levels in a cohort of B/AAs (n = 58) and NHWs (n = 67) including subjects with normal cognition (B/AA: n = 25, NHW: n = 28), mild cognitive impairment (MCI) (B/AA: n = 24, NHW: n = 24), or AD dementia (B/AA: n = 9, NHW: n = 15). Additionally, we used non-reducing western blot analysis to assess the distribution of plasma apoE into monomers/disulfide-linked dimers. Plasma total apoE, apoE isoform levels, and % apoE monomers/dimers were assessed for correlations with cognition, cerebrospinal fluid (CSF) AD biomarkers, sTREM2, neurofilament light protein (NfL), and plasma lipids. RESULTS Plasma apoE was predominantly monomeric in both racial groups and the monomer/dimer distribution was not affected by disease status, or correlated with CSF AD biomarkers, but associated with plasma lipids. Plasma total apoE levels were not related to disease status and only in the NHW subjects we observed lower plasma apoE levels in the APOEε4/ε4-carriers. Total plasma apoE levels were 13% higher in B/AA compared to NHW APOEε4/ε4 subjects and associated with plasma high-density lipoprotein (HDL) in NHW subjects but with low-density lipoprotein levels (LDL) in the B/AA subjects. Higher plasma apoE4 levels, exclusively in APOEε3/ε4 B/AA subjects, were linked to higher plasma total cholesterol and LDL levels. In the controls, NHWs and B/AAs exhibited opposite associations between plasma apoE and CSF t-tau. CONCLUSIONS The previously reported lower APOEε4-promoted risk of AD in B/AA subjects may be associated with differences in plasma apoE levels and lipoprotein association. Whether differences in plasma apoE levels between races/ethnicities result from altered APOEε4 expression or turnover, needs further elucidation.
Collapse
Affiliation(s)
- Andreas Giannisis
- Department of Biochemistry and Biophysics, Stockholm University, Svante Arrhenius Väg 16B, 114 18, Stockholm, Sweden
| | - Asma Al-Grety
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
| | - Henrik Carlsson
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
| | | | - William T Hu
- Department of Neurology, Emory University, Atlanta, GA, USA
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, and Institute for Health, Health Care Policy, and Aging Research, New Brunswick, NJ, USA
| | - Kim Kultima
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
| | - Henrietta M Nielsen
- Department of Biochemistry and Biophysics, Stockholm University, Svante Arrhenius Väg 16B, 114 18, Stockholm, Sweden.
| |
Collapse
|
18
|
Hu WT, Nayyar A, Kaluzova M. Charting the Next Road Map for CSF Biomarkers in Alzheimer's Disease and Related Dementias. Neurotherapeutics 2023; 20:955-974. [PMID: 37378862 PMCID: PMC10457281 DOI: 10.1007/s13311-023-01370-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/13/2023] [Indexed: 06/29/2023] Open
Abstract
Clinical prediction of underlying pathologic substrates in people with Alzheimer's disease (AD) dementia or related dementia syndromes (ADRD) has limited accuracy. Etiologic biomarkers - including cerebrospinal fluid (CSF) levels of AD proteins and cerebral amyloid PET imaging - have greatly modernized disease-modifying clinical trials in AD, but their integration into medical practice has been slow. Beyond core CSF AD biomarkers (including beta-amyloid 1-42, total tau, and tau phosphorylated at threonine 181), novel biomarkers have been interrogated in single- and multi-centered studies with uneven rigor. Here, we review early expectations for ideal AD/ADRD biomarkers, assess these goals' future applicability, and propose study designs and performance thresholds for meeting these ideals with a focus on CSF biomarkers. We further propose three new characteristics: equity (oversampling of diverse populations in the design and testing of biomarkers), access (reasonable availability to 80% of people at risk for disease, along with pre- and post-biomarker processes), and reliability (thorough evaluation of pre-analytical and analytical factors influencing measurements and performance). Finally, we urge biomarker scientists to balance the desire and evidence for a biomarker to reflect its namesake function, indulge data- as well as theory-driven associations, re-visit the subset of rigorously measured CSF biomarkers in large datasets (such as Alzheimer's disease neuroimaging initiative), and resist the temptation to favor ease over fail-safe in the development phase. This shift from discovery to application, and from suspended disbelief to cogent ingenuity, should allow the AD/ADRD biomarker field to live up to its billing during the next phase of neurodegenerative disease research.
Collapse
Affiliation(s)
- William T Hu
- Department of Neurology, Rutgers Biomedical and Health Sciences, Rutgers-Robert Wood Johnson Medical School, 125 Paterson Street, Suite 6200, New Brunswick, NJ, 08901, USA.
- Center for Innovation in Health and Aging Research, Institute for Health, Health Care Policy, and Aging Research, Rutgers Biomedical and Health Sciences, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA.
| | - Ashima Nayyar
- Department of Neurology, Rutgers Biomedical and Health Sciences, Rutgers-Robert Wood Johnson Medical School, 125 Paterson Street, Suite 6200, New Brunswick, NJ, 08901, USA
| | - Milota Kaluzova
- Department of Neurology, Rutgers Biomedical and Health Sciences, Rutgers-Robert Wood Johnson Medical School, 125 Paterson Street, Suite 6200, New Brunswick, NJ, 08901, USA
| |
Collapse
|
19
|
Kuhn MK, Fleeman RM, Beidler LM, Snyder AM, Chan DC, Proctor EA. Alzheimer's disease-specific cytokine secretion suppresses neuronal mitochondrial metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.07.536014. [PMID: 37066287 PMCID: PMC10104145 DOI: 10.1101/2023.04.07.536014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Introduction Neuroinflammation and metabolic dysfunction are early alterations in Alzheimer's disease brain that are thought to contribute to disease onset and progression. Glial activation due to protein deposition results in cytokine secretion and shifts in brain metabolism, which have been observed in Alzheimer's disease patients. However, the mechanism by which this immunometabolic feedback loop can injure neurons and cause neurodegeneration remains unclear. Methods We used Luminex XMAP technology to quantify hippocampal cytokine concentrations in the 5xFAD mouse model of Alzheimer's disease at milestone timepoints in disease development. We used partial least squares regression to build cytokine signatures predictive of disease progression, as compared to healthy aging in wild-type littermates. We applied the disease-defining cytokine signature to wild-type primary neuron cultures and measured downstream changes in gene expression using the NanoString nCounter system and mitochondrial function using the Seahorse Extracellular Flux live-cell analyzer. Results We identified a pattern of up-regulated IFNγ, IP-10, and IL-9 as predictive of advanced disease. When healthy neurons were exposed to these cytokines in proportions found in diseased brain, gene expression of mitochondrial electron transport chain complexes, including ATP synthase, was suppressed. In live cells, basal and maximal mitochondrial respiration were impaired following cytokine stimulation. Conclusions An Alzheimer's disease-specific pattern of cytokine secretion reduces expression of mitochondrial electron transport complexes and impairs mitochondrial respiration in healthy neurons. We establish a mechanistic link between disease-specific immune cues and impaired neuronal metabolism, potentially causing neuronal vulnerability and susceptibility to degeneration in Alzheimer's disease.
Collapse
Affiliation(s)
- Madison K. Kuhn
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA
- Center for Neural Engineering, Pennsylvania State University, University Park, PA, USA
| | - Rebecca M. Fleeman
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
| | - Lynne M. Beidler
- Department of Microbiology & Immunology, Penn State College of Medicine, Hershey, PA, USA
| | - Amanda M. Snyder
- Department of Neurology, Penn State College of Medicine, Hershey, PA, USA
| | - Dennis C. Chan
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA
- Center for Neural Engineering, Pennsylvania State University, University Park, PA, USA
| | - Elizabeth A. Proctor
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA
- Department of Engineering Science & Mechanics, Pennsylvania State University, University Park, PA, USA
- Center for Neural Engineering, Pennsylvania State University, University Park, PA, USA
| |
Collapse
|
20
|
Liu Y, Tan Y, Zhang Z, Li H, Yi M, Zhang Z, Hui S, Peng W. Neuroimmune mechanisms underlying Alzheimer's disease: Insights into central and peripheral immune cell crosstalk. Ageing Res Rev 2023; 84:101831. [PMID: 36565960 DOI: 10.1016/j.arr.2022.101831] [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: 08/24/2022] [Revised: 12/15/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
Alzheimer's disease (AD) is a highly life-threatening neurodegenerative disease. Dysregulation of the immune system plays a critical role in promoting AD, which has attracted extensive attention recently. Central and peripheral immune responses are involved in the pathogenesis of AD. Immune changes precede Aβ-associated senile plaque formation and tau-related neurofibrillary tangles, which are the recognised pathological features of AD. Therefore, elucidating immune-related mechanisms underlying the development of AD can help to prevent and treat AD at the source by blocking its progression before the development of pathological changes. To understand the specific pathogenesis of AD, it is important to examine the role of central and peripheral immunity in AD. This review summarises immune-related mechanisms underlying the pathogenesis of AD, focusing on the effect of various central and peripheral immune cells, and describes the possible crosstalk between central and peripheral immunity during the development of AD. This review provides novel insights into the treatment of AD and offers a new direction for immune-related research on AD in the future.
Collapse
Affiliation(s)
- Yuqing Liu
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China; National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.
| | - Yejun Tan
- School of Mathematics, University of Minnesota Twin Cities, Minneapolis, MN, USA.
| | - Zheyu Zhang
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China; National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.
| | - Hongli Li
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China; National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.
| | - Min Yi
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China; National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.
| | - Zhen Zhang
- YangSheng College of Traditional Chinese Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, Guizhou, China.
| | - Shan Hui
- Department of Geratology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410005, China.
| | - Weijun Peng
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China; National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.
| |
Collapse
|
21
|
Felsky D, Santa-Maria I, Cosacak MI, French L, Schneider JA, Bennett DA, De Jager PL, Kizil C, Tosto G. The Caribbean-Hispanic Alzheimer's disease brain transcriptome reveals ancestry-specific disease mechanisms. Neurobiol Dis 2023; 176:105938. [PMID: 36462719 PMCID: PMC10039465 DOI: 10.1016/j.nbd.2022.105938] [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: 08/12/2022] [Revised: 09/21/2022] [Accepted: 11/29/2022] [Indexed: 12/05/2022] Open
Abstract
Identifying ancestry-specific molecular profiles of late-onset Alzheimer's Disease (LOAD) in brain tissue is crucial to understand novel mechanisms and develop effective interventions in non-European, high-risk populations. We performed gene differential expression (DE) and consensus network-based analyses in RNA-sequencing data of postmortem brain tissue from 39 Caribbean Hispanics (CH). To identify ancestry-concordant and -discordant expression profiles, we compared our results to those from two independent non-Hispanic White (NHW) samples (n = 731). In CH, we identified 2802 significant DE genes, including several LOAD known-loci. DE effects were highly concordant across ethnicities, with 373 genes transcriptome-wide significant in all three cohorts. Cross-ancestry meta-analysis found NPNT to be the top DE gene. We replicated over 82% of meta-analyses genome-wide signals in single-nucleus RNA-seq data (including NPNT and LOAD known-genes SORL1, FBXL7, CLU, ABCA7). Increasing representation in genetic studies will allow for deeper understanding of ancestry-specific mechanisms and improving precision treatment options in understudied groups.
Collapse
Affiliation(s)
- Daniel Felsky
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, 250 College St., M5T 1R8, Canada; Department of Psychiatry, University of Toronto, 27 King's College Circle, Toronto, Ontario M5S 1A1, Canada; Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Medical Center, 630 West 168th Street, New York, NY 10032, USA
| | - Ismael Santa-Maria
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, 630 West 168th Street, New York, NY 10032, USA; Department of Pathology and Cell Biology, Columbia University Medical Center, 630 West 168th Street, New York, NY 10032, USA
| | - Mehmet Ilyas Cosacak
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Helmholtz Association, Tatzberg 41, 01307 Dresden, Germany
| | - Leon French
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, 250 College St., M5T 1R8, Canada; Department of Psychiatry, University of Toronto, 27 King's College Circle, Toronto, Ontario M5S 1A1, Canada
| | - Julie A Schneider
- Department of Neurology, Rush University Medical Center, 1653 West Congress Parkway, Chicago, IL 60612, USA; Rush Alzheimer's Disease Center, Rush University Medical Center, 1653 West Congress Parkway, Chicago, IL 60612, USA
| | - David A Bennett
- Department of Neurology, Rush University Medical Center, 1653 West Congress Parkway, Chicago, IL 60612, USA; Rush Alzheimer's Disease Center, Rush University Medical Center, 1653 West Congress Parkway, Chicago, IL 60612, USA
| | - Philip L De Jager
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Medical Center, 630 West 168th Street, New York, NY 10032, USA; The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, 630 West 168th Street, New York, NY 10032, USA
| | - Caghan Kizil
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, 630 West 168th Street, New York, NY 10032, USA; German Center for Neurodegenerative Diseases (DZNE) Dresden, Helmholtz Association, Tatzberg 41, 01307 Dresden, Germany; The Department of Neurology, Columbia University Medical Center, 630 West 168th Street, New York, NY 10032, USA
| | - Giuseppe Tosto
- The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, 630 West 168th Street, New York, NY 10032, USA; The Department of Neurology, Columbia University Medical Center, 630 West 168th Street, New York, NY 10032, USA; Gertrude H. Sergievsky Centre, Columbia University Medical Center, 630 West 168th St., New York, NY 10032, USA.
| |
Collapse
|
22
|
Adhikari UK, Khan R, Mikhael M, Balez R, David MA, Mahns D, Hardy J, Tayebi M. Therapeutic anti-amyloid β antibodies cause neuronal disturbances. Alzheimers Dement 2022. [PMID: 36515320 DOI: 10.1002/alz.12833] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 09/07/2022] [Accepted: 09/19/2022] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Recent published clinical trial safety data showed that 41% of Alzheimer patients experienced amyloid-related imaging abnormalities (ARIA), marks of microhemorrhages and edema in the brain, following administration of Biogen's Aduhelm/aducanumab (amino acids 3-7 of the Aβ peptide). Similarly, Janssen/Pfizer's Bapineuzumab (amino acids 1-5 of the Aβ peptide) and Roche's Gantenerumab (amino acids 2-11/18-27 of the Aβ peptide) also displayed ARIA in clinical trials, including microhemorrhage and focal areas of inflammation or vasogenic edema, respectively. The molecular mechanisms underlying ARIA caused by therapeutic anti-Aβ antibodies remain largely unknown, however, recent reports demonstrated that therapeutic anti-prion antibodies activate neuronal allergenic proteomes following cross-linking cellular prion protein. METHODS Here, we report that treatment of human induced pluripotent stem cells- derived neurons (HSCN) from a non-demented donor, co-cultured with human primary microglia with anti-Aβ1-6, or anti-Aβ17-23 antibodies activate a significant number of allergenic-related proteins as assessed by mass spectrometry. RESULTS Interestingly, a large proportion of the identified proteins included cytokines such as interleukin (IL)-4, IL-12, and IL-13 suggesting a type-1 hypersensitivity response. Following flow cytometry analysis, several proinflammatory cytokines were significantly elevated following anti-Aβ1-6, or anti-Aβ17-23 antibody treatment. DISCUSSION These results justify further and more robust investigation of the molecular mechanisms of ARIA during immunotherapy study trials of AD. HIGHLIGHTS Allergenic-related proteins are often linked with Alzheimer's disease (AD). We investigated the effects of amyloid beta (Aβ) immunotherapy on stem cell derived neurons and primary neuronal cells co-cultured with microglia. Anti-Aβ antibody treatment of neurons or neurons co-cultured with microglia led to activation of a substantial number of allergenic-related genes. These allergenic-related genes are associated with endothelial dysfunction possibly responsible for ARIA.
Collapse
Affiliation(s)
- Utpal Kumar Adhikari
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Rizwan Khan
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Meena Mikhael
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Rachelle Balez
- Illawarra Health and Medical Research Institute, Wollongong, New South Wales, Australia
- School of Chemistry and Molecular Bioscience and Molecular Horizons, University of Wollongong, Wollongong, New South Wales, Australia
| | | | - David Mahns
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - John Hardy
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, UK
| | - Mourad Tayebi
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| |
Collapse
|
23
|
Pak VM, Paul S, Swieboda D, Balthazar MS, Wharton W. Sleep duration and biomarkers of inflammation in African American and white participants with a parental history of Alzheimer's disease. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2022; 8:e12332. [PMID: 36177446 PMCID: PMC9473642 DOI: 10.1002/trc2.12332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 03/24/2022] [Accepted: 06/01/2022] [Indexed: 01/13/2023]
Abstract
Introduction African Americans (AA)s have worse inflammation, worse sleep, and a greater incidence of Alzheimer's disease (AD) compared to whites; however, no studies have examined associations between biomarkers, sleep, and cognition, and differences by race. Methods Seventy-six cognitively normal, middle aged (45-65 years) adults with a parental history of AD were included in this study. Associations between biomarkers (tumor necrosis factor-α [TNF-α], interleukin-10 [IL-10], intercellular adhesion molecule-1 [ICAM-1],, and C-reactive protein [CRP]) and self-reported sleep or cognition measures, were assessed. Results Average sleep duration was significantly lower for AA versus whites (average[SD]) in hours: 6.02(1.18) versus 7.23(0.91), P = .000004). We found a statistically significant association between plasma IL-10 and sleep duration (Spearman's ρ = 0.26, P = .04) and CSF ICAM-1 and sleep quality (Spearman's ρ = 0.30, P = .03). Discussion Longer sleep duration is positively associated with plasma IL-10 levels irrespective of race. Sleep quality was positively associated with CSF ICAM-1 only in African Americans.
Collapse
Affiliation(s)
| | | | | | - Monique S. Balthazar
- School of NursingEmory UniversityAtlantaGeorgiaUSA
- Byrdine F. Lewis College of Nursing and Health ProfessionsGeorgia State UniversityAtlantaGeorgiaUSA
| | - Whitney Wharton
- School of NursingEmory UniversityAtlantaGeorgiaUSA
- Cognitive Neurology DepartmentEmory UniversityAtlantaGeorgiaUSA
| |
Collapse
|
24
|
Saiyasit N, Butlig EAR, Chaney SD, Traylor MK, Hawley NA, Randall RB, Bobinger HV, Frizell CA, Trimm F, Crook ED, Lin M, Hill BD, Keller JL, Nelson AR. Neurovascular Dysfunction in Diverse Communities With Health Disparities-Contributions to Dementia and Alzheimer's Disease. Front Neurosci 2022; 16:915405. [PMID: 35844216 PMCID: PMC9279126 DOI: 10.3389/fnins.2022.915405] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 05/31/2022] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease and related dementias (ADRD) are an expanding worldwide crisis. In the absence of scientific breakthroughs, the global prevalence of ADRD will continue to increase as more people are living longer. Racial or ethnic minority groups have an increased risk and incidence of ADRD and have often been neglected by the scientific research community. There is mounting evidence that vascular insults in the brain can initiate a series of biological events leading to neurodegeneration, cognitive impairment, and ADRD. We are a group of researchers interested in developing and expanding ADRD research, with an emphasis on vascular contributions to dementia, to serve our local diverse community. Toward this goal, the primary objective of this review was to investigate and better understand health disparities in Alabama and the contributions of the social determinants of health to those disparities, particularly in the context of vascular dysfunction in ADRD. Here, we explain the neurovascular dysfunction associated with Alzheimer's disease (AD) as well as the intrinsic and extrinsic risk factors contributing to dysfunction of the neurovascular unit (NVU). Next, we ascertain ethnoregional health disparities of individuals living in Alabama, as well as relevant vascular risk factors linked to AD. We also discuss current pharmaceutical and non-pharmaceutical treatment options for neurovascular dysfunction, mild cognitive impairment (MCI) and AD, including relevant studies and ongoing clinical trials. Overall, individuals in Alabama are adversely affected by social and structural determinants of health leading to health disparities, driven by rurality, ethnic minority status, and lower socioeconomic status (SES). In general, these communities have limited access to healthcare and healthy food and other amenities resulting in decreased opportunities for early diagnosis of and pharmaceutical treatments for ADRD. Although this review is focused on the current state of health disparities of ADRD patients in Alabama, future studies must include diversity of race, ethnicity, and region to best be able to treat all individuals affected by ADRD.
Collapse
Affiliation(s)
- Napatsorn Saiyasit
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Evan-Angelo R. Butlig
- Department of Neurology, Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, Intellectual and Developmental Disabilities Research Center, University of California, Los Angeles, Los Angeles, CA, United States
| | - Samantha D. Chaney
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Miranda K. Traylor
- Department of Health, Kinesiology, and Sport, University of South Alabama, Mobile, AL, United States
| | - Nanako A. Hawley
- Department of Psychology, University of South Alabama, Mobile, AL, United States
| | - Ryleigh B. Randall
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Hanna V. Bobinger
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Carl A. Frizell
- Department of Physician Assistant Studies, University of South Alabama, Mobile, AL, United States
| | - Franklin Trimm
- College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Errol D. Crook
- Department of Internal Medicine, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Mike Lin
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Benjamin D. Hill
- Department of Psychology, University of South Alabama, Mobile, AL, United States
| | - Joshua L. Keller
- Department of Health, Kinesiology, and Sport, University of South Alabama, Mobile, AL, United States
| | - Amy R. Nelson
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| |
Collapse
|
25
|
Relationship between parental history of dementia, motor-cognitive and executive function performance in African American women. J Neurol Sci 2022; 439:120305. [DOI: 10.1016/j.jns.2022.120305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 04/19/2022] [Accepted: 05/28/2022] [Indexed: 11/23/2022]
|
26
|
Brijnath B, Croy S, Sabates J, Thodis A, Ellis S, de Crespigny F, Moxey A, Day R, Dobson A, Elliott C, Etherington C, Geronimo MA, Hlis D, Lampit A, Low L, Straiton N, Temple J. Including ethnic minorities in dementia research: Recommendations from a scoping review. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2022; 8:e12222. [PMID: 35505899 PMCID: PMC9053375 DOI: 10.1002/trc2.12222] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 08/18/2021] [Accepted: 10/27/2021] [Indexed: 11/30/2022]
Abstract
Introduction Ethnicity influences dementia etiology, prognosis, and treatment, while culture shapes help-seeking and care. Despite increasing population diversity in high-income settlement countries, ethnic minorities remain underrepresented in dementia research. We investigated approaches to enhance the recruitment, and consistent collection and analysis of variables relevant to, ethnic minorities in dementia studies to make recommendations for consistent practice in dementia research. Methods We did a scoping review, searching Embase, PsycINFO, Medline, CENTRAL, and CINAHL between January 1, 2010 and January 7, 2020. Dementia clinical and cohort studies that actively recruited ethnic minorities in high-income countries were included. A steering group of experts developed criteria through which high-quality studies were identified. Results Sixty-six articles were retrieved (51 observational; 15 experimental). Use of interpreters and translators (n = 17) was the most common method to facilitate participant recruitment. Race and ethnicity (n = 59) were the most common variables collected, followed by information on native language (n = 14), country of birth (n = 9), and length of time in country of settlement (n = 8). Thirty-three studies translated or used a culturally validated instrument. Twenty-three articles conducted subgroup analyses based on ethnicity. Six high-quality studies facilitated inclusion through community engagement, collected information on multiple aspects of ethnic diversity, and adjusted/substratified to analyze the impact of ethnicity on dementia. Discussion We make recommendations for consistent recruitment, collection, and reporting of variables relating to ethnic and cultural diversity in dementia research.
Collapse
Affiliation(s)
- Bianca Brijnath
- National Ageing Research InstituteParkvilleVictoriaAustralia
- School of Social SciencesUniversity of Western AustraliaWestern AustraliaPerchAustralia
| | - Samantha Croy
- Centre for Population GenomicsMurdoch Children's Research InstituteMelbourneVictoriaAustralia
| | - Julieta Sabates
- Academic Unit for Psychiatry of Old AgeUniversity of MelbourneParkvilleVictoriaAustralia
| | - Antonia Thodis
- National Ageing Research InstituteParkvilleVictoriaAustralia
| | | | - Fleur de Crespigny
- Australian Institute of Health and WelfareCanberraAustralian Capital TerritoryAustralia
| | - Annette Moxey
- Dementia Australia Research FoundationGriffithAustralian Capital TerritoryAustralia
| | - Robert Day
- Australian Government Department of HealthCanberraAustralian Capital TerritoryAustralia
| | - Annette Dobson
- School of Public HealthUniversity of QueenslandHerstonQueenslandAustralia
| | | | - Cathy Etherington
- Australian Bureau of StatisticsBelconnenAustralian Capital TerritoryAustralia
| | - Mary Ann Geronimo
- Federation of Ethnic Community Councils of AustraliaDeakinAustralian Capital TerritoryAustralia
| | | | - Amit Lampit
- Academic Unit for Psychiatry of Old AgeUniversity of MelbourneParkvilleVictoriaAustralia
| | - Lee‐Fay Low
- Sydney School of Health SciencesUniversity of SydneyCamperdownNew South WalesAustralia
| | | | - Jeromey Temple
- School of Population and Global HealthUniversity of MelbourneParkvilleVictoriaAustralia
| |
Collapse
|
27
|
Khan MJ, Chung NA, Hansen S, Dumitrescu L, Hohman TJ, Kamboh MI, Lopez OL, Robinson RAS. Targeted Lipidomics To Measure Phospholipids and Sphingomyelins in Plasma: A Pilot Study To Understand the Impact of Race/Ethnicity in Alzheimer's Disease. Anal Chem 2022; 94:4165-4174. [PMID: 35235294 PMCID: PMC9126486 DOI: 10.1021/acs.analchem.1c03821] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The number of people suffering from Alzheimer's disease (AD) is increasing rapidly every year. One aspect of AD that is often overlooked is the disproportionate incidence of AD among African American/Black populations. With the recent development of novel assays for lipidomics analysis in recent times, there has been a drastic increase in the number of studies focusing on changes of lipids in AD. However, very few of these studies have focused on or even included samples from African American/Black individuals samples. In this study, we aimed to determine if the lipidome in AD is universal across non-Hispanic White and African American/Black individuals. To accomplish this, a targeted mass spectrometry lipidomics analysis was performed on plasma samples (N = 113) obtained from cognitively normal (CN, N = 54) and AD (N = 59) individuals from African American/Black (N = 56) and non-Hispanic White (N = 57) backgrounds. Five lipids (PS 18:0_18:0, PS 18:0_20:0, PC 16:0_22:6, PC 18:0_22:6, and PS 18:1_22:6) were altered between AD and CN sample groups (p value < 0.05). Upon racial stratification, there were notable differences in lipids that were unique to African American/Black or non-Hispanic White individuals. PS 20:0_20:1 was reduced in AD in samples from non-Hispanic White but not African American/Black adults. We also tested whether race/ethnicity significantly modified the association between lipids and AD status by including a race × diagnosis interaction term in a linear regression model. PS 20:0_20:1 showed a significant interaction (p = 0.004). The discovery of lipid changes in AD in this study suggests that identifying relevant lipid biomarkers for diagnosis will require diversity in sample cohorts.
Collapse
Affiliation(s)
- Mostafa J Khan
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Nadjali A Chung
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Shania Hansen
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, Tennessee 37212, United States
| | - Logan Dumitrescu
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, Tennessee 37212, United States.,Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States.,Vanderbilt Brain Institute, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States.,Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Timothy J Hohman
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, Tennessee 37212, United States
| | - M Ilyas Kamboh
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United States.,Department of Human Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United States.,Department of Epidemiology, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United States
| | - Oscar L Lopez
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United States.,Department of Neurology, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United States
| | - Renã A S Robinson
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States.,Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, Tennessee 37212, United States.,Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States.,Vanderbilt Brain Institute, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States.,Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37232, United States
| |
Collapse
|
28
|
Fazeli PL, Woods SP, Lambert CC, Li W, Hopkins CN, Vance DE. Differential Associations Between BDNF and Memory Across Older Black and White Adults With HIV Disease. J Acquir Immune Defic Syndr 2022; 89:129-135. [PMID: 34629411 PMCID: PMC8752478 DOI: 10.1097/qai.0000000000002831] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/27/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND Brain-derived neurotrophic factor (BDNF) shows consistent associations with memory across many clinical populations, including dementia. Less is understood about the association between BDNF and memory functioning in people living with HIV (PWH). METHODS A sample of 173 adults aged 50+ (n = 100 HIV+ and n = 73 HIV seronegative) completed a comprehensive neurobehavioral assessment and blood draw. Linear regressions predicting memory domains (learning, delayed recall, and recognition) were conducted including race (White vs. Black/African American), HIV status, BDNF, and their interactions. RESULTS For learning and delayed recall, significant (P < 0.05) main effects for race and interactions for BDNF x race and HIV status x race were found, whereas for recognition, only a BDNF x race interaction emerged. In adjusted models, BDNF x race interactions remained for learning and delayed recall. To determine effect size, correlations were conducted between BDNF and memory domains stratified by HIV serostatus and race, and small-medium associations between BDNF and learning and delayed recall (rho = 0.29, P < 0.01; rho = 0.22, P = 0.045), but no recognition (rho = 0.12, P = 0.29) were found among Black/African American PWH. BDNF was not significantly associated with memory domains in White PWH or either HIV- sample. Follow-up analyses showed BDNF-memory specificity, such that race X BDNF interactions did not emerge for other cognitive domains. CONCLUSIONS While limited by cross-sectional design among a small sample, particularly of White individuals, results indicate that BDNF may serve as a promising biomarker reflecting memory functioning in PWH, particularly Black/African Americans. Further work is needed to replicate findings and determine mechanisms for racial differences in BDNF associations with memory.
Collapse
Affiliation(s)
- Pariya L. Fazeli
- School of Nursing, University of Alabama at Birmingham, Birmingham, AL
| | | | | | - Wei Li
- School of Health Professions, University of Alabama at Birmingham, Birmingham, AL
| | - Cierra N. Hopkins
- School of Nursing, University of Alabama at Birmingham, Birmingham, AL
| | - David E. Vance
- School of Nursing, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
29
|
Dabravolski SA, Nikiforov NG, Zhuravlev AD, Orekhov NA, Grechko AV, Orekhov AN. Role of the mtDNA Mutations and Mitophagy in Inflammaging. Int J Mol Sci 2022; 23:ijms23031323. [PMID: 35163247 PMCID: PMC8836173 DOI: 10.3390/ijms23031323] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/12/2022] [Accepted: 01/17/2022] [Indexed: 12/14/2022] Open
Abstract
Ageing is an unavoidable multi-factorial process, characterised by a gradual decrease in physiological functionality and increasing vulnerability of the organism to environmental factors and pathogens, ending, eventually, in death. One of the most elaborated ageing theories implies a direct connection between ROS-mediated mtDNA damage and mutations. In this review, we focus on the role of mitochondrial metabolism, mitochondria generated ROS, mitochondrial dynamics and mitophagy in normal ageing and pathological conditions, such as inflammation. Also, a chronic form of inflammation, which could change the long-term status of the immune system in an age-dependent way, is discussed. Finally, the role of inflammaging in the most common neurodegenerative diseases, such as Alzheimer’s and Parkinson’s, is also discussed.
Collapse
Affiliation(s)
- Siarhei A. Dabravolski
- Department of Clinical Diagnostics, Vitebsk State Academy of Veterinary Medicine [UO VGAVM], 7/11 Dovatora Str., 210026 Vitebsk, Belarus
- Correspondence:
| | - Nikita G. Nikiforov
- AP Avtsyn Research Institute of Human Morphology, 3 Tsyurupa Street, 117418 Moscow, Russia; (N.G.N.); (A.D.Z.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilova Street, 119334 Moscow, Russia
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia
| | - Alexander D. Zhuravlev
- AP Avtsyn Research Institute of Human Morphology, 3 Tsyurupa Street, 117418 Moscow, Russia; (N.G.N.); (A.D.Z.)
| | - Nikolay A. Orekhov
- Institute for Atherosclerosis Research, Osennyaya Street 4-1-207, 121609 Moscow, Russia; (N.A.O.); (A.N.O.)
| | - Andrey V. Grechko
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 14-3 Solyanka Street, 109240 Moscow, Russia;
| | - Alexander N. Orekhov
- Institute for Atherosclerosis Research, Osennyaya Street 4-1-207, 121609 Moscow, Russia; (N.A.O.); (A.N.O.)
| |
Collapse
|
30
|
Noren Hooten N, Pacheco NL, Smith JT, Evans MK. The accelerated aging phenotype: The role of race and social determinants of health on aging. Ageing Res Rev 2022; 73:101536. [PMID: 34883202 PMCID: PMC10862389 DOI: 10.1016/j.arr.2021.101536] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 11/12/2021] [Accepted: 12/03/2021] [Indexed: 02/06/2023]
Abstract
The pursuit to discover the fundamental biology and mechanisms of aging within the context of the physical and social environment is critical to designing interventions to prevent and treat its complex phenotypes. Aging research is critically linked to understanding health disparities because these inequities shape minority aging, which may proceed on a different trajectory than the overall population. Health disparities are characteristically seen in commonly occurring age-associated diseases such as cardiovascular and cerebrovascular disease as well as diabetes mellitus and cancer. The early appearance and increased severity of age-associated disease among African American and low socioeconomic status (SES) individuals suggests that the factors contributing to the emergence of health disparities may also induce a phenotype of 'premature aging' or 'accelerated aging' or 'weathering'. In marginalized and low SES populations with high rates of early onset age-associated disease the interaction of biologic, psychosocial, socioeconomic and environmental factors may result in a phenotype of accelerated aging biologically similar to premature aging syndromes with increased susceptibility to oxidative stress, premature accumulation of oxidative DNA damage, defects in DNA repair and higher levels of biomarkers of oxidative stress and inflammation. Health disparities, therefore, may be the end product of this complex interaction in populations at high risk. This review will examine the factors that drive both health disparities and the accelerated aging phenotype that ultimately contributes to premature mortality.
Collapse
Affiliation(s)
- Nicole Noren Hooten
- Laboratory of Epidemiology and Population Science, National Institute on Aging, National Institutes of Health, 251 Bayview Boulevard, Baltimore, MD 21224, USA
| | - Natasha L Pacheco
- Laboratory of Epidemiology and Population Science, National Institute on Aging, National Institutes of Health, 251 Bayview Boulevard, Baltimore, MD 21224, USA
| | - Jessica T Smith
- Laboratory of Epidemiology and Population Science, National Institute on Aging, National Institutes of Health, 251 Bayview Boulevard, Baltimore, MD 21224, USA
| | - Michele K Evans
- Laboratory of Epidemiology and Population Science, National Institute on Aging, National Institutes of Health, 251 Bayview Boulevard, Baltimore, MD 21224, USA.
| |
Collapse
|
31
|
Cerebrospinal fluid biomarkers in Parkinson's disease with freezing of gait: an exploratory analysis. NPJ Parkinsons Dis 2021; 7:105. [PMID: 34845234 PMCID: PMC8629994 DOI: 10.1038/s41531-021-00247-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 10/27/2021] [Indexed: 01/06/2023] Open
Abstract
We explore the association between three Alzheimer’s disease-related and ten inflammation-related CSF markers and freezing of gait (FOG) in patients with Parkinson’s disease (PD). The study population includes PD patients with FOG (PD-FOG, N = 12), without FOG (PD-NoFOG, N = 19), and healthy controls (HC, N = 12). Age and PD duration are not significantly different between groups. After adjusting for covariates and multiple comparisons, the anti-inflammatory marker, fractalkine, is significantly decreased in the PD groups compared to HC (P = 0.002), and further decreased in PD-FOG compared to PD-NoFOG (P = 0.007). The Alzheimer’s disease-related protein, Aβ42, is increased in PD-FOG compared to PD-NoFOG and HC (P = 0.001). Group differences obtained in individual biomarker analyses are confirmed with multivariate discriminant partial least squares regression (P < 0.001). High levels of Aβ42 in PD-FOG patients supports an increase over time from early to advanced state. Low levels of fractalkine might suggest anti-inflammatory effect. These findings warrant replication.
Collapse
|
32
|
Ghosh P, Singh R, Ganeshpurkar A, Pokle AV, Singh RB, Singh SK, Kumar A. Cellular and molecular influencers of neuroinflammation in Alzheimer's disease: Recent concepts & roles. Neurochem Int 2021; 151:105212. [PMID: 34656693 DOI: 10.1016/j.neuint.2021.105212] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 09/22/2021] [Accepted: 10/10/2021] [Indexed: 01/21/2023]
Abstract
Alzheimer's disease (AD), an extremely common neurodegenerative disorder of the older generation, is one of the leading causes of death globally. Besides the conventional hallmarks i.e. Amyloid-β (Aβ) plaques and neurofibrillary tangles (NFTs), neuroinflammation also serves as a major contributing factor in the pathogenesis of AD. There are mounting evidences to support the fundamental role of cellular (microglia, astrocytes, mast cells, and T-cells) and molecular (cytokines, chemokines, caspases, and complement proteins) influencers of neuroinflammation in producing/promoting neurodegeneration and dementia in AD. Genome-wide association studies (GWAS) have revealed the involvement of various single nucleotide polymorphisms (SNPs) of genes related to neuroinflammation with the risk of developing AD. Modulating the release of the neuroinflammatory molecules and targeting their relevant mechanisms may have beneficial effects on the onset, progress and severity of the disease. Here, we review the distinct role of various mediators and modulators of neuroinflammation that impact the pathogenesis and progression of AD as well as incite further research efforts for the treatment of AD through a neuroinflammatory approach.
Collapse
Affiliation(s)
- Powsali Ghosh
- Pharmaceutical Chemistry Research Laboratory 1, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Ravi Singh
- Pharmaceutical Chemistry Research Laboratory 1, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Ankit Ganeshpurkar
- Pharmaceutical Chemistry Research Laboratory 1, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Ankit Vyankatrao Pokle
- Pharmaceutical Chemistry Research Laboratory 1, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Ravi Bhushan Singh
- Institute of Pharmacy Harischandra PG College, Bawanbigha, Varanasi, India
| | - Sushil Kumar Singh
- Pharmaceutical Chemistry Research Laboratory 1, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Ashok Kumar
- Pharmaceutical Chemistry Research Laboratory 1, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India.
| |
Collapse
|
33
|
Hu WT, Ozturk T, Kollhoff A, Wharton W, Christina Howell J. Higher CSF sTNFR1-related proteins associate with better prognosis in very early Alzheimer's disease. Nat Commun 2021; 12:4001. [PMID: 34183654 PMCID: PMC8238986 DOI: 10.1038/s41467-021-24220-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 06/08/2021] [Indexed: 02/06/2023] Open
Abstract
Neuroinflammation is associated with Alzheimer's disease, but the application of cerebrospinal fluid measures of inflammatory proteins may be limited by overlapping pathways and relationships between them. In this work, we measure 15 cerebrospinal proteins related to microglial and T-cell functions, and show them to reproducibly form functionally-related groups within and across diagnostic categories in 382 participants from the Alzheimer's Disease Neuro-imaging Initiative as well participants from two independent cohorts. We further show higher levels of proteins related to soluble tumor necrosis factor receptor 1 are associated with reduced risk of conversion to dementia in the multi-centered (p = 0.027) and independent (p = 0.038) cohorts of people with mild cognitive impairment due to predicted Alzheimer's disease, while higher soluble TREM2 levels associated with slower decline in the dementia stage of Alzheimer's disease. These inflammatory proteins thus provide prognostic information independent of established Alzheimer's markers.
Collapse
Affiliation(s)
- William T Hu
- Department of Neurology and Center for Neurodegenerative Diseases, School of Medicine, Emory University, Atlanta, GA, USA.
- Rutgers Robert Wood Johnson Medical School and Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers Biomedical and Health Sciences, New Brunswick, NJ, USA.
| | - Tugba Ozturk
- Department of Neurology and Center for Neurodegenerative Diseases, School of Medicine, Emory University, Atlanta, GA, USA
| | - Alexander Kollhoff
- Department of Neurology and Center for Neurodegenerative Diseases, School of Medicine, Emory University, Atlanta, GA, USA
| | - Whitney Wharton
- Nell Hodgson School of Nursing, Emory University, Atlanta, GA, USA
| | - J Christina Howell
- Department of Neurology and Center for Neurodegenerative Diseases, School of Medicine, Emory University, Atlanta, GA, USA
| |
Collapse
|
34
|
Llibre-Guerra JJ, Li Y, Allen IE, Llibre-Guerra JC, Rodríguez Salgado AM, Peñalver AI, Sanchez AA, Yokoyama JS, Grinberg L, Valcour V, Miller BL, Llibre-Rodríguez JJ. Race, genetic admixture and cognitive performance in the Cuban population. J Gerontol A Biol Sci Med Sci 2021; 77:331-338. [PMID: 33649769 DOI: 10.1093/gerona/glab063] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Population aging will lead to a dramatic increase in dementia prevalence, which will disproportionally affect racial minorities. The presence of racial differences in dementia prevalence has been widely reported in United States, but there are no relevant studies on this topic in low-middle income countries (LMIC). METHODS In a cross-sectional survey, 2,944 older Cubans were recruited at a community-based level aimed to identify the effects of self-identified race and genetic admixture on cognitive performance. Dementia diagnosis was established using 10/66 Dementia and DSM-IV criteria. APOE-ε4 genotype was determined in 2,511 (85%) and genetic admixture was completed for all dementia cases and in a randomly selected sample of cognitive healthy participants (218 dementia cases and 367 participants without dementia). RESULTS The overall prevalence of dementia was 8.7%, without large or statistically significant differences on dementia prevalence (p=0.12) by self-identified race. Mean cognitive scores were similar across racial groups (p=0.46). After controlling for age, sex and education, greater proportion of African ancestry was not associated with cognitive performance (p=0.17). CONCLUSIONS We found no evidence of an independent effect of self-identified race and/or population ancestry on dementia prevalence or cognitive performance. This suggests that observed differences in dementia prevalence among diverse populations may be driven primarily by social determinants of health.
Collapse
Affiliation(s)
- Jorge J Llibre-Guerra
- Department of Neurology. Washington University in St Louis, St Louis, MO, USA.,Global Brain Health Institute, University of California San Francisco, San Francisco, CA, USA
| | - Yan Li
- Department of Neurology. Washington University in St Louis, St Louis, MO, USA
| | - Isabel Elaine Allen
- Department of Epidemiology & Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | | | - Ana M Rodríguez Salgado
- Global Brain Health Institute, University of California San Francisco, San Francisco, CA, USA
| | - Ana Ibis Peñalver
- Department of Neurology. National Institute of Neurology and Neurosurgery, La Havana, Cuba
| | | | - Jennifer S Yokoyama
- Global Brain Health Institute, University of California San Francisco, San Francisco, CA, USA.,Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Lea Grinberg
- Global Brain Health Institute, University of California San Francisco, San Francisco, CA, USA.,Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Victor Valcour
- Global Brain Health Institute, University of California San Francisco, San Francisco, CA, USA.,Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Bruce L Miller
- Global Brain Health Institute, University of California San Francisco, San Francisco, CA, USA.,Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | | |
Collapse
|
35
|
Trotti LM, Bliwise DL, Keating GL, Rye DB, Hu WT. Cerebrospinal Fluid Hypocretin and Nightmares in Dementia Syndromes. Dement Geriatr Cogn Dis Extra 2021; 11:19-25. [PMID: 33790936 PMCID: PMC7989783 DOI: 10.1159/000509585] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 06/19/2020] [Indexed: 01/08/2023] Open
Abstract
Background/Aims Hypocretin promotes wakefulness and modulates REM sleep. Alterations in the hypocretin system are increasingly implicated in dementia. We evaluated relationships among hypocretin, dementia biomarkers, and sleep symptoms in elderly participants, most of whom had dementia. Methods One-hundred twenty-six adults (mean age 66.2 ± 8.4 years) were recruited from the Emory Cognitive Clinic. Diagnoses were Alzheimer disease (AD; n = 60), frontotemporal dementia (FTD; n = 21), and dementia with Lewy bodies (DLB; n = 20). We also included cognitively normal controls (n = 25). Participants and/or caregivers completed sleep questionnaires and lumbar puncture was performed for cerebrospinal fluid (CSF) assessments. Results Except for sleepiness (worst in DLB) and nocturia (worse in DLB and FTD) sleep symptoms did not differ by diagnosis. CSF hypocretin concentrations were available for 87 participants and normal in 70, intermediate in 16, and low in 1. Hypocretin levels did not differ by diagnosis. Hypocretin levels correlated with CSF total τ levels only in men (r = 0.34; p = 0.02). Lower hypocretin levels were related to frequency of nightmares (203.9 ± 29.8 pg/mL in those with frequent nightmares vs. 240.4 ± 46.1 pg/mL in those without; p = 0.05) and vivid dreams (209.1 ± 28.3 vs. 239.5 ± 47.8 pg/mL; p = 0.014). Cholinesterase inhibitor use was not associated with nightmares or vivid dreaming. Conclusion Hypocretin levels did not distinguish between dementia syndromes. Disturbing dreams in dementia patients may be related to lower hypocretin concentrations in CSF.
Collapse
Affiliation(s)
- Lynn Marie Trotti
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, USA.,Emory Sleep Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Donald L Bliwise
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, USA.,Emory Sleep Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Glenda L Keating
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - David B Rye
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, USA.,Emory Sleep Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - William T Hu
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
36
|
Perales-Puchalt J, Gauthreaux K, Shaw A, McGee JL, Teylan MA, Chan KCG, Rascovsky K, Kukull WA, Vidoni ED. Risk of mild cognitive impairment among older adults in the United States by ethnoracial group. Int Psychogeriatr 2021; 33:51-62. [PMID: 31948505 PMCID: PMC7365740 DOI: 10.1017/s1041610219002175] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
OBJECTIVES To compare the risk of mild cognitive impairment (MCI) among a wide range of ethnoracial groups in the US. DESIGN Non-probabilistic longitudinal clinical research. SETTING Participants enrolling into the National Alzheimer's Coordinating Center Unified Data Set recruited via multiple approaches including clinician referral, self-referral by patients or family members, or active recruitment through community organizations. PARTICIPANTS Cognitively normal individuals 55 and older at the initial visit, who reported race and ethnicity information, with at least two visits between September 2005 and November 2018. MEASUREMENTS Ethnoracial information was self-reported and grouped into non-Latino Whites, Asian Americans, Native Americans, African Americans (AAs), and individuals simultaneously identifying as AAs and another minority race (AA+), as well as Latinos of Caribbean, Mexican, and Central/South American origin. MCI was evaluated clinically following standard criteria. Four competing risk analysis models were used to calculate MCI risk adjusting for risk of death, including an unadjusted model, and models adjusting for non-modifiable and modifiable risk factors. RESULTS After controlling for sex and age at initial visit, subhazard ratios of MCI were statistically higher than non-Latino Whites among Native Americans (1.73), Caribbean Latinos (1.80), and Central/South American Latinos (1.55). Subhazard ratios were higher among AA+ compared to non-Latino Whites only in the model controlling for all risk factors (1.40). CONCLUSION Compared to non-Latino Whites, MCI risk was higher among Caribbean and South/Central American Latinos as well as Native Americans and AA+. The factors explaining the differential MCI risk among ethnoracial groups are not clear and warrant future research.
Collapse
Affiliation(s)
| | - Kathryn Gauthreaux
- Department of Epidemiology, National Alzheimer's Coordinating Center, University of Washington, Seattle, WA98195, USA
| | - Ashley Shaw
- Alzheimer's Disease Center, University of Kansas, MS6002, Fairway, KS66205, USA
| | - Jerrihlyn L McGee
- School of Nursing, University of Kansas, MS 4043, Kansas City, KS66160, USA
| | - Merilee A Teylan
- Department of Epidemiology, National Alzheimer's Coordinating Center, University of Washington, Seattle, WA98195, USA
| | - Kwun C G Chan
- Department of Biostatistics, National Alzheimer's Coordinating Center, University of Washington, Seattle, WA98195, USA
| | - Katya Rascovsky
- Department of Neurology, Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Walter A Kukull
- Department of Epidemiology, National Alzheimer's Coordinating Center, University of Washington, Seattle, WA98195, USA
| | - Eric D Vidoni
- Alzheimer's Disease Center, University of Kansas, MS6002, Fairway, KS66205, USA
| |
Collapse
|
37
|
Khan MJ, Desaire H, Lopez OL, Kamboh MI, Robinson RA. Why Inclusion Matters for Alzheimer's Disease Biomarker Discovery in Plasma. J Alzheimers Dis 2021; 79:1327-1344. [PMID: 33427747 PMCID: PMC9126484 DOI: 10.3233/jad-201318] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
BACKGROUND African American/Black adults have a disproportionate incidence of Alzheimer's disease (AD) and are underrepresented in biomarker discovery efforts. OBJECTIVE This study aimed to identify potential diagnostic biomarkers for AD using a combination of proteomics and machine learning approaches in a cohort that included African American/Black adults. METHODS We conducted a discovery-based plasma proteomics study on plasma samples (N = 113) obtained from clinically diagnosed AD and cognitively normal adults that were self-reported African American/Black or non-Hispanic White. Sets of differentially-expressed proteins were then classified using a support vector machine (SVM) to identify biomarker candidates. RESULTS In total, 740 proteins were identified of which, 25 differentially-expressed proteins in AD came from comparisons within a single racial and ethnic background group. Six proteins were differentially-expressed in AD regardless of racial and ethnic background. Supervised classification by SVM yielded an area under the curve (AUC) of 0.91 and accuracy of 86%for differentiating AD in samples from non-Hispanic White adults when trained with differentially-expressed proteins unique to that group. However, the same model yielded an AUC of 0.49 and accuracy of 47%for differentiating AD in samples from African American/Black adults. Other covariates such as age, APOE4 status, sex, and years of education were found to improve the model mostly in the samples from non-Hispanic White adults for classifying AD. CONCLUSION These results demonstrate the importance of study designs in AD biomarker discovery, which must include diverse racial and ethnic groups such as African American/Black adults to develop effective biomarkers.
Collapse
Affiliation(s)
- Mostafa J. Khan
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
| | - Heather Desaire
- Department of Chemistry, University of Kansas, Lawrence, KS, USA
| | - Oscar L. Lopez
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - M. Ilyas Kamboh
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Renã A.S. Robinson
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
38
|
Wharton W, Jeong L, Ni L, Bay AA, Shin RJ, McCullough LE, Silverstein H, Hart AR, Swieboda D, Hu W, Hackney ME. A Pilot randomized clinical trial of adapted tango to improve cognition and psychosocial function in African American women with family history of Alzheimer's disease (ACT trial). CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2021; 2:100018. [PMID: 36324714 PMCID: PMC9616328 DOI: 10.1016/j.cccb.2021.100018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/30/2021] [Accepted: 06/17/2021] [Indexed: 11/24/2022]
Abstract
the adapted tango intervention was very well received as demonstrated by minimal participant attrition, satisfaction questionnaires that indicated high satisfaction, and anecdotal reports. Adapted tango intervention may be helpful in controlling or reducing markers of inflammation in AA women with a parental history of AD. Participants in tango demonstrated improvements in whole-body spatial cognition and short-term and working memory, and reduced deterioration of executive function. Although our tango group did not show large positive effect in cumulative caregiver burden post intervention, the large positive effect in role Captivity, caregiver confidence, and deprivation of intimate exchange displays some of the positive effects of this dance intervention for the caregivers.
Alzheimer's disease (AD) is a devastating, progressive neurodegenerative disease resulting in memory loss and a severe reduction in the ability to perform activities of daily living. Ethnicity-related genetic factors promoting the development of dementias among African Americans (AA) and increased risk among women for developing AD indicates that AA women with a parental history of AD are at great risk for developing AD. This phase I study assessed the impact of a 12 week, 20-lesson adapted Argentine Tango intervention (n = 24) to a no-contact control group (n = 10) on measures of plasma inflammatory markers, cognition, and motor and psychosocial performance in middle-aged AA woman at increased risk for AD by virtue of parental history. Some woman (n = 16) were also caregivers; thus, the impact of the intervention on caregiving burden was examined in this subset. Preliminary analysis of efficacy was conducted with significance tests on biomarkers and key measures of cognition, including visuospatial and executive function, balance, and strength. After 12 weeks, Tango participants had significantly decreased inflammatory cytokine, including reductions in IL-7 (p = 0.003), IFN-γ (p = 0.011), TNFα (p = 0.011), and MCP-1 (p = 0.042) compared to controls. Large effects were noted for the Tango group on tests of executive functioning (d = 0.89), and inhibition (p = 0.031). Participants in Tango improved in dynamic and static balance (p = 0.018) and functional lower body strength (p = 0.023). Secondary assessment revealed trends favoring the intervention group were noted in spatial cognition and executive function. Moderate effects were noted in caregiving burden measures among the subset of caregivers. These data demonstrate substantial reductions in inflammatory biomarkers along with cognitive and motor improvements through a non-pharmacologic, affordable intervention among a small, well-characterized cohort of AA women with a parental history of AD.
Collapse
Affiliation(s)
| | - Leanne Jeong
- Emory University College of Arts and Sciences, 550 Asbury Circle, Atlanta, GA, 30322, USA
| | - Liang Ni
- Division of Geriatrics and Gerontology, Department of Medicine, Emory School of Medicine, 1841 Clifton Rd NE, Atlanta, GA, 30307, USA
| | - Allison A. Bay
- Division of Geriatrics and Gerontology, Department of Medicine, Emory School of Medicine, 1841 Clifton Rd NE, Atlanta, GA, 30307, USA
| | - Ryan J. Shin
- Emory University College of Arts and Sciences, 550 Asbury Circle, Atlanta, GA, 30322, USA
| | - Lauren E. McCullough
- Emory University Rollins School of Public Health, 1518 Clifton Rd., Atlanta, GA, 30329, USA
| | - Hayley Silverstein
- Division of Geriatrics and Gerontology, Department of Medicine, Emory School of Medicine, 1841 Clifton Rd NE, Atlanta, GA, 30307, USA
| | | | | | - William Hu
- Division of Cognitive Neurology, Rutgers Robert Wood Johnson Medical School, 125 Paterson Street, New Brunswick, NJ 08901, USA
| | - Madeleine E. Hackney
- Emory University School of Nursing, Atlanta, GA, 30307, USA
- Division of Geriatrics and Gerontology, Department of Medicine, Emory School of Medicine, 1841 Clifton Rd NE, Atlanta, GA, 30307, USA
- Atlanta VA Center for Visual and Neurocognitive Rehabilitation, 1670 Clairmont Rd., Decatur, GA, 30033, USA
- Department of Rehabilitation Medicine, Emory School of Medicine, 1648 Pierce Dr. NE, Atlanta, GA, 30307, USA
- Birmingham/Atlanta VA Geriatric Research Clinical and Education Center
- Corresponding author at: Emory University School of Medicine, Department of Medicine, Division of Geriatrics and Gerontology, Research Health Scientist, Atlanta VA Rehabilitation R&D Center for Visual and Neurocognitive Rehabilitation, 1841 Clifton Rd. NE, #553, Atlanta, GA 30329, USA.
| |
Collapse
|
39
|
Stepler KE, Mahoney ER, Kofler J, Hohman TJ, Lopez OL, Robinson RAS. Inclusion of African American/Black adults in a pilot brain proteomics study of Alzheimer's disease. Neurobiol Dis 2020; 146:105129. [PMID: 33049317 PMCID: PMC7990397 DOI: 10.1016/j.nbd.2020.105129] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/02/2020] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease (AD) disproportionately affects certain racial and ethnic subgroups, such as African American/Black and Hispanic adults. Genetic, comorbid, and socioeconomic risk factors contribute to this disparity; however, the molecular contributions have been largely unexplored. Herein, we conducted a pilot proteomics study of postmortem brains from African American/Black and non-Hispanic White adults neuropathologically diagnosed with AD compared to closely-matched cognitively normal individuals. Examination of hippocampus, inferior parietal lobule, and globus pallidus regions using quantitative proteomics resulted in 568 differentially-expressed proteins in AD. These proteins were consistent with the literature and included glial fibrillary acidic protein, peroxiredoxin-1, and annexin A5. In addition, 351 novel proteins in AD were identified, which could partially be due to cohort diversity. From linear regression analyses, we identified 185 proteins with significant race x diagnosis interactions across various brain regions. These differences generally were reflective of differential expression of proteins in AD that occurred in only a single racial/ethnic group. Overall, this pilot study suggests that disease understanding can be furthered by including diversity in racial/ethnic groups; however, this must be done on a larger scale.
Collapse
Affiliation(s)
- Kaitlyn E Stepler
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, United States of America
| | - Emily R Mahoney
- Vanderbilt Memory & Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN 37212, United States of America; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37232, United States of America
| | - Julia Kofler
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, United States of America
| | - Timothy J Hohman
- Vanderbilt Memory & Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN 37212, United States of America; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37232, United States of America; Department of Neurology, Vanderbilt University Medical Center, Nashville, TN 37232, United States of America; Vanderbilt Brain Institute, Vanderbilt University Medical Center, Nashville, TN 37232, United States of America
| | - Oscar L Lopez
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, United States of America; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, United States of America
| | - Renã A S Robinson
- Department of Chemistry, Vanderbilt University, Nashville, TN 37235, United States of America; Vanderbilt Memory & Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN 37212, United States of America; Department of Neurology, Vanderbilt University Medical Center, Nashville, TN 37232, United States of America; Vanderbilt Brain Institute, Vanderbilt University Medical Center, Nashville, TN 37232, United States of America; Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN 37232, United States of America.
| |
Collapse
|
40
|
Fowler AJ, Hebron M, Balaraman K, Shi W, Missner AA, Greenzaid JD, Chiu TL, Ullman C, Weatherdon E, Duka V, Torres-Yaghi Y, Pagan FL, Liu X, Ressom H, Ahn J, Wolf C, Moussa C. Discoidin Domain Receptor 1 is a therapeutic target for neurodegenerative diseases. Hum Mol Genet 2020; 29:2882-2898. [PMID: 32776088 PMCID: PMC7566445 DOI: 10.1093/hmg/ddaa177] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/29/2020] [Accepted: 08/04/2020] [Indexed: 12/13/2022] Open
Abstract
The role of Discoidin Domain Receptors (DDRs) is poorly understood in neurodegeneration. DDRs are upregulated in Alzheimer's and Parkinson's disease (PD), and DDRs knockdown reduces neurotoxic protein levels. Here we show that potent and preferential DDR1 inhibitors reduce neurotoxic protein levels in vitro and in vivo. Partial or complete deletion or inhibition of DDR1 in a mouse model challenged with α-synuclein increases autophagy and reduces inflammation and neurotoxic proteins. Significant changes of cerebrospinal fluid microRNAs that control inflammation, neuronal injury, autophagy and vesicular transport genes are observed in PD with and without dementia and Lewy body dementia, but these changes are attenuated or reversed after treatment with the DDR1 inhibitor, nilotinib. Collectively, these data demonstrate that DDR1 regulates autophagy and reduces neurotoxic proteins and inflammation and is a therapeutic target in neurodegeneration.
Collapse
Affiliation(s)
- Alan J Fowler
- Department of Neurology, Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Lewy Body Dementia Association, Research Center of Excellence, Georgetown University Medical Center, Washington, DC 20057, USA
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC 20057, USA
- Georgetown Howard Universities Center for Clinical and Translational Sciences, Translational Biomedical Sciences Program, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Michaeline Hebron
- Department of Neurology, Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Lewy Body Dementia Association, Research Center of Excellence, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Kaluvu Balaraman
- Department of Chemistry, Georgetown University and Medicinal Chemistry Shared Resource, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Wangke Shi
- Department of Neurology, Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Lewy Body Dementia Association, Research Center of Excellence, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Alexander A Missner
- Department of Neurology, Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Lewy Body Dementia Association, Research Center of Excellence, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Jonathan D Greenzaid
- Department of Neurology, Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Lewy Body Dementia Association, Research Center of Excellence, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Timothy L Chiu
- Department of Neurology, Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Lewy Body Dementia Association, Research Center of Excellence, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Clementina Ullman
- Department of Neurology, Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Lewy Body Dementia Association, Research Center of Excellence, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Ethan Weatherdon
- Department of Neurology, Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Lewy Body Dementia Association, Research Center of Excellence, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Val Duka
- Department of Neurology, Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Lewy Body Dementia Association, Research Center of Excellence, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Yasar Torres-Yaghi
- MedStar Georgetown University Hospital, Movement Disorders Clinic, Department of Neurology, Washington, DC 20057, USA
| | - Fernando L Pagan
- MedStar Georgetown University Hospital, Movement Disorders Clinic, Department of Neurology, Washington, DC 20057, USA
| | - Xiaoguang Liu
- Department of Neurology, Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Lewy Body Dementia Association, Research Center of Excellence, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Habtom Ressom
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Jaeil Ahn
- Department of Bioinformatics, Biostatistics, and Biomathematics, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Christian Wolf
- Department of Chemistry, Georgetown University and Medicinal Chemistry Shared Resource, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Charbel Moussa
- Department of Neurology, Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Lewy Body Dementia Association, Research Center of Excellence, Georgetown University Medical Center, Washington, DC 20057, USA
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC 20057, USA
| |
Collapse
|
41
|
Fan W, Mai L, Zhu X, Huang F, He H. The Role of Microglia in Perioperative Neurocognitive Disorders. Front Cell Neurosci 2020; 14:261. [PMID: 32973455 PMCID: PMC7461842 DOI: 10.3389/fncel.2020.00261] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 07/27/2020] [Indexed: 12/12/2022] Open
Abstract
Perioperative neurocognitive disorder (PND) is a common phenomenon associated with anesthesia and surgery and has been frequently described in the elderly and susceptible individuals. Microglia, which are the brain’s major resident immune cells, play critical roles in maintaining neuronal homeostasis and synaptic plasticity. Accumulating evidence suggests microglial dysfunction occurring after anesthesia and surgery might perturb neuronal function and induce PND. This review aims to provide an overview of the involvement of microglia in PND to date. Possible cellular and molecular mechanisms regarding the connection between microglial activation and PND are discussed.
Collapse
Affiliation(s)
- Wenguo Fan
- Department of Anesthesiology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Lijia Mai
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Xiao Zhu
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China
| | - Fang Huang
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Hongwen He
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| |
Collapse
|
42
|
Sayed A, Bahbah EI, Kamel S, Barreto GE, Ashraf GM, Elfil M. The neutrophil-to-lymphocyte ratio in Alzheimer's disease: Current understanding and potential applications. J Neuroimmunol 2020; 349:577398. [PMID: 32977249 DOI: 10.1016/j.jneuroim.2020.577398] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/17/2020] [Accepted: 09/17/2020] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disorder affecting millions of people, and its prevalence is expected to continue to grow in the older age population. It is characterized by cognitive impairment and dementia on the long term leading to a wide spectrum of social and financial burdens. Due to these burdens, there is a need to have a better understanding of the disease pathophysiology as well as to come up with more readily available and cost-effective screening tools to provide an acceptable estimate of the disease risk/diagnosis in the early years of the disease before dementia develops as it is in these early years that lifestyle modifications can be more effective in protecting against and delaying the frank cognitive impairment associated with AD. Recently, there has been a more detailed, yet incomplete, comprehension of the inflammatory component of the AD pathophysiology. The inflammatory response in AD entails hyperactivation of neutrophils with noticeable changes in their subsets and increased migration of lymphocytes into the central nervous system (CNS) across the compromised blood-brain barrier (BBB). These changes in the counts of the different immune cells in AD allowed for pursuing a new cost-effective, and more widely accessible diagnostic tool, which is the neutrophil-to-lymphocyte ratio (NLR). In this review, we aimed to discuss the inflammatory response in AD, and how this response is reflected in the counts of different immune cells, mainly neutrophils and lymphocytes which can be implemented in the utility of NLR as a diagnostic tool in AD patients.
Collapse
Affiliation(s)
- Ahmed Sayed
- Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Eshak I Bahbah
- Faculty of Medicine, Al-Azhar University, Damietta, Egypt
| | | | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland; Health Research Institute, University of Limerick, Limerick, Ireland
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Mohamed Elfil
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
43
|
Patel RA, Wharton W, Bay AA, Nid L, Barter JD, Hackney ME. Association between anti-inflammatory interleukin-10 and executive function in African American women at risk for Alzheimer's disease. J Clin Exp Neuropsychol 2020; 42:647-659. [PMID: 32781877 PMCID: PMC9747330 DOI: 10.1080/13803395.2020.1798879] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION African-Americans (AAs) are 64% more likely to be diagnosed with AD than non-Hispanic Whites. AAs with elevated AD biomarkers exhibit greater neurodegeneration in AD signature regions compared to non-Hispanic Whites with elevated AD biomarkers. This pilot trial examined whether normal or elevated plasma levels of interleukin (IL)-10 are associated with changes in executive function and short-term memory in AA women at risk for developing AD due to parental history. METHOD Observational study comparing groups with elevated and normal plasma IL-10 levels. Study included 31 AA women (age=58.9±8 years) with parental history of AD. Measures included inflammatory blood biomarkers, executive function and visuospatial short-term memory tests. Multivariate linear regression with adjustment for comorbidities, and Bonferroni corrections for multiple comparisons were used to compare groups. Effect sizes (Cohen's d) were generated. Using endpoints with moderate-large effects between groups, Pearson correlations determined associations between biomarker levels and cognitive performance. RESULTS The elevated IL-10 group performed worse on the Trail-Making Test proportional score ((B-A)/A) (effect size (d =-0.87 (-1.6, -.1)). Moderate effects with large confident intervals were noted in inhibition, set-switching, and body position spatial memory. Significant differences between groups in levels of other inflammatory markers were noted, including IL-7 (p=0.002) and interferon γ (p=0.02). IL-7 remained significant after Bonferroni correction. Correlation matrices revealed moderate-large, significant correlations (yet with wide confidence intervals) between levels of IL-10 and IL-9 with BPST total correct trials, and between interferon γ and delayed recall. CONCLUSIONS Interleukins may incite inflammation, leading to impaired aspects of executive function and short-term memory in this sample of African American women at risk for developing AD. This research provides effect sizes that will be used to power future research that will further investigate the relationship between inflammation, AD biomarkers, and cognitive function in an understudied population.
Collapse
Affiliation(s)
- Ruhee A. Patel
- Emory University College of Arts and Sciences, Atlanta, USA
| | - Whitney Wharton
- Emory University School of Nursing, Atlanta, USA,Department of Neurology, Emory University School of Medicine, Atlanta, USA
| | - Allison A. Bay
- Division of General Medicine and Geriatrics, Department of Medicine, Emory University School of Medicine, Atlanta, USA
| | - Liang Nid
- Division of General Medicine and Geriatrics, Department of Medicine, Emory University School of Medicine, Atlanta, USA,Emory University Rollins School of Public Health, Atlanta, USA
| | - Jolie D. Barter
- Division of General Medicine and Geriatrics, Department of Medicine, Emory University School of Medicine, Atlanta, USA
| | - Madeleine E. Hackney
- Emory University School of Nursing, Atlanta, USA,Division of General Medicine and Geriatrics, Department of Medicine, Emory University School of Medicine, Atlanta, USA,Department of Rehabilitation Medicine, Emory University School of Medicine, Atlanta, USA,Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Decatur, GA, USA,Birmingham/Atlanta VA Geriatric Research Clinical and Education Center, Atlanta, USA
| |
Collapse
|
44
|
Benameur K, Agarwal A, Auld SC, Butters MP, Webster AS, Ozturk T, Howell JC, Bassit LC, Velasquez A, Schinazi RF, Mullins ME, Hu WT. Encephalopathy and Encephalitis Associated with Cerebrospinal Fluid Cytokine Alterations and Coronavirus Disease, Atlanta, Georgia, USA, 2020. Emerg Infect Dis 2020; 26:2016-2021. [PMID: 32487282 PMCID: PMC7454059 DOI: 10.3201/eid2609.202122] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
There are few detailed investigations of neurologic complications in severe acute respiratory syndrome coronavirus 2 infection. We describe 3 patients with laboratory-confirmed coronavirus disease who had encephalopathy and encephalitis develop. Neuroimaging showed nonenhancing unilateral, bilateral, and midline changes not readily attributable to vascular causes. All 3 patients had increased cerebrospinal fluid (CSF) levels of anti-S1 IgM. One patient who died also had increased levels of anti-envelope protein IgM. CSF analysis also showed markedly increased levels of interleukin (IL)-6, IL-8, and IL-10, but severe acute respiratory syndrome coronavirus 2 was not identified in any CSF sample. These changes provide evidence of CSF periinfectious/postinfectious inflammatory changes during coronavirus disease with neurologic complications.
Collapse
|
45
|
Misiura MB, Howell JC, Wu J, Qiu D, Parker MW, Turner JA, Hu WT. Race modifies default mode connectivity in Alzheimer's disease. Transl Neurodegener 2020; 9:8. [PMID: 32099645 PMCID: PMC7029517 DOI: 10.1186/s40035-020-0186-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 02/04/2020] [Indexed: 12/11/2022] Open
Abstract
Background Older African Americans are more likely to develop Alzheimer's disease (AD) than older Caucasians, and this difference cannot be readily explained by cerebrovascular and socioeconomic factors alone. We previously showed that mild cognitive impairment and AD dementia were associated with attenuated increases in the cerebrospinal fluid (CSF) levels of total and phosphorylated tau in African Americans compared to Caucasians, even though there was no difference in beta-amyloid 1-42 level between the two races. Methods We extended our work by analyzing early functional magnetic resonance imaging (fMRI) biomarkers of the default mode network in older African Americans and Caucasians. We calculated connectivity between nodes of the regions belonging to the various default mode network subsystems and correlated these imaging biomarkers with non-imaging biomarkers implicated in AD (CSF amyloid, total tau, and cognitive performance). Results We found that race modifies the relationship between functional connectivity of default mode network subsystems and cognitive performance, tau, and amyloid levels. Conclusion These findings provide further support that race modifies the AD phenotypes downstream from cerebral amyloid deposition, and identifies key inter-subsystem connections for deep imaging and neuropathologic characterization.
Collapse
Affiliation(s)
- Maria B Misiura
- 1Department of Psychology, Georgia State University, Atlanta, GA USA.,2Departments of Neurology, Emory University, 615 Michael Street, Suite 505, Atlanta, GA 30322 USA
| | - J Christina Howell
- 2Departments of Neurology, Emory University, 615 Michael Street, Suite 505, Atlanta, GA 30322 USA
| | - Junjie Wu
- 3Departments of Radiology, Emory University, Atlanta, GA USA
| | - Deqiang Qiu
- 3Departments of Radiology, Emory University, Atlanta, GA USA
| | - Monica W Parker
- 2Departments of Neurology, Emory University, 615 Michael Street, Suite 505, Atlanta, GA 30322 USA
| | - Jessica A Turner
- 1Department of Psychology, Georgia State University, Atlanta, GA USA
| | - William T Hu
- 2Departments of Neurology, Emory University, 615 Michael Street, Suite 505, Atlanta, GA 30322 USA
| |
Collapse
|