LIVIVO - The Search Portal for Life Sciences

zur deutschen Oberfläche wechseln
Advanced search

Search results

Result 1 - 10 of total 19

Search options

  1. Article: Human disease-specific cell signatures in non-lesional tissue in Multiple Sclerosis detected by single-cell and spatial transcriptomics.

    Lam, Matti / Lee, Dylan / Kosater, Ivy / Khairallah, Anthony / Taga, Mariko / Zhang, Ya / Fujita, Masashi / Nag, Sukriti / Bennett, David A / De Jager, Philip / Menon, Vilas

    bioRxiv : the preprint server for biology

    2023  

    Abstract: Recent investigations of cell type changes in Multiple Sclerosis (MS) using single-cell profiling methods have focused on active lesional and peri-lesional brain tissue, and have implicated a number of peripheral and central nervous system cell types. ... ...

    Abstract Recent investigations of cell type changes in Multiple Sclerosis (MS) using single-cell profiling methods have focused on active lesional and peri-lesional brain tissue, and have implicated a number of peripheral and central nervous system cell types. However, an important question is the extent to which so-called "normal-appearing" non-lesional tissue in individuals with MS accumulate changes over the lifespan. Here, we compared post-mortem non-lesional brain tissue from donors with a pathological or clinical diagnosis of MS from the Religious Orders Study or Rush Memory and Aging Project (ROSMAP) cohorts to age- and sex-matched brains from persons without MS (controls). We profiled three brain regions using single-nucleus RNA-seq: dorsolateral prefrontal cortex (DLPFC), normal appearing white matter (NAWM) and the pulvinar in thalamus (PULV), from 15 control individuals, 8 individuals with MS, and 5 individuals with other detrimental pathologies accompanied by demyelination, resulting in a total of 78 samples. We identified region- and cell type-specific differences in non-lesional samples from individuals diagnosed with MS and/or exhibiting secondary demyelination with other neurological conditions, as compared to control donors. These differences included lower proportions of oligodendrocytes with expression of myelination related genes MOBP, MBP, PLP1, as well as higher proportions of CRYAB+ oligodendrocytes in all three brain regions. Among microglial signatures, we identified subgroups that were higher in both demyelination (TMEM163+/ERC2+), as well as those that were specifically higher in MS donors (HIF1A+/SPP1+) and specifically in donors with secondary demyelination (SOCS6+/MYO1E+), in both white and grey matter. To validate our findings, we generated Visium spatial transcriptomics data on matched tissue from 13 donors, and recapitulated our observations of gene expression differences in oligodendrocytes and microglia. Finally, we show that some of the differences observed between control and MS donors in NAWM mirror those previously reported between control WM and active lesions in MS donors. Overall, our investigation sheds additional light on cell type- and disease-specific differences present even in non-lesional white and grey matter tissue, highlighting widespread cellular signatures that may be associated with downstream pathological changes.
    Language English
    Publishing date 2023-12-20
    Publishing country United States
    Document type Preprint
    DOI 10.1101/2023.12.20.572491
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  2. Article ; Online: Multicellular communities are perturbed in the aging human brain and Alzheimer's disease.

    Cain, Anael / Taga, Mariko / McCabe, Cristin / Green, Gilad S / Hekselman, Idan / White, Charles C / Lee, Dylan I / Gaur, Pallavi / Rozenblatt-Rosen, Orit / Zhang, Feng / Yeger-Lotem, Esti / Bennett, David A / Yang, Hyun-Sik / Regev, Aviv / Menon, Vilas / Habib, Naomi / De Jager, Philip L

    Nature neuroscience

    2023  Volume 26, Issue 7, Page(s) 1267–1280

    Abstract: The role of different cell types and their interactions in Alzheimer's disease (AD) is a complex and open question. Here, we pursued this question by assembling a high-resolution cellular map of the aging frontal cortex using single-nucleus RNA ... ...

    Abstract The role of different cell types and their interactions in Alzheimer's disease (AD) is a complex and open question. Here, we pursued this question by assembling a high-resolution cellular map of the aging frontal cortex using single-nucleus RNA sequencing of 24 individuals with a range of clinicopathologic characteristics. We used this map to infer the neocortical cellular architecture of 638 individuals profiled by bulk RNA sequencing, providing the sample size necessary for identifying statistically robust associations. We uncovered diverse cell populations associated with AD, including a somatostatin inhibitory neuronal subtype and oligodendroglial states. We further identified a network of multicellular communities, each composed of coordinated subpopulations of neuronal, glial and endothelial cells, and we found that two of these communities are altered in AD. Finally, we used mediation analyses to prioritize cellular changes that might contribute to cognitive decline. Thus, our deconstruction of the aging neocortex provides a roadmap for evaluating the cellular microenvironments underlying AD and dementia.
    MeSH term(s) Humans ; Alzheimer Disease/metabolism ; Endothelial Cells/metabolism ; Brain/metabolism ; Aging/pathology ; Cognitive Dysfunction/pathology ; Neocortex/pathology
    Language English
    Publishing date 2023-06-19
    Publishing country United States
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't
    ZDB-ID 1420596-8
    ISSN 1546-1726 ; 1097-6256
    ISSN (online) 1546-1726
    ISSN 1097-6256
    DOI 10.1038/s41593-023-01356-x
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  3. Article: Cellular dynamics across aged human brains uncover a multicellular cascade leading to Alzheimer's disease.

    Green, Gilad Sahar / Fujita, Masashi / Yang, Hyun-Sik / Taga, Mariko / McCabe, Cristin / Cain, Anael / White, Charles C / Schmidtner, Anna K / Zeng, Lu / Wang, Yangling / Regev, Aviv / Menon, Vilas / Bennett, David A / Habib, Naomi / De Jager, Philip L

    bioRxiv : the preprint server for biology

    2023  

    Abstract: Alzheimer's Disease (AD) is a progressive neurodegenerative disease seen with advancing age. Recent studies have revealed diverse AD-associated cell states, yet when and how they impact the causal chain leading to AD remains unknown. To reconstruct the ... ...

    Abstract Alzheimer's Disease (AD) is a progressive neurodegenerative disease seen with advancing age. Recent studies have revealed diverse AD-associated cell states, yet when and how they impact the causal chain leading to AD remains unknown. To reconstruct the dynamics of the brain's cellular environment along the disease cascade and to distinguish between AD and aging effects, we built a comprehensive cell atlas of the aged prefrontal cortex from 1.64 million single-nucleus RNA-seq profiles. We associated glial, vascular and neuronal subpopulations with AD-related traits for 424 aging individuals, and aligned them along the disease cascade using causal modeling. We identified two distinct lipid-associated microglial subpopulations, one contributed to amyloid-β proteinopathy while the other mediated the effect of amyloid-β in accelerating tau proteinopathy, as well as an astrocyte subpopulation that mediated the effect of tau on cognitive decline. To model the coordinated dynamics of the entire cellular environment we devised the BEYOND methodology which uncovered two distinct trajectories of brain aging that are defined by distinct sequences of changes in cellular communities. Older individuals are engaged in one of two possible trajectories, each associated with progressive changes in specific cellular communities that end with: (1) AD dementia or (2) alternative brain aging. Thus, we provide a cellular foundation for a new perspective of AD pathophysiology that could inform the development of new therapeutic interventions targeting cellular communities, while designing a different clinical management for those individuals on the path to AD or to alternative brain aging.
    Language English
    Publishing date 2023-03-09
    Publishing country United States
    Document type Preprint
    DOI 10.1101/2023.03.07.531493
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  4. Article ; Online: INPP5D regulates inflammasome activation in human microglia.

    Chou, Vicky / Pearse, Richard V / Aylward, Aimee J / Ashour, Nancy / Taga, Mariko / Terzioglu, Gizem / Fujita, Masashi / Fancher, Seeley B / Sigalov, Alina / Benoit, Courtney R / Lee, Hyo / Lam, Matti / Seyfried, Nicholas T / Bennett, David A / De Jager, Philip L / Menon, Vilas / Young-Pearse, Tracy L

    Nature communications

    2023  Volume 14, Issue 1, Page(s) 7552

    Abstract: Microglia and neuroinflammation play an important role in the development and progression of Alzheimer's disease (AD). Inositol polyphosphate-5-phosphatase D (INPP5D/SHIP1) is a myeloid-expressed gene genetically-associated with AD. Through unbiased ... ...

    Abstract Microglia and neuroinflammation play an important role in the development and progression of Alzheimer's disease (AD). Inositol polyphosphate-5-phosphatase D (INPP5D/SHIP1) is a myeloid-expressed gene genetically-associated with AD. Through unbiased analyses of RNA and protein profiles in INPP5D-disrupted iPSC-derived human microglia, we find that reduction in INPP5D activity is associated with molecular profiles consistent with disrupted autophagy and inflammasome activation. These findings are validated through targeted pharmacological experiments which demonstrate that reduced INPP5D activity induces the formation of the NLRP3 inflammasome, cleavage of CASP1, and secretion of IL-1β and IL-18. Further, in-depth analyses of human brain tissue across hundreds of individuals using a multi-analytic approach provides evidence that a reduction in function of INPP5D in microglia results in inflammasome activation in AD. These findings provide insights into the molecular mechanisms underlying microglia-mediated processes in AD and highlight the inflammasome as a potential therapeutic target for modulating INPP5D-mediated vulnerability to AD.
    MeSH term(s) Humans ; Inflammasomes/metabolism ; NLR Family, Pyrin Domain-Containing 3 Protein/genetics ; NLR Family, Pyrin Domain-Containing 3 Protein/metabolism ; Microglia/metabolism ; Alzheimer Disease/metabolism ; Brain/metabolism ; Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/metabolism
    Chemical Substances Inflammasomes ; NLR Family, Pyrin Domain-Containing 3 Protein ; INPP5D protein, human (EC 3.1.3.86) ; Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases (EC 3.1.3.86)
    Language English
    Publishing date 2023-11-29
    Publishing country England
    Document type Journal Article
    ZDB-ID 2553671-0
    ISSN 2041-1723 ; 2041-1723
    ISSN (online) 2041-1723
    ISSN 2041-1723
    DOI 10.1038/s41467-023-42819-w
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  5. Article ; Online: Cell-type-specific regulation of APOE and CLU levels in human neurons by the Alzheimer's disease risk gene SORL1.

    Lee, Hyo / Aylward, Aimee J / Pearse, Richard V / Lish, Alexandra M / Hsieh, Yi-Chen / Augur, Zachary M / Benoit, Courtney R / Chou, Vicky / Knupp, Allison / Pan, Cheryl / Goberdhan, Srilakshmi / Duong, Duc M / Seyfried, Nicholas T / Bennett, David A / Taga, Mariko F / Huynh, Kevin / Arnold, Matthias / Meikle, Peter J / De Jager, Philip L /
    Menon, Vilas / Young, Jessica E / Young-Pearse, Tracy L

    Cell reports

    2023  Volume 42, Issue 8, Page(s) 112994

    Abstract: SORL1 is implicated in the pathogenesis of Alzheimer's disease (AD) through genetic studies. To interrogate the roles of SORL1 in human brain cells, SORL1-null induced pluripotent stem cells (iPSCs) were differentiated to neuron, astrocyte, microglial, ... ...

    Abstract SORL1 is implicated in the pathogenesis of Alzheimer's disease (AD) through genetic studies. To interrogate the roles of SORL1 in human brain cells, SORL1-null induced pluripotent stem cells (iPSCs) were differentiated to neuron, astrocyte, microglial, and endothelial cell fates. Loss of SORL1 leads to alterations in both overlapping and distinct pathways across cell types, with the greatest effects in neurons and astrocytes. SORL1 loss induces a neuron-specific reduction in apolipoprotein E (APOE) and clusterin (CLU) and altered lipid profiles. Analyses of iPSCs derived from a large cohort reveal a neuron-specific association between SORL1, APOE, and CLU levels, a finding validated in postmortem brain. Enhancement of retromer-mediated trafficking rescues tau phenotypes observed in SORL1-null neurons but does not rescue APOE levels. Pathway analyses implicate transforming growth factor β (TGF-β)/SMAD signaling in SORL1 function, and modulating SMAD signaling in neurons alters APOE RNA levels in a SORL1-dependent manner. Taken together, these data provide a mechanistic link between strong genetic risk factors for AD.
    MeSH term(s) Humans ; Clusterin/genetics ; Alzheimer Disease/genetics ; Neurons ; Cell Growth Processes ; Apolipoproteins E/genetics ; LDL-Receptor Related Proteins/genetics ; Membrane Transport Proteins
    Chemical Substances Clusterin ; Apolipoproteins E ; SORL1 protein, human ; LDL-Receptor Related Proteins ; Membrane Transport Proteins ; CLU protein, human
    Language English
    Publishing date 2023-08-22
    Publishing country United States
    Document type Journal Article ; Research Support, N.I.H., Extramural
    ZDB-ID 2649101-1
    ISSN 2211-1247 ; 2211-1247
    ISSN (online) 2211-1247
    ISSN 2211-1247
    DOI 10.1016/j.celrep.2023.112994
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  6. Article ; Online: Deconvolving the contributions of cell-type heterogeneity on cortical gene expression.

    Patrick, Ellis / Taga, Mariko / Ergun, Ayla / Ng, Bernard / Casazza, William / Cimpean, Maria / Yung, Christina / Schneider, Julie A / Bennett, David A / Gaiteri, Chris / De Jager, Philip L / Bradshaw, Elizabeth M / Mostafavi, Sara

    PLoS computational biology

    2020  Volume 16, Issue 8, Page(s) e1008120

    Abstract: Complexity of cell-type composition has created much skepticism surrounding the interpretation of bulk tissue transcriptomic studies. Recent studies have shown that deconvolution algorithms can be applied to computationally estimate cell-type proportions ...

    Abstract Complexity of cell-type composition has created much skepticism surrounding the interpretation of bulk tissue transcriptomic studies. Recent studies have shown that deconvolution algorithms can be applied to computationally estimate cell-type proportions from gene expression data of bulk blood samples, but their performance when applied to brain tissue is unclear. Here, we have generated an immunohistochemistry (IHC) dataset for five major cell-types from brain tissue of 70 individuals, who also have bulk cortical gene expression data. With the IHC data as the benchmark, this resource enables quantitative assessment of deconvolution algorithms for brain tissue. We apply existing deconvolution algorithms to brain tissue by using marker sets derived from human brain single cell and cell-sorted RNA-seq data. We show that these algorithms can indeed produce informative estimates of constituent cell-type proportions. In fact, neuronal subpopulations can also be estimated from bulk brain tissue samples. Further, we show that including the cell-type proportion estimates as confounding factors is important for reducing false associations between Alzheimer's disease phenotypes and gene expression. Lastly, we demonstrate that using more accurate marker sets can substantially improve statistical power in detecting cell-type specific expression quantitative trait loci (eQTLs).
    MeSH term(s) Algorithms ; Brain/cytology ; Brain/metabolism ; Computational Biology ; Gene Expression Profiling/methods ; Humans ; Immunohistochemistry ; Organ Specificity/genetics ; Phenotype ; Quantitative Trait Loci/genetics ; Sequence Analysis, RNA/methods ; Single-Cell Analysis ; Transcriptome/genetics
    Language English
    Publishing date 2020-08-17
    Publishing country United States
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't
    ZDB-ID 2193340-6
    ISSN 1553-7358 ; 1553-734X
    ISSN (online) 1553-7358
    ISSN 1553-734X
    DOI 10.1371/journal.pcbi.1008120
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  7. Article ; Online: A cortical immune network map identifies distinct microglial transcriptional programs associated with β-amyloid and Tau pathologies.

    Patrick, Ellis / Olah, Marta / Taga, Mariko / Klein, Hans-Ulrich / Xu, Jishu / White, Charles C / Felsky, Daniel / Agrawal, Sonal / Gaiteri, Chris / Chibnik, Lori B / Mostafavi, Sara / Schneider, Julie A / Bennett, David A / Bradshaw, Elizabeth M / De Jager, Philip L

    Translational psychiatry

    2021  Volume 11, Issue 1, Page(s) 50

    Abstract: Microglial dysfunction has been proposed as one of the many cellular mechanisms that can contribute to the development of Alzheimer's disease (AD). Here, using a transcriptional network map of the human frontal cortex, we identify five modules of co- ... ...

    Abstract Microglial dysfunction has been proposed as one of the many cellular mechanisms that can contribute to the development of Alzheimer's disease (AD). Here, using a transcriptional network map of the human frontal cortex, we identify five modules of co-expressed genes related to microglia and assess their role in the neuropathologic features of AD in 540 subjects from two cohort studies of brain aging. Two of these transcriptional programs-modules 113 and 114-relate to the accumulation of β-amyloid, while module 5 relates to tau pathology. We replicate these associations in brain epigenomic data and in two independent datasets. In terms of tau, we propose that module 5, a marker of activated microglia, may lead to tau accumulation and subsequent cognitive decline. We validate our model further by showing that three representative module 5 genes (ACADVL, TRABD, and VASP) encode proteins that are upregulated in activated microglia in AD.
    MeSH term(s) Alzheimer Disease/genetics ; Amyloid beta-Peptides/metabolism ; Brain/metabolism ; Cognitive Dysfunction ; Humans ; Microglia/metabolism ; tau Proteins/genetics ; tau Proteins/metabolism
    Chemical Substances Amyloid beta-Peptides ; tau Proteins
    Language English
    Publishing date 2021-01-14
    Publishing country United States
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't
    ZDB-ID 2609311-X
    ISSN 2158-3188 ; 2158-3188
    ISSN (online) 2158-3188
    ISSN 2158-3188
    DOI 10.1038/s41398-020-01175-9
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  8. Article ; Online: Modulation of oxidative stress and tau phosphorylation by the mTOR activator phosphatidic acid in SH-SY5Y cells.

    Taga, Mariko / Mouton-Liger, François / Paquet, Claire / Hugon, Jacques

    FEBS letters

    2011  Volume 585, Issue 12, Page(s) 1801–1806

    Abstract: The mammalian target of rapamycin complex 1 (mTORC1) pathway including p70(S6K) (the 70-kDa p70 S6 kinase) and S6, controls protein synthesis, has anti-apoptotic functions and can phosphorylate tau protein. mTORC1 is triggered by nutrients such as ... ...

    Abstract The mammalian target of rapamycin complex 1 (mTORC1) pathway including p70(S6K) (the 70-kDa p70 S6 kinase) and S6, controls protein synthesis, has anti-apoptotic functions and can phosphorylate tau protein. mTORC1 is triggered by nutrients such as phosphatidic acid (PA). Previous experimental studies have shown that oxidative stress may down-regulate this pathway leading to neuronal death. Our results showed that in human neuroblastoma cells, PA exposure can reduce H(2)O(2)-induced apoptosis and can increase tau protein phosphorylation on Ser214 via p70(S6K) activation. These findings reveal that PA, via the mTOR kinase, can trigger tau phosphorylation on a site known to reduce paired helical filament (PHF) formation.
    MeSH term(s) Cell Line, Tumor ; Humans ; Neuroblastoma/metabolism ; Neuroblastoma/pathology ; Oxidative Stress ; Phosphatidic Acids/physiology ; Phosphorylation ; TOR Serine-Threonine Kinases/metabolism ; tau Proteins/metabolism
    Chemical Substances MAPT protein, human ; Phosphatidic Acids ; tau Proteins ; MTOR protein, human (EC 2.7.1.1) ; TOR Serine-Threonine Kinases (EC 2.7.1.1)
    Language English
    Publishing date 2011-06-23
    Publishing country England
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 212746-5
    ISSN 1873-3468 ; 0014-5793
    ISSN (online) 1873-3468
    ISSN 0014-5793
    DOI 10.1016/j.febslet.2011.04.022
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  9. Article ; Online: Metaflammasome components in the human brain: a role in dementia with Alzheimer's pathology?

    Taga, Mariko / Minett, Thais / Classey, John / Matthews, Fiona E / Brayne, Carol / Ince, Paul G / Nicoll, James Ar / Hugon, Jacques / Boche, Delphine

    Brain pathology (Zurich, Switzerland)

    2016  Volume 27, Issue 3, Page(s) 266–275

    Abstract: Epidemiological and genetic studies have identified metabolic disorders and inflammation as risk factors for Alzheimer's disease (AD). Evidence in obesity and type-2 diabetes suggests a role for a metabolic inflammasome ("metaflammasome") in mediating ... ...

    Abstract Epidemiological and genetic studies have identified metabolic disorders and inflammation as risk factors for Alzheimer's disease (AD). Evidence in obesity and type-2 diabetes suggests a role for a metabolic inflammasome ("metaflammasome") in mediating chronic inflammation in peripheral organs implicating IKKβ (inhibitor of nuclear factor kappa-B kinase subunit beta), IRS1 (insulin receptor substrate 1), JNK (c-jun N-terminal kinase), and PKR (double-stranded RNA protein kinase). We hypothesized that these proteins are expressed in the brain in response to metabolic risk factors in AD. Neocortex from 299 participants from the MRC Cognitive Function and Ageing Studies was analysed by immunohistochemistry for the expression of the phosphorylated (active) form of IKKβ [pSer
    MeSH term(s) Aged ; Aged, 80 and over ; Alzheimer Disease/genetics ; Alzheimer Disease/metabolism ; Alzheimer Disease/pathology ; Apolipoproteins E/genetics ; Cohort Studies ; Diabetes Mellitus, Type 2/metabolism ; Diabetes Mellitus, Type 2/pathology ; Female ; Humans ; I-kappa B Kinase/metabolism ; Immunohistochemistry ; Insulin Receptor Substrate Proteins/metabolism ; MAP Kinase Kinase 4/metabolism ; Male ; Mental Status Schedule ; Neocortex/metabolism ; Neocortex/pathology ; Phosphorylation ; Polymorphism, Genetic ; Risk Factors ; eIF-2 Kinase/metabolism
    Chemical Substances Apolipoproteins E ; IRS1 protein, human ; Insulin Receptor Substrate Proteins ; EIF2AK2 protein, human (EC 2.7.11.1) ; eIF-2 Kinase (EC 2.7.11.1) ; I-kappa B Kinase (EC 2.7.11.10) ; IKBKB protein, human (EC 2.7.11.10) ; MAP Kinase Kinase 4 (EC 2.7.12.2)
    Language English
    Publishing date 2016-06-08
    Publishing country Switzerland
    Document type Journal Article ; Multicenter Study ; Research Support, Non-U.S. Gov't
    ZDB-ID 1051484-3
    ISSN 1750-3639 ; 1015-6305
    ISSN (online) 1750-3639
    ISSN 1015-6305
    DOI 10.1111/bpa.12388
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  10. Article ; Online: Microglial immunophenotype in dementia with Alzheimer's pathology.

    Minett, Thais / Classey, John / Matthews, Fiona E / Fahrenhold, Marie / Taga, Mariko / Brayne, Carol / Ince, Paul G / Nicoll, James A R / Boche, Delphine

    Journal of neuroinflammation

    2016  Volume 13, Issue 1, Page(s) 135

    Abstract: Background: Genetic risk factors for Alzheimer's disease imply that inflammation plays a causal role in development of the disease. Experimental studies suggest that microglia, as the brain macrophages, have diverse functions, with their main role in ... ...

    Abstract Background: Genetic risk factors for Alzheimer's disease imply that inflammation plays a causal role in development of the disease. Experimental studies suggest that microglia, as the brain macrophages, have diverse functions, with their main role in health being to survey the brain parenchyma through highly motile processes.
    Methods: Using the Medical Research Council Cognitive Function and Ageing Studies resources, we have immunophenotyped microglia to investigate their role in dementia with Alzheimer's pathology. Cerebral cortex obtained at post-mortem from 299 participants was analysed by immunohistochemistry for cluster of differentiation (CD)68 (phagocytosis), human leukocyte antigen (HLA)-DR (antigen-presenting function), ionized calcium-binding adaptor molecule (Iba1) (microglial motility), macrophage scavenger receptor (MSR)-A (plaque-related phagocytosis) and CD64 (immunoglobulin Fcγ receptor I).
    Results: The presence of dementia was associated positively with CD68 (P < 0.001), MSR-A (P = 0.010) and CD64 (P = 0.007) and negatively with Iba1 (P < 0.001). Among participants without dementia, the cognitive function according to the Mini-Mental State Examination was associated positively with Iba1 (P < 0.001) and negatively with CD68 (P = 0.033), and in participants with dementia and Alzheimer's pathology, positively with all microglial markers except Iba1. Overall, in participants without dementia, the relationship with Alzheimer's pathology was negative or not significant, and positive in participants with dementia and Alzheimer's pathology. Apolipoprotein E (APOE) ε2 allele was associated with expression of Iba1 (P = 0.001) and MSR-A (P < 0.001) and APOE ε4 with CD68, HLA-DR and CD64 (P < 0.001).
    Conclusions: Our findings raise the possibility that in dementia with Alzheimer's pathology, microglia lose motility (Iba-1) necessary to support neurons. Conversely, other microglial proteins (CD68, MSR-A), the role of which is clearance of damaged cellular material, are positively associated with Alzheimer's pathology and impaired cognitive function. In addition, our data imply that microglia may respond differently to Aβ and tau in participants with and without dementia so that the microglial activity could potentially influence the likelihood of developing dementia, as supported by genetic studies, highlighting the complexity and diversity of microglial responses.
    MeSH term(s) Aged ; Aged, 80 and over ; Alzheimer Disease/complications ; Alzheimer Disease/pathology ; Antigens, CD/metabolism ; Antigens, Differentiation, Myelomonocytic/metabolism ; Calcium-Binding Proteins ; Cohort Studies ; Cytokines/metabolism ; DNA-Binding Proteins/metabolism ; Dementia/complications ; Dementia/pathology ; Diagnosis ; Female ; HLA-DR Antigens/metabolism ; Humans ; Male ; Mental Status Schedule ; Methionine Sulfoxide Reductases/metabolism ; Microfilament Proteins ; Microglia/metabolism ; Neuropsychological Tests ; Receptors, IgG/metabolism
    Chemical Substances AIF1 protein, human ; Antigens, CD ; Antigens, Differentiation, Myelomonocytic ; CD68 antigen, human ; Calcium-Binding Proteins ; Cytokines ; DNA-Binding Proteins ; FCGR1A protein, human ; HLA-DR Antigens ; Microfilament Proteins ; Receptors, IgG ; Methionine Sulfoxide Reductases (EC 1.8.4.-) ; methionine sulfoxide reductase (EC 1.8.4.11)
    Language English
    Publishing date 2016-06-02
    Publishing country England
    Document type Journal Article
    ISSN 1742-2094
    ISSN (online) 1742-2094
    DOI 10.1186/s12974-016-0601-z
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

To top