Significance

This study changes our view of Alzheimer’s disease (AD) initiation and progression. Mutations promoting cerebral beta-amyloid (Aβ) deposition guarantee rare genetic forms of AD. Thus, the prevailing hypothesis has been that Aβ is central to initiation and progression of all AD, despite contrary animal and patient evidence. We show that age-related T cells generate neurodegeneration with compelling features of AD in mice, with distinct T cell functions required for pathological initiation and neurodegenerative progression. Knowledge from these mice was applied to successfully predict previously unrecognized features of human AD and generate tools for its clinical management.

Abstract

Cerebral (Aβ) plaque and (pTau) tangle deposition are hallmarks of Alzheimer’s disease (AD), yet are insufficient to confer complete AD-like neurodegeneration experimentally. Factors acting upstream of Aβ/pTau in AD remain unknown, but their identification could enable earlier diagnosis and more effective treatments. T cell abnormalities are emerging AD hallmarks, and CD8 T cells were recently found to mediate neurodegeneration downstream of tangle deposition in hereditary neurodegeneration models. The precise impact of T cells downstream of Aβ/pTau, however, appears to vary depending on the animal model. Our prior work suggested that antigen-specific memory CD8 T (“hiT”) cells act upstream of Aβ/pTau after brain injury. Here, we examine whether hiT cells influence sporadic AD-like pathophysiology upstream of Aβ/pTau. Examining neuropathology, gene expression, and behavior in our hiT mouse model we show that CD8 T cells induce plaque and tangle-like deposition, modulate AD-related genes, and ultimately result in progressive neurodegeneration with both gross and fine features of sporadic human AD. T cells required Perforin to initiate this pathophysiology, and IFNγ for most gene expression changes and progression to more widespread neurodegenerative disease. Analogous antigen-specific memory CD8 T cells were significantly elevated in the brains of human AD patients, and their loss from blood corresponded to sporadic AD and related cognitive decline better than plasma pTau-217, a promising AD biomarker candidate. We identify an age-related factor acting upstream of Aβ/pTau to initiate AD-like pathophysiology, the mechanisms promoting its pathogenicity, and its relevance to human sporadic AD.
Deposits of aggregated amyloid plaques containing beta-amyloid (Aβ) and neurofibrillary tangles (NFTs) comprised of hyperphosphorylated tau (pTau) are the primary neuropathological hallmarks of Alzheimer’s disease (AD). Aβ deposition precedes that of NFTs in all forms of AD, and rare inherited forms of the disease are due to mutated (AD-mut) genes that promote the deposition of toxic Aβ in the brain. Thus, a prominent hypothesis is that Aβ is central to initiation and progression of all AD. While Aβ deposition clearly plays an important role, evidence that it is sufficient to initiate and/or maintain AD pathogenesis is lacking. For example, laboratory animals expressing one or even several human AD-mut transgenes that guarantee AD in their human carriers fail to exhibit either robust neurodegeneration or NFTs (1, 2). Moreover, targeting Aβ deposition has resulted in hundreds of failed clinical trials, with modest and sometimes controversial clinical efficacy demonstrated in a few trials only recently (3). Finally, up to a third of elderly individuals possess Aβ deposition sufficient for AD diagnosis, yet are cognitively normal and may remain so for life (4). These findings suggest that Aβ requires distinct cofactors to initiate AD. The search for such factors, with occasional exceptions (5), has primarily focused on processes downstream of Aβ. A recent finding, however, documented that a patient harboring a deterministic AD-mut gene normally guaranteeing neurodegeneration by age 45, exhibited no clinical symptoms into their 70s (6). This protection was evidently afforded by a second germline mutation in the ApoE3 gene that disrupts lipid metabolism (7), highlighting the possibility that the pathogenic effects of Aβ may be dominantly inhibited by preexisting physiological processes upstream of Aβ. While the precise nature of such processes remains unknown, identifying them could allow earlier diagnosis and lead to more effective AD treatments.
Among Aβ-independent factors potentially contributing to AD pathogenesis, CD8 T cell abnormalities have recently emerged as some of the most intriguing. Recent studies have verified and extended earlier reports of T cell abnormalities in AD-related conditions, including resident memory CD8 T (TRM) and other memory T cell accumulation in CSF and/or brain of aging and AD-afflicted individuals (8, 9). CD8 T cells within CSF also dysregulate chemokine signaling when cognitive impairment is evident, regulate neuronal and synaptic gene expression when they accumulate in the hippocampus, and promote neuroinflammation and cognitive decline, in transgenic (Tg) mice (1012). Moreover, T cells increase in areas of AD-mut Tg (AD-Tg) mouse brain and human AD brain with tau neuropathology (10), which correlates with cognitive symptoms in AD (13, 14). Most surprisingly, CD8 T cells together with microglia and interferon-gamma (IFNγ), were recently shown to mediate neurodegeneration in tau-transgenic mice (15). Finally, a very recent study on CD8 T cells in AD-Tg mice presented evidence that they actually dampen Aβ/pTau pathology through CXCL16-CXCR6 intercellular communication between T cells and microglia (16). Thus, the role of CD8 T cells appears variable, at least in transgenic rodents. Moreover, T cell changes in AD are often presumed to follow Aβ deposition. In all of the studies above, for example, T cell abnormalities were examined after AD pathology was established. The potential role of CD8 T cells upstream of Aβ (or tau) has thus not been examined. Nevertheless, the potential relationship of tissue-resident CD8 T cells in particular to ApoE-mediated AD risk and/or protection makes them especially intriguing in this regard (17).
To examine the effect of aging T cells upstream of Aβ/pTau pathology, we recently developed the “hiT” model of rapid CD8 T cell aging in young mice. In this model, age-related homeostatic CD8 T cell expansion is mimicked on a much faster time scale (18). This results in morphological, functional, and genotypic changes in homeostatically induced (“hi”) T cells similar to those occurring with age. These changes include human-like predominance of CD103+ memory CD8 T cells in the general circulation, and accumulation of a non-Aβ antigen (amyloid precursor protein; APP)-specific age-related subpopulation of T cells in the brain. Transfer of these age-related CD8 T cells from hiT mice into wild-type B6 mice increased human-like amyloid and fibrillar pTau in the brain after percussive brain injury, as well as neuronal marker loss regardless of injury (18). This suggested that age-related CD8 T cells act upstream to promote both Aβ and fibrillar pTau accumulation in a non-AD setting. Here, we examined the relationship of age-related CD8 T cells to events upstream of Aβ, and possibly to other aspects of AD pathophysiology, by first interrogating proteinopathy, neurodegeneration, and behavior in the hiT mouse model. We then examined mechanisms whereby hiT cells mediate pathological characteristics using T cells from functional knockout donors as well as RNAseq analysis. Finally, we examined the clinical relevance of antigen-specific and parental CD8 T cell populations to human AD using flow cytometry, tissue staining, and protein analysis in human patients.

Results

Creation of hiT Mouse.

PBS, or CD8 T cells from mouse wild-type donors were injected into young (6 to 8 wk) B6.Foxn1 hosts (PBS and wt-CD8 groups, respectively). Limited analyses of young B6.Foxn1 hosts injected with wild-type CD4 T cells or CD4 plus CD8 T cells together (wt-CD4 and wt-CD8+CD4 groups, respectively), were also performed (SI Appendix, Fig. S1A). Females were exclusively used to avoid male-specific autoimmune disease dynamics, some of which are associated with CD8 T cell homeostatic expansion, in related mouse strains (19, 20). The donor T cells acquire age-related resident memory-like (TRM) phenotype, as well as age-related genotype and function as they expand. CD8 T cells reactive to a non-Aβ epitope on APP then selectively accumulate in the brain of these hiT (for “homeostatically induced T cell”) mice (18). Circulating APP-reactive CD8 TRM cells were rapidly homeostatically expanded in hiT mice (SI Appendix, Fig. S1B), similar to the reported gradual expansion of age-related CD8 TRM cells in aging humans (21).

Aβ and Neurofibrillary Deposition.

We examined Triton-soluble brain extracts as we previously described (22), to assess potentially toxic Aβ in hiT mice by western blot (WB) using 4G8 (human- and rodent-specific APP/Aβ) and ab14220 (rodent-specific APP/Aβ) antibodies. This revealed relative upregulation of high molecular weight species presumed to be APP and its cleavage products (APPCl) in brains of wt-CD8 group hiT mice 3 and 10 wk after injection (Fig. 1A and SI Appendix, Fig. S2 A and B). Additional isoform- and species-specific antibodies in ELISA revealed that endogenous Aβ1-40 but not Aβ1-42 was relatively increased in wt-CD8 group mice 10 wk postinjection (Fig. 1B), with WBs and tissue staining using 4G8 antibody (recognizing rodent and human Aβ/APP) confirming prominent amyloidosis in the cortex and hippocampus (SI Appendix, Fig. S2B and Fig. 1 CF, respectively). By 6 mo postinjection, wt-CD8 and wt-CD8+CD4 groups exhibited increased Aβ deposition in brain vasculature, whereas wt-CD4 group did not (Fig. 1 D and E and SI Appendix, Fig. S2 C and D), further suggesting CD8 T cells are necessary and sufficient to promote amyloidosis. Aβ plaques in wt-CD8 group brains 15 mo postinjection exhibited typical morphology, but exhibited small compact cores at best, were chiefly detergent-soluble (SI Appendix, Fig. S3), and were minimally stained by curcumin or ThioS (Fig. 1 C and F).
Fig. 1.
Amyloid and Tau pathology. WBs of APP (100 kDa) and its cleavage products (APPCl; <100 kDa) in the brain using antibody 4G8 (upper blot; 10 wk postinjection) and ab14220 (lower blot; 3 wk postinjection) after T cell injection (→) into recipients (A). Expanded versions of these WBs are shown in SI Appendix, Fig. S2 A and B. Forebrain Aβ1-40 ELISA in B6.Foxn1 recipients of PBS of wt-CD8 T cells 15 mo postinjection (B). 4G8-positive Plaques ± pTau/curcumin staining in brains of the above mice, and in 18-mo-old Tg2576 (AD-Tg) brain (C). ThioS staining of vasculature in the wt-CD8 group cortex (D), and 4G8 staining in the periventricular region (E) 6 mo postinjection. Representative plaque morphology and size in the wt-CD8 brain 15 mo postinjection (F). Forebrain pTau and Tau PHF WBs 10 wk postinjection (G), and compiled pTau and PHF values (H). Gallyas/silver-stained cells in hiT mouse groups 6 mo after i.v. control/cell injection, and in 14-mo-old Tg2576 (AD-Tg) mice (I). Hippocampal sections from the indicated groups (all B6.Foxn1 recipients, except AD-Tg = Tg2576 mice), were stained with 4G8 (Aβ) and curcumin 6 mo postinjection, or at 14 mo of age for AD-Tg (J). Right-facing arrows highlight Aβ deposits with no curcumin costaining. Up-facing arrows depict colocalized Aβ and curcumin deposits. Down-facing arrows depict curcumin+ structures with no Aβ costaining, i.e., nonamyloid fibrillar deposits. No DAPI was used; blue background is provided for anatomical context. ThioS staining of dentate gyrus in the same hiT mice (PBS and wt-CD8 group mice 6 mo after control/cell injection), and in 20-mo-old AD-Tg rats (K). Individual and overlaid images of sequential 4G8 (green)/pTau (red) → Gallyas (black) stains in the wt-CD8 group hippocampus 15 mo postinjection (L). Plots depict averages ± SEM. *P < 0.05, **P < 0.01, ***P < 0.005 by the two-sided T-test, relative to the PBS group.
Detergent-soluble phospho-tau (pTau) was slightly (30%) but significantly increased at 10 wk postinjection in the wt-CD8 group forebrain, while pTau paired helical filaments (PHFs, which mature to form NFTs in AD) were increased nearly fivefold on WBs (Fig. 1 G and H). Starting at 6 mo postinjection, fibril-staining reagents (Gallyas silver, curcumin, and ThioS), each stained cellular inclusions within the wt-CD8 group hippocampus (Fig. 1 IK and SI Appendix, Fig. S4A), with similar structures seen in simultaneously stained human AD cortex (SI Appendix, Fig. S4B), but not in brains of either AD-transgenic Tg2576 (AD-Tg) mice or AD-Tg rats (Fig. 1 IK) (22). Tissue immunofluorescence (IF) for pTau, Aβ, and DAPI prior to Gallyas staining revealed these structures were derived from pTau+ neurons with intact nuclei, and that silver staining was superimposable with that of pTau but not Aβ (Fig. 1L). These data suggest that the predominant T cells in hiT mice (which resemble resident-memory phenotype CD8 T cells or CD8 TRM) promote coordinated and robust deposition of Aβ and fibrillar NFT-like inclusions in the mouse brain.

Immune and Neuroinflammatory Infiltration.

Additional images support our previous findings that CD8 T cell infiltration, microgliosis, and astrogliosis (SI Appendix, Fig. S5 AC, D and E, and FI, respectively), are exclusively seen in wt-CD8 group mice (18). Cerebral plaques in these animals were closely associated with gliosis, as is common in human AD (SI Appendix, Fig. S5 H and I). In brain regions containing both T and glial cells, Aβ plaque burden correlated more strongly with hippocampal CD8 T cell levels than astrocytic or microglial levels (SI Appendix, Fig. S5 JL), suggesting a unique relationship between CD8 T cells and amyloid pathology.

Mechanisms of hiT Mouse Neuropathology.

To examine mechanisms of hiT cell mediated neuropathology, we injected PBS, or CD8 T cells from wild-type, Perforin 1-deficient, or IFNγ-deficient donors into young (6 to 8 wk) B6.Foxn1 hosts (PBS, wt-CD8, PrfKO-CD8, and IfnγKO-CD8 groups, respectively), and analyzed neuropathology by ELISA or WB, as well as by tissue staining. CD8 T cells from all donor strains expanded in circulation of B6.Foxn1 recipients (18). In marked contrast to wt-CD8 or IfnγKO-CD8, however, PrfKO-CD8 brains exhibited no significant increase in Aβ, plaques, pTau, PHFs, or NFT-like inclusions in any region. At 15 mo postinjection, Aβ1-40 was significantly elevated only in the wt-CD8 group brain by ELISA (Fig. 2A). Nevertheless, Aβ plaques and silver-stained neurons were evident within the entorhinal cortex and hippocampus by tissue IF in both wt-CD8 and IfnγKO-CD8 groups (Fig. 2B). Tau PHFs were also only elevated in wt-CD8 brains at 15 mo by WB (Fig. 2C), but Gallyas tissue staining of IfnγKO-CD8 brains similarly revealed NFT-like inclusions in the entorhinal cortex and hippocampus, but not in the cingulate cortex as in wt-CD8 group brains (Fig. 2D). Silver-stained neurons in IfnγKO-CD8 brain were derived from pTau+ neurons by sequential staining as shown for wt-CD8 brain (Fig. 2E). Aβ plaques in wt-CD8 and IfnγKO-CD8 group brains were of similar morphology (Fig. 2 F and G), but those in the IfnγKO-CD8 group were significantly smaller (Fig. 2 F and H) and appeared only in the entorhinal cortex and hippocampus but not in the cingulate cortex as in the wt-CD8 group (Fig. 2B). These data suggested that blocking Perforin production completely prevented all overt AD-like pathology, while blocking IFNγ production led to smaller Aβ plaques and to limited distribution of NFT-like structures reminiscent of early-stage AD (23).
Fig. 2.
Amyloid and Tau pathology in hiT mice with T cell functional inhibition. Compiled Aβ ELISA (A), 4G8 plaque burden (B) pTau and PHF signal (pS199/202 antibody [Invitrogen] used for pTau in WBs and tissue staining; Phospho-PHF-tau pSer202+Thr205 Antibody [AT8] used for Tau PHF in WBs) (C). Gallyas silver-stained cells in PBS, wt-CD8, PrfKO-CD8, and IfnγKO-CD8 hiT recipients 15 mo after control/cell injection and in 18-mo-old Tg2576 (AD-Tg) brain regions (D). Gallyas+ neurons from the IfnγKO-CD8 group cortex (E). Representative plaque morphology and size (F and G) in IfnγKO-CD8 brain 15 mo postinjection, and comparison to plaque size in the wt-CD8 group (H). Plots depict averages ± SEM. *P < 0.05, **P < 0.01, ***P < 0.005 by the two-sided T-test, relative to the PBS group.

Neuronal Loss and Cerebral Atrophy.

To determine whether neurodegeneration was evident in hiT mice, we stained and counted neurons positive for the neuron specific nuclear protein, NeuN, in CA1, CA2, and CA3 of the hippocampus. Brain mass was also assessed, and NeuN and synaptic protein signals quantified on WBs. Loss of NeuN+ cells in wt-CD8 group mice was visually apparent in hippocampal immunostains (Fig. 3 A and B), and was verified by NeuN+ cell counts at 15 mo postinjection (Fig. 3C). Loss of brain mass in the wt-CD8 group progressed from 5% at 6 mo, to 10% 15 mo postinjection (Fig. 3D), comparable to terminal brain atrophy in human AD (24). WBs of brains 15 mo postinjection confirmed proportional decreases in NeuN, the postsynaptic submembrane protein, drebrin, and the presynaptic vesicle protein, Synaptophysin (Fig. 3 E and F). Loss in brain mass correlated with decreased NeuN within PBS and CD8 injection groups, establishing a direct relationship between brain atrophy and neuronal loss (Fig. 3G). Significant brain loss was not observed in wt-CD4 group but was evident in wt-CD8+CD4 group, suggesting that CD4 T cells fail to either mediate or mitigate neuronal loss, or modulate its induction by CD8 T cells (SI Appendix, Fig. S6A).
Fig. 3.
Neurodegeneration and cognition in nude mice harboring hiT cells. Cell/control recipients in all panels are B6.Foxn1 exclusively. NeuN and GFAP staining (A and B), and cell counts in the hippocampus, 15 mo after cell/control injection (C). Brain atrophy over time in PBS and wt-CD8 groups (mass normalized to PBS controls at each time point; D). Representative forebrain westerns (E), and GAPDH-normalized NeuN, drebrin, and synaptophysin (SYNP) western signals (F). Correlation of NeuN with brain weight (G). Plots depict averages ± SEM. *P < 0.05, **P < 0.01, ***P < 0.005 by the two-sided T-test, relative to the PBS group. Representative open-field test at 13 mo (H). Fear conditioning (FC) over time (I), and spontaneous alternation behavior (SAB) at 12 mo (J). Barnes maze (BM) learning/training (K), retention (L), and reversal (M and N) phases, at 14 mo (black, colored symbols = P relative to PBS, wt-CD8, respectively). Plots depict averages ± SEM. *P < 0.05, **P < 0.01, ***P < 0.005 by two-sided ANOVA (panel K) or two-sided T-test (all others), relative to the PBS group unless otherwise indicated.

Severe Cognitive Impairment.

We assessed cognitive performance in hiT mice using three independent tests for learning and memory: contextual and cued FC, SAB in the Y-maze, and BM performance. Open-field testing (OFT) was performed prior to each of these three behavioral tests to rule out influences due to injection-associated mobility variations (Fig. 3H and SI Appendix, Fig. S7A), as well as to monitor motor dysfunction evident in other T cell-associated neurological conditions such as multiple sclerosis.
Significant motor deficits occurred with age but did not distinguish any groups including the PBS group from others (SI Appendix, Fig. S7A). In contrast, contextual FC was reduced in the wt-CD8 group relative to PBS controls 6 mo after T cell injection, with both contextual and cued learning impaired at 11 mo (Fig. 3I). These results suggest that CD8 T cells in hiT mice alone mediate early damage to the hippocampus (required for contextual FC), with additional later damage to the amygdala (involved in cued FC). This progressive behavioral deficit pattern mimics one typical of cognitive decline in human AD (25). Intriguingly, only cued learning was impaired in wt-CD8+CD4 but not wt-CD4 group mice at the earlier time point (SI Appendix, Fig. S6B), suggesting that CD4 T cells modulate whether the hippocampus or amygdala is damaged first in hiT mice, but neither induce cognitive decline themselves nor alter the ability of CD8 T cells to do so.
SAB in the Y-maze is a well-established assay for hippocampal integrity (26, 27), based on the preference of mice to alternately explore two alleys. The lowest possible score (50%) indicates random alley choice, due to either no working memory of the previous alley entered, or complete lack of preference. SAB testing 12 mo postinjection revealed a score of 55 to 56% in PBS controls, comparable to wild-type mice (28), but was reduced to 50% in the wt-CD8 group (Fig. 3J). This is consistent with an absence of working memory in the wt-CD8 group.
BM testing was performed at 14 mo postinjection and represents a more focused measure of hippocampus-dependent learning and memory. This test is not confounded by decreased ability to swim, or stress-induced behaviors caused by swim tests (29). Relative to PBS controls, wt-CD8 mice showed no ability to learn the maze over the initial 4-d training period (Fig. 3K). Given this initial learning deficit, it was not surprising that wt-CD8 mice were also profoundly impaired in subsequent memory retention and reversal phases of the maze (Fig. 3 LN).
Contextual FC at 6 and 11 mo correlated with brain mass (SI Appendix, Fig. S7B), as did latency to solve the BM (SI Appendix, Fig. S7C), further underscoring the relationship of cognitive decline to physical neurodegeneration. Poor performance of hiT mice on BM (total latency below median = BMlo) also exhibited significant association with increased pTau PHFs exclusively (SI Appendix, Fig. S7D), but not with any form of Aβ or detergent-soluble pTau (SI Appendix, Fig. S7 E and F).
Taken together, these three independent tests suggest that fully functional CD8 T cells in hiT mice mediate severe, progressive impairment of hippocampus-dependent learning and memory independent of locomotor activity, and that both the progressive pattern of cognitive loss and its association with cerebral pathology in the hiT model mirrors features typical of clinical AD (30).
Neuronal and cognitive loss differences were also evident between wt-CD8 and KO-CD8 groups: Neither IfnγKO-CD8 nor PrfKO-CD8 brains showed significant evidence of neuronal loss, and both were indistinguishable from PBS controls in level of spontaneous alternation (Fig. 3J). IfnγKO-CD8 mice, however, appeared initially similar to PBS controls in the training sessions of the BM, but became more similar to the impaired wt-CD8 group mice at many of the later stages (Fig. 3 KN). This suggests that both IFNγ and PRF1 deficiency eliminates robust neuronal loss and either subdues cognitive decline (in IfnγKO-CD8 group) or eliminates it altogether (in PrfKO-CD8 group).

Reduction of Doublecortin (DCX)+ Neurons in hiT Mice.

The dramatic impact of PRF1 deficiency on all aspects of AD-like neurodegeneration is most consistent with a direct role for lytic elimination of neural cells by CD8 T cells in the hiT mouse model. CD8 T cells normally reside in the brain, including those of C57BL/6 mice (31). These cells require expression of MHC I on their targets to eliminate them in an antigen-specific manner. Most neuronal lineage cells are MHC I-negative unless induced by cytokines such as IFNγ, but catecholaminergic neurons within the locus coeruleus (32) and neural progenitors constitutively express MHC I (33). To determine whether these cells were eliminated early and independent of IFNγ, we examined Doublecortin(DCX)-positive neural progenitors within the subventricular zone (SVZ) of mature (6-mo-old) wild-type C57BL/6 mice, as well as in wt-CD8 and IfnγKO-CD8 group hiT mice 15 mo postinjection. CD8 T cells were observed in close proximity to DCX+ neural progenitors in the SVZ rostral migratory stream of wild-type C57BL/6 mice, consistent with the possibility that CD8 T cells might directly eliminate these cells under disease conditions (SI Appendix, Fig. S8A). Accordingly, DCX+ neurons were dramatically decreased in SVZ and throughout the brain in wt-CD8 and IfnγKO-CD8 group hiT mice (SI Appendix, Fig. S8 B and C). Thus, even in the absence of more widespread neuronal loss (as in the IfnγKO-CD8 group), CD8 T cells in hiT mice promoted DCX+ neural progenitor elimination. This suggests that neuronal lineage cells that constitutively express MHC I are eliminated first by hiT cells.

RNAseq Analysis on Brains of hiT Mice.

To gain further mechanistic insights into how CD8 T cells in hiT mice promote AD-like pathology, we performed RNAseq analysis on mouse forebrains from experimental and control groups. Focusing on changes ≥10% relative to PBS, each of wt-CD8, IfnγKO-CD8, and PrfKO-CD8 groups exhibited significant upregulation of the T cell-specific marker, CD3ε, while they also altered 1,947, 309, and 1,901 additional transcripts, respectively (SI Appendix, Fig. S9 AJ). Since upregulation of CD3ε was at odds with our previous observation that CD8 T cells were undetectable in PrfKO-CD8 group brain by tissue staining (18), we reexamined T cells in this group using potentially more sensitive flow cytometric analysis (SI Appendix, Fig. S10). We confirmed significant accumulation of CD8 T cells in PrfKO-CD8 group brain relative to control B6.Foxn1, albeit lower than in wild-type C57BL/6 controls previously reported as comparable to the wt-CD8 group (SI Appendix, Fig. S9 A and B) (18). Thus, all groups injected with CD8 T cells harbored elevated levels of those cells in the brain, consistent with their CD8ε mRNA upregulation. This provides relevant context for further gene expression analysis.
Two gene expression pathways were prominently altered in the wt-CD8 group exclusively: BioPlanet AD and a group of largely overlapping pathways related to cytoplasmic ribosomal proteins and translation (SI Appendix, Figs. S9H and S11 AC). In addition, a group of largely overlapping pathways related to electron transport chain and oxidative phosphorylation was up-regulated in both wt-CD8 and IfnγKO-CD8 but not PrfKO-CD8 brain (SI Appendix, Figs. S9H and S11 AC), and as such was uniquely associated with both delayed/arrested and later AD-like neuropathology. Electron transport chain- and ribosomal protein-related pathways are known to be involved in CD8 T cell-mediated effector activity as well as AD pathophysiology, and IFNγ is also known to modulate ribosomal proteins (34). Within cell type-specific genes, only neuronal genes exhibited net upregulation, whereas most nonneuronal genes were down-regulated (Fig. 4A). Moreover, multiple pathways implicated in AD were affected in the wt-CD8 group (SI Appendix, Figs. S9 HJ and S12). To further determine relevance of these modest changes to AD, we focused on 82 genes identified in prior genome-wide epidemiological studies (“GWAS genes”) (35) whose AD-associated variants often manifest as subtle expression changes. Among this GWAS subset, 14, 2, and 11 genes were significantly altered in wt-CD8, IfnγKO-CD8, and PrfKO-CD8 groups, respectively, with 8 and 1 of these also altered in wt-CD8 and IfnγKO-CD8 groups respectively, relative to PrfKO-CD8 (P = 0.034; two-sided Fisher’s exact test; Fig. 4B). Known functions of GWAS genes altered in wt-CD8 and PrfKO-CD8 brains included multiple biological pathways implicated in AD, including Aβ/Tau pathology gene expression and cell signaling, endocytosis, inflammation/immunity, and lipid/cholesterol metabolism (SI Appendix, Fig. S9 H and J), whereas those altered in IfnγKO-CD8 brains covered a more restricted subset involved in Aβ/Tau pathology and inflammation/immunity. This supports a model in which Perforin controls a critical disease-initiating event reflected by electron transport chain gene modulation, whereas IFNγ modulates most other gene expression risk factors that may worsen disease pace, assure disease progression, and/or increase disease severity (Fig. 4C).
Fig. 4.
Gene expression changes in the hiT mouse brain. RNAseq analysis on hiT and control forebrains. Most cell type-specific genes were down-regulated with the exception of neuron-specific genes (A). Differentially regulated genes among 84 AD-associated loci from GWAS; bolded black font depicts genes uniquely regulated in each group (B). Mechanistic model of gene, pathway, and disease induction by hiT cells based on combined pathological, knockout, and gene expression analysis (C). Abbreviations: CD8, cluster of differentiation antigen 8; Peptide Ag, peptide antigen; APP, Amyloid Precursor Protein; MHC I (HLA), major histocompatibility complex class I protein (Human Leukocyte Antigen); PRF1, Perforin 1 protein; IFNγ, Interferon-gamma (proinflammatory) cytokine; GZMB, Granzyme B (lytic) protein.

hiT Cell Metrics in Human AD.

Given the varied impact of CD8 T cells on different rodent models of hereditary neurodegeneration, it was imperative that the potential impact of hiT analogues was examined in human patients. We first examined levels of the larger “parental” memory/aging (KLRG1+) CD8 population in blood, and then the APP-specific KLRG1+ CD8 subpopulation derived from it, by flow cytometry in HLA-A2+ individuals from two separate cohorts (Fig. 5 AF, with gating parameters in SI Appendix, Figs. S13 and S15A). As expected, the parental KLRG1+ CD8 population increased with age while KLRG1- CD8 cells did not (SI Appendix, Fig. S14). The KLRG1+ subpopulation also exhibited slight expansion in age-related cognitive decline or MCI-AD (Fig. 5 B and D; and SI Appendix, Fig. S15 B and C, with representative gating shown in SI Appendix, Fig. S15A). By contrast, APP-specific KLRG1+ CD8 T cells were markedly decreased in MCI and/or AD (Fig. 5 C and D), and this trend was maintained in both females and males (SI Appendix, Fig. S16 A and B). The decrease in APP-specific KLRG1+ CD8 T cells correlated with poor cognitive performance in both cohorts independent of age (Fig. 5 E and F), exhibiting a linear correlation in the cohort subjected to the more sensitive MoCA test (Fig. 5E).
Fig. 5.
hiT parameters in human Alzheimer’s. Patient cohorts: University of Antwerp = “UA”; Cedars-Sinai Medical Center = “CSMC”; University of California, Davis = “UCD”; West Los Angeles Veteran’s Administration Hospital = “WLAVA” (A). KLRG1+ (B) and APP(471–479)/HLA-A2-reactive KLRG1+ (C) CD8 T cells in CTRL, MCI ± CSF AD biomarkers (MCI, MCI–AD), and verified Alzheimer’s (AD) blood. T cell subpopulations vs. MoCA score (D), and correlation of APP(471–479)/HLA-A2-reactive KLRG1+ CD8 with Mini Mental State Exam (MMSE) score and age (with negative pHLA multimer staining subtracted; significance of both APP-specific T cells and age with MMSE score was increased without such subtraction, with P = 0.01 and 0.02, respectively) (E and F). PRF1 WB and IF representative examples of PRF1 staining in presumptive vesicles with or without (arrows) costained Aβ (G), with compiled IF and WB quantifications in age-matched CTRL and AD brains (H). Representative APP(471-479)/HLA-A2-reactive CD8 staining (I) and compiled quantification (J) in the brain. Plots depict averages ± SEM. *P < 0.05, **P < 0.01, ***P < 0.005, ****P < 0.001 by the two-sided T-test, relative to CTRL unless otherwise indicated.
Cytolytic and antigen-specific T cell markers in AD brains were next examined (Fig. 5 A and GJ). Upregulation of Perforin1 in the AD brain was evident in two independent patient cohorts, corresponded with CD8 upregulation on WBs, and exhibited the expected punctate (endocytic) cellular pattern by tissue IF (Fig. 5 G and H). Similarly, T cells stained by CD8 and APP-specific pHLA multimers were more prevalent by tissue IF in the AD brain from the UCD cohort, despite no prescreening of specimens for HLA-A2 to which the APP-specific multimer binds (Fig. 5 I and J). Taken together, these data suggest that parental KLRG1+ CD8 T cells accumulate with aging in blood. By contrast, APP-specific KLRG1+ CD8 T cells in blood decrease in proportion to cognitive decline in AD and prodromal conditions, just as they accumulate in the AD brain.
Decreased APP-specific KLRG1+ CD8 T cells in blood correlated significantly with reduced Aβ1-42 and increased total Tau (but only marginally with p-tau181) in CSF within the largest patient cohort (UA; Fig. 6A), suggesting correspondence with existing AD biomarkers. Levels of parental KLRG1+ CD8 T cells also correlated uniquely with CSF Aβ1-42 in AD patients independent of age (Fig. 6B). While neither parental nor APP-specific CD8 cells correlated linearly with MMSE (as with MoCA) (Fig. 6 A and B), the latter did exhibit a correlation with hi/lo MMSE score (Fig. 5F). The lower sensitivity of MMSE vs. MoCA in detecting cognitive impairment may account for this difference (36). Overall, these trends suggested association of age-related CD8 T cell dynamics with some of the most specific established AD biomarkers, hinting at potential AD biomarker utility for both parental CD8 TRM and APP-specific subpopulations.
Fig. 6.
CD8 TRM biomarker potential in human Alzheimer’s. Correlation of APP(471–479)/HLA-A2-reactive KLRG1+ CD8 levels with Aβ1-42, total-tau, and P-tau181 in CSF, and with MMSE score in all AU patients combined (A), and correlation of parental KLRG+CD8+ T cells with these parameters in AU Alzheimer’s patients only (note: several higher APP-specific T cell levels were in control patients that were not subjected to MMSE testing) (B). Combined variance of TRM markers, CD8A, CD44, and CD103 yielded the indicated areas under the curve (AUC) in Receiver Operating Characteristic (ROC) plots of T cell-high and T cell-low samples from a publicly available patient dataset (C). AUC was not significantly altered when CD103 variance was used alone in this cohort (0.781, T cell-high; 0.548, T cell-low). ROC plots of APP(471–479)/HLA-A2 multimer-reactive KLRG1+ CD8 T cells in blood relative to normal aging controls, both from UA cohort (D). Mild Cognitive Impairment without (MCI-normal bio) and with (MCI-AD bio) CSF biomarkers consistent with AD, and confirmed AD patients ages 57 to 84 (AD-all). AD-age-matched indicates ROC analysis on 10 AD patients for whom precisely age-matched controls were available (±1 y; n = 10). P < 0.001 for all curves except MCI – normal bio (P = 0.003).
To test this, we first examined publicly accessible data for multiple CD8 TRM markers in blood from independent patient cohorts (CD3D/E, TCRZ, CD8A/CD8B, CD103, CD122, CD127, CD44, as available in database). Using GFAP as an up-regulated standard (SI Appendix, Fig. S17 A and B), or simply compared to normal aging controls (SI Appendix, Fig. S17 C and D), we verified significant upregulation of multiple CD8 TRM markers at various stages of AD in two independent cohorts, with CD8 and TCRZ genes up-regulated at the earliest stage of AD in a severity-graded cohort (SI Appendix, Fig. S17B). We separated the largest of these cohorts into T cell high and low groups based on median CD3 expression to exclude patients with age-related T cell lymphopenia, and excluded those under 65 to eliminate potentially confounding early-onset AD patients and young controls (SI Appendix, Fig. S6D). Comodulation of CD103, CD44, and CD8A as a group, as well as levels of CD103 alone, exhibited intriguing biomarker potential in ROC plots (Fig. 6C). In addition, APP-specific KLRG1+ CD8 T cell levels quantified by flow cytometry exhibited even better biomarker potential than CD8 TRM genes (Fig. 6D), corresponding better to MCI-AD than the highly touted plasma biomarker, pTau-217 (SI Appendix, Fig. S17 A–C), corresponded to AD itself (37). This demonstrates that the APP-specific CD8 T cell subpopulation and its parental KLRG1+ population are uniquely modulated in AD, and suggests that their levels may be useful and potentially superior disease biomarkers.

Discussion

The hiT mouse model demonstrated several key features reminiscent of AD in humans. These included coupled accumulation of amyloid plaques and NFT-like fibrillar inclusions in tau+ neurons, as well as robust neuronal loss and profound cognitive decline, each demonstrated by at least three separate methodologies. This combination of AD-like features has not previously been induced in animal models by a single factor or transgene, as rodents typically fail to develop either NFT-like inclusions or robust neuronal loss without the addition of transgenes unrelated to human AD (1, 38, 39). hiT mice also exhibited finer features often seen in human AD, including cognitive loss progressing from hippocampus- to amygdala-dependent tasks over time (25), progressive brain atrophy related to neuronal loss, unique association of cognitive loss with fibrillar tau pathology (30), and modulation of multiple risk-associated genes.
Mechanistically, Perforin 1 in CD8 T cells was required for all aspects of neurodegenerative pathology, with the exception of most gene expression alterations. While this potentially involved the target-lysis function of Perforin in T cells, we’d previously reported that Perforin also precluded the accumulation of T cells in brains of hiT mice (18), a finding inconsistent with the minimal impact on gene expression Perforin knockout conferred on hiT mouse brains. Follow-up analysis showed that Prf1KO CD8 T cells did in fact accumulate and remain in hiT brains long term, but at lower levels than wild-type CD8 T cells. While this offers an explanation for the changes in brain gene expression seen in Prf1KO-CD8 group mice, it does not discount the possibility that lower levels of brain T cells could be a contributing factor in the lack of neuropathology observed in this group, particularly given that Perforin deficiency has been implicated in clinical disorders of T cell tissue homeostasis previously (40).
By contrast to Perforin 1, IFNγ in CD8 T cells promoted widespread neuronal loss and cognitive decline in hiT mice and increased the distribution and size of amyloid plaques. This parallels the impact of this cytokine on mouse models of amyloidosis (41). On a molecular level, Perforin1 is known to promote mitochondrial ROS and oxidative stress (very early events in AD) by facilitating Granzyme-mediated disruption of the electron transport chain in target cells (42, 43). Perforin 1 also promotes internalization of Aβ in neurons (44). IFNγ on the other hand is a well-known proinflammatory effector, as well as a potent modulator of gene expression in cells expressing its receptor (45). The distinct involvement of these two genes is thus consistent with Perforin-mediated attack of neurons by hiT cells, which simultaneously initiates limited neurodegeneration, mitochondrial oxidative stress, and inflammatory cytokine production in the brain. IFNγ accelerates progression of the neuropathology, likely through induction of MHC I and modulation of multiple risk-associated genes. CD4 T cells appear to play a dispensable role in induction and progression of neuropathology in hiT mice, but appear to subtly influence the brain region damaged by CD8 hiT cells, to the extent that it resembles a distinct AD behavioral subtype (46). While intriguing, rigorous validation of this working molecular mechanism requires further study.
The apparent arrest of IfnγKO-CD8 group mice at a stage resembling early AD made them attractive to examine potential targets of T cell elimination in the absence of more widespread neuronal loss. CD8 T cells require MHC I on target cells to recognize and destroy them, but only small anatomically restricted subpopulations of neuronal lineage cells constitutively express MHC I without induction by cytokines such as IFNγ. Such cells include catecholaminergic neurons within locus coeruleus (32), among the earliest sites affected by AD neuropathology (47). Neuroblasts mediate adult neurogenesis, a process thought to be critically impaired very early in AD, and also constitutively express MHC I (33, 48). The elimination of Doublecortin (DCX)+ neuroblasts by CD8 T cells in the IfnγKO-CD8 group suggests that neuronal lineage cells constitutively expressing MHC I are initial targets of CD8 T cell elimination in the hiT model, while more widespread neurodegeneration requires production of IFNγ, a known inducer of MHC I on distinct neural cells (45).
While several aspects of the hi T model paralleled features of human AD, it is not a perfect phenocopy of the disease. For example, Aβ1–40 and diffuse plaques predominate in hiT mice, whereas Aβ1–42 and compact plaques typically predominate in most forms of human AD. In addition, “ghost” tangles—remnants of NFTs in dead neurons—were not seen in hiT mice. Both these differences may well be explained by hiT mouse pathology occurring in the context of the endogenous mouse rather than transgenic human Aβ- and/or tau-encoding genes. Rodents, for example, are known to be deficient in clearing endogenous Aβ1–42, and exhibit Aβ1–40 preference in amyloid fibril formation (49, 50), which could favor diffuse plaque and Aβ1–40 predominance in hiT mice. Similarly, the absence of ghost tangles is likely due to the virtual absence of the ghost tangle-promoting isoforms of MAPT, the gene encoding tau proteins, in adult mice (51). In addition, the hiT model was analyzed exclusively in female mice due to concerns over male-specific autoimmune dynamics in related strains (19, 20). This rendered our finding that human APP-specific CD8 T cell levels are associated with AD in both males and females a critical observation.
Reports of rodents exhibiting neurodegeneration linked to amyloid and fibrillar tau pathology have been published previously, most notably upon infection of mice with Porphyromonas gingivalis (52). Introduction of multiple AD-associated human Aβ-related transgenes into rats (AD-Tg rats) has similarly been reported to produce neurodegeneration with NFT-like silver-stained neurons, although this model appears more relevant to dominantly inherited AD (22). Evidence of NFTs in both these models, however, was extremely limited, reliant on single staining reagents, and not statistically validated. For this reason, we sought to include one of these models as a control for NFT-like structures. In our hands, however, NFT-like structures failed to appear in the same AD-Tg rat model under staining procedures that readily revealed NFTs in hiT mice and human AD brains alike. Further, neither infection nor transgenic models have offered mechanistic explanations for anatomically specific disease etiology, generated pathophysiology corresponding to well-defined aspects of human AD progression, or demonstrated translational relevance more generally.
In addition to mirroring gross and fine aspects of human AD, hiT mice successfully predicted that CD8 TRM-marker genes and Perforin protein are up-regulated in the AD brain. Similarly, the model successfully predicted that APP-specific CD8 T cell levels would correspond to disease status in both females and males with AD and related MCI, as well as to cognitive decline in multiple cohorts. Finally, observations in hiT mice led to the prediction that levels of hiT cell analogues in blood would constitute uniquely useful tools for the clinical management of AD, with their utility for AD diagnosis particularly highlighted in this study.
While the hiT mouse model is perhaps not the first to replicate key aspects of AD via an inductive factor upstream of Aβ (39), it does so without the use of transgenes not associated with the human disease. More importantly, it exhibits clear translational relevance to sporadic AD. As such, it helps to dispel some of the confusion that surrounds the role of T cells in Alzheimer’s and related neurodegenerative diseases. Critically, the hiT model reproduced compelling AD-like characteristics through introduction of age-related T cells and without deterministic transgenes. Functional impairment of the T cells attenuated AD-like features, and their analogues were uniquely associated with human AD. Thus, three critical criteria for causation (disease recapitulation by agent introduction into animals, disease mitigation upon impairment of agent, and preferential association of agent dynamics to human disease) appear satisfied by this model. Nevertheless, the exclusive use of female nude mice qualifies such a conclusion and opens the possibility that additional factors associated with gender and/or mouse strain genetics could also impact neurodegenerative induction. Similarly, human AD may in fact possess multiple distinct inductive events or processes. Hence, it will be critical for future studies to discern the extent to which hiT cells and related events contribute to AD induction and in what proportion of patients they do so.
Conservatively, however, our study reveals evidence for the existence of a discrete and clinically useful factor upstream of Aβ that may contribute to amyloid- and tau-associated neurodegeneration. Further assessment of the potential of hiT cell metrics as diagnostic biomarkers for human AD nevertheless requires multicenter validation, prospective and longitudinal analyses that include more extensive non-AD dementia cohorts. It will be equally important to determine whether modulation of antigen-specific hiT cell presence or function can alter the course of AD-like neurodegeneration in therapeutic studies and determine whether effective treatments for AD critically modulate these cells in humans. Finally, continued examination of hiT mice on multiple strain backgrounds, and harboring risk factors for AD or other age-related disorders, may lead to a more comprehensive understanding of age-related disease biology, pathology, and immune characteristics more generally.

Materials and Methods

Animal Subjects.

Female C57BL/6, B6.Foxn1 mice, and congenic and/or syngeneic knockout strains were housed in a pathogen–free vivarium under standard conditions on a 12-h light/12-h dark cycle. Recipient animals were 8- to 10-wk-old female B6.Foxn1 mice; donors were 5- to 8-wk-old female C57BL/6 or B6.CD45.1 congenic mice. Numbers of animals used per test are specified in SI Appendix, Table S1.

Adoptive Transfer of T Cells.

Splenic CD8+ T cells from C57BL/6J female donors (5 to 8 wk old) were purified using anti-CD8 or anti-CD4 immunobeads (Miltenyi Biotech, Sunnyvale, CA). 3 × 106 purified CD8 or CD4 T cells, or 3 × 106 native splenocytes for CD8+CD4 T cell hosts, were intravenously injected in 50 µL of PBS into female B6.Foxn1 nude hosts. Cell numbers were based on prior publications demonstrating homeostatic expansion at similar doses (18, 53, 54). Transfer efficiency into B6.Foxn1 hosts was validated by persistence of ≥5% CD8+, CD4+, or both, T cells within splenic lymphocytes 3 wk after injection (18).

Tissue Processing.

The brain and spleen were harvested from mice and perfused with saline under deep anesthesia using a ketamine and xylazine (40 to 50 mg/kg i.p.) cocktail. Whole brains were weighed after removal of the cerebellum, brainstem, and olfactory bulb. Right hemispheres were flash frozen at −80 °C for protein studies, and homogenized. Cell lysates were separated into detergent-soluble and detergent-insoluble fractions. Left hemispheres were immersion fixed in 4% paraformaldehyde for 24 to 48 h and used for immunohistochemical staining.

Antibodies for Tissue Staining and WB Analyses.

Brain sections were mounted on slides and incubated at 4 °C overnight with primary antibody in blocking buffer, rinsed, and incubated 90 min in fluorochrome- or biotin-conjugated secondary antibody, with or without curcumin, or with ThioS alone. Sections were mounted with DAPI (Invitrogen) unless otherwise indicated, and images analyzed with ImageJ (NIH). Antibodies used: anti-Aβ/APP antibody (ab14220; clone 4G8\); Anti-pTau pS199/202 antibody (Invitrogen) and AT8; β-actin (clone AC-74, Sigma) due to cross-reactivity of GAPDH with IgG H chain (55); GAPDH used as control for all other markers; anti-GFAP (Dako); anti-NeuN antibody (Chemicon); anti-Iba1 (Wako, Ltd.); anti-CD8 (clone 53-6.72, BD Pharmingen); anti-doublecortin (DCX; polyclonal sc-8066, Santa Cruz Biotechnology); secondary antibodies (HRP, Alexa Flour-488, -594, -647; Invitrogen). control HLA-A2 and custom APP epitope dextramers (predicted affinity < 100 nM per NetMHC version 3.4 [APP(471–479)/HLA-A2], were manufactured by Immudex.

WB for Amyloid, Tau, Neural, and Immunological Markers.

Triton-soluble cell lysates were electrophoresed and blotted onto 0.2 µm nitrocellulose. Membranes were blocked and incubated in primary and secondary antibodies 1 h at room temperature with ≥3 washes between each, developed with ECL (GE Healthcare Biosciences; Pittsburgh, PA), and exposed onto Hyperfilm. WBs were performed a minimum of three times with comparable results per antibody. Bands were quantified using ImageJ software and expressed as (% area of test band)/(average % area of control bands).

ELISA.

Supernatant from homogenized brain was used for Triton-soluble Aβ. Insoluble pellets from Triton-homogenized brain were resuspended in 10 volumes 5M guanidine HCl 4 h to generate guanidine-soluble Aβ. Triton- and guanidine-soluble samples were subjected to analysis using species- and isoform-specific antibodies in Soluble and Insoluble Aβ ELISA kits (Invitrogen, Life Technologies). Absorbance was read on a SPECTRAmax Plus384 microplate reader (Molecular Devices, Sunnyvale, CA) and data analyzed in Graphpad PRISM. ELISAs were carried out a minimum of three times with comparable results per antibody/kit, with samples tested in triplicate and standard curves included on each plate.

Flow Cytometry.

Lymphocytes stained with respective antibodies were analyzed by three-color flow cytometry. Flow cytometry was performed with CD8-APC, HLA-A2-PerCP-Cy5.5, KLRG1-FITC antibodies (all Biolegend), and MHC A*0201-PE (Immudex) dextramers on a FACSCanto flow cytometer (BD Biosciences, San Jose, CA). Antibodies and multimers were incubated with whole-blood single-cell suspension in PBS with 2% FBS, on ice for 30 min, washed, and analyzed. FSC vs SSC was used to distinguish between lymphocytes and dead cells/debris, with exclusion of cell multiplets. 100,000 to 300,000 flow events were acquired and analyzed. FlowJo software was used for data analysis using identical gating (shown in SI Appendix, Figs. S13 and S15A).

Gallyas Silver Staining.

Free floating brain sections were placed in 5% periodic acid 3 min, washed 2×, and placed in silver iodide solution 1 min, followed by incubation in 0.5% acetic acid 5 min (2×), and rinsed with dH20. Sections were incubated in developer for ~10 min until sections were pale brown/gray, stopped in 0.5% acetic acid 5 min, rinsed in dH2O, and mounted. Stained sections were examined by microscopy. Stained neurons were counted from CA2, and proportions within total neurons visually quantified in triplicate from entorhinal and cingulate cortex.

Neuronal Counts.

Whole-number neuronal estimates were performed using the optical fractionator method (56) with stereological software (Stereo Investigator; MBF Bioscience). Paramedian sagittal serial sections spaced 50 µm apart were stained with NeuN. CA1, CA2, CA3, and other regions of interest were defined according to the Paxinos and Watson mouse brain atlas. A grid was placed randomly over the ROI, and cells were counted within three-dimensional optical dissectors (50 µm, 50 µm, 10 µm) using a 100× objective. Estimated totals weighted by section thickness were obtained with Stereo Investigator software, yielding a coefficient of error 0.10.

Behavioral Testing: General.

Testing order was randomized by alternating control and treatment group animal runs. Testing started at the same time (±1.5 h) for tests run over multiple days, with early and late times alternated for intergroup randomization. Testing was performed during the light phase exclusively. OFT was performed preceding all other behavioral tests to rule out motility effects.

BM test.

BM testing was performed a single time only, 14 mo post-cell or -control injection. Mice were assessed for their ability to learn the location of an escape box over the course of 9 d in the BM apparatus as previously described (57).

Y-maze SAB.

Mice were tested for SA a single time only, at 12 mo post-cell or -control injection. SAB was measured by individually placing animals in one arm of a symmetric Y-maze made of opaque black acrylic plastic, and the sequence of arm entries and total number of entries recorded over a period of 8 min.

Flinch-jump/FC tests.

Flinch-jump/FC freezing times were determined 6 and 11 mo postcell or postcontrol injection. The apparatus (Freeze Monitor™) was a Plexiglas box with a stainless-steel grid floor with acoustic stimulus unit on top of the box, and the box ringed with photo beams and computer-linked optical sensors. On day 1, mice were presented with foot-shock associated with an auditory tone. On day 2, contextual retrieval was determined by placing the mice into the same test box, but without either tone or foot shocks. On day 3, cue conditioning was assessed in an altered environment with delivery of the auditory tone. Freezing time was recorded each day.

RNAseq and Gene Expression Analysis.

rRNA was depleted with NEBNext® rRNA Depletion Kit v2 and libraries prepared. Data were analyzed by ROSALIND® (https://rosalind.bio/), with their HyperScale architecture (San Diego, CA). Database sources referenced for enrichment analysis included Interpro, NCBI, MSigDB, REACTOME, and WikiPathways. Enrichment was calculated relative to a set of background genes relevant for the experiment.

Human Subjects.

Cohort 1: 40 control individuals (CTRL); 52 MCI patients with an AD-characteristic CSF biomarker profile (MCI-AD); 36 MCI patients not displaying an AD-characteristic CSF biomarker profile (MCI); 50 sporadic AD patients with an AD-characteristic CSF biomarker profile (AD). CSF samples were collected at Middelheim General Hospital (Antwerp, Belgium) according to described protocols (58). Inclusion criteria for controls: 1) no neurological or psychiatric antecedents and 2) no organic disease involving the central nervous system upon extensive clinical examination. MCI patients were diagnosed applying Petersen’s diagnostic criteria (59) without dementia (60). AD dementia was clinically diagnosed according to NINCDS/ADRDA and IWG-2 criteria (61).
Cohort 2: Cognitive Testing. 29 self-referred memory clinic patients from Cedars-Sinai Department of Neurosurgery clinically diagnosed as normal (n = 6), MCI (n = 18), dementia (n = 3), or uncertain (n = 2; on basis of cognitive testing) and were administered Montreal Cognitive Assessment (MoCA) (n = 22). HLA-A2-negative patients determined by flow cytometry were excluded from analysis, as were patients with ≤2.5% CD8+ cells in lymphocyte gates, or ≥20% variation in any staining parameter between duplicate samples.
Cohort 3: Brain western and IHC. Hippocampal lysates from 13 autopsy-confirmed sporadic AD patients, and 5 age-matched normal controls were run on WBs using anti-CD8 or anti-PRF1. Hippocampal sections from 10 autopsy-confirmed sporadic AD patients, and 10 age-matched normal controls were stained with anti-CD8-fluorescein plus APP(471–479)/HLA-A2-PE. HLA-A2-negative samples were not excluded from western and IHC/IF analysis.

Statistical Analysis.

Power analyses for sample sizes and statistical methods are detailed in SI Appendix, Supplementary Test: Detailed Materials & Methods. Briefly, GraphPad Prism (version 5.0b; San Diego, CA) was used to analyze the data using appropriate statistical tests when data was normally or nonnormally distributed. Subject numbers and methods of reagent validation are shown in SI Appendix, Table S1, and all histograms depict average ± SEM.

Study Approval.

All animal procedures were approved prior to performance by the Cedars-Sinai Institutional Animal Care and Use Committee. The Cedars-Sinai Institutional Review Board designated the analysis of deidentified human brain specimens from UC Davis exempt from committee review. Brain specimens were collected, stored, and disseminated with prior approval by the UC Davis Medical Center Institutional Review Board. Sampling for cohort 1 was approved by the Medical Ethics Committee of the Hospital Network Antwerp (ZNA), Antwerp, Belgium.

Data, Materials, and Software Availability

RNAseq, Western blot quantitation, behavioral test data, flow cytometry data have been deposited in Open Science Framework (https://doi.org/10.17605/OSF.IO/354JS) (62). Model Organisms and/or the means to generate them will be made generally available for research (non-commercial) use.

Acknowledgments

We gratefully acknowledge the patients and families who provided tissue and blood for analysis; the Cedars-Sinai Research Institute Biobehavioral Core for conducting mouse behavioral tests; Dr. Igor Antoshechkin for RNAseq performance; Dr. Jeremy Sanders and Dr. Sol Katzman for guidance in gene expression analysis; Ms. Hannah Schubloom and Mia Oviatt for excellent administrative support and editing; and Dr. Kristina Trujillo for editing and proofreading. NIH Grant (UC Davis Alzheimer’s Disease Center) P30AG10129 (L.-W.J.); NIH Grant R21NSO54162 (C.J.W.); NIH Grant R21AG033394 (R.M.C.); Cedars-Sinai Medical Center Biobehavioral Core (R.N.P.); Joseph Drown Foundation (C.J.W.); Maxine Dunitz Neurosurgical Institute (C.J.W.); and Maxine Dunitz Neurosurgical Institute (D.K.I.).

Author contributions

R.C., R.N.P., J.A.R., D.K.I., B.A.W., and C.J.W. designed research; A.P., A.R., M.J., R.M.C., R.C., N.G., R.N.P., G.D., A.M., D.G., L.-W.J., D.V.D., Y.V., H.D.R., P.P.D.D., D.K.I., B.A.W., and C.J.W. performed research; A.R., R.C., R.N.P., L.-W.J., Y.V., H.D.R., P.P.D.D., K.L.B., D.K.I., B.A.W., and C.J.W. contributed new reagents/analytic tools; A.P., A.R., M.J., R.M.C., R.C., N.G., R.N.P., G.D., A.M., H.S., D.V.D., Y.V., H.D.R., P.P.D.D., D.K.I., B.A.W., and C.J.W. analyzed data; R.N.P., L.-W.J., and P.P.D.D. advised on paper framing & design; J.A.R. facilitated collaborations, advised on paper framing & design; K.L.B. and C.J.W. facilitated collaborations, supervised facilities and personnel; and A.P., A.R., R.C., and C.J.W. wrote the paper.

Competing interests

C.J.W. is the author of patents PCT/US2016/049598, WO2017/040594, and PCT/US2019/017879. R.C. and K.L.B. are co-authors on patent PCT/US2019/017879. PCT/US2016/049598 and WO 2017/040594 are licensed by Cedars-Sinai Medical Center to T-Neuro Pharma, Inc. C.J.W. has received salary and ownership interest in T-Neuro Pharma, Inc.

Supporting Information

Appendix 01 (PDF)

References

1
A. Codita, B. Winblad, A. H. Mohammed, Of mice and men: More neurobiology in dementia. Curr. Opin. Psychiatry 19, 555–563 (2006).
2
O. Wirths, S. Zampar, Neuron loss in Alzheimer’s disease: Translation in transgenic mouse models. Int. J. Mol. Sci. 21, 8144 (2020).
3
E. S. Musiek, D. A. Bennett, Aducanumab and the “post-amyloid” era of Alzheimer research? Neuron 109, 3045–3047 (2021).
4
G. Chételat et al., Amyloid imaging in cognitively normal individuals, at-risk populations and preclinical Alzheimer’s disease. Neuroimage Clin. 2, 356–365 (2013).
5
J. Yao et al., Mitochondrial bioenergetic deficit precedes Alzheimer’s pathology in female mouse model of Alzheimer’s disease. Proc. Natl. Acad. Sci. U.S.A. 106, 14670–14675 (2009).
6
J. F. Arboleda-Velasquez et al., Resistance to autosomal dominant Alzheimer’s disease in an APOE3 Christchurch homozygote: A case report. Nat. Med. 25, 1680–1683 (2019).
7
M. R. Wardell, S. O. Brennan, E. D. Janus, R. Fraser, R. W. Carrell, Apolipoprotein E2-Christchurch (136 Arg––Ser). New variant of human apolipoprotein E in a patient with type III hyperlipoproteinemia. J. Clin. Invest. 80, 483–490 (1987).
8
D. Gate et al., Clonally expanded CD8 T cells patrol the cerebrospinal fluid in Alzheimer’s disease. Nature 577, 399–404 (2020).
9
B. Altendorfer et al., Transcriptomic profiling identifies CD8(+) T cells in the brain of aged and Alzheimer’s disease transgenic mice as tissue-resident memory T cells. J. Immunol. 209, 1272–1285 (2022).
10
C. Laurent et al., Hippocampal T cell infiltration promotes neuroinflammation and cognitive decline in a mouse model of tauopathy. Brain 140, 184–200 (2017).
11
M. S. Unger et al., CD8(+) T-cells infiltrate Alzheimer’s disease brains and regulate neuronal- and synapse-related gene expression in APP-PS1 transgenic mice. Brain Behav. Immun. 89, 67–86 (2020).
12
N. Piehl et al., Cerebrospinal fluid immune dysregulation during healthy brain aging and cognitive impairment. Cell 185, 5028–5039.e5013 (2022).
13
G. K. Wilcock, M. M. Esiri, Plaques, tangles and dementia. A quantitative study. J. Neurol. Sci. 56, 343–356 (1982).
14
P. V. Arriagada, J. H. Growdon, E. T. Hedley-Whyte, B. T. Hyman, Neurofibrillary tangles but not senile plaques parallel duration and severity of Alzheimer’s disease. Neurology 42, 631–639 (1992).
15
X. Chen et al., Microglia-mediated T cell infiltration drives neurodegeneration in tauopathy. Nature 615, 668–677 (2023).
16
W. Su et al., CXCR6 orchestrates brain CD8(+) T cell residency and limits mouse Alzheimer’s disease pathology. Nat. Immunol. 24, 1735–1747 (2023).
17
C. J. Wheeler, Evolutionary selection of APOEɛ4 encourages increased focus on immunity in Alzheimer’s disease. J. Alzheimer’s Dis. 93, 919–921 (2023).
18
A. Panwar et al., Functional recreation of age-related CD8 T cells in young mice identifies drivers of aging- and human-specific tissue pathology. Mech. Ageing Dev. 191, 111351 (2020).
19
B. R. Lawson et al., The role of alpha beta+ T cells and homeostatic T cell proliferation in Y-chromosome-associated murine lupus. J. Immunol. 167, 2354–2360 (2001).
20
M. G. Pitts, D. Nardo, C. M. Isom, V. J. Venditto, Autoantibody responses to apolipoprotein A-I are not diet- or sex-linked in C57BL/6 mice. Immunohorizons 4, 455–463 (2020).
21
R. Schwab et al., Expanded CD4+ and CD8+ T cell clones in elderly humans. J. Immunol. 158, 4493–4499 (1997).
22
R. M. Cohen et al., A transgenic Alzheimer rat with plaques, tau pathology, behavioral impairment, oligomeric abeta, and frank neuronal loss. J. Neurosci. 33, 6245–6256 (2013).
23
C. Reitz, L. Honig, J. P. Vonsattel, M. X. Tang, R. Mayeux, Memory performance is related to amyloid and tau pathology in the hippocampus. J. Neurol. Neurosurg. Psychiatry 80, 715–721 (2009).
24
J. D. Sluimer et al., Whole-brain atrophy rate and cognitive decline: longitudinal MR study of memory clinic patients. Radiology 248, 590–598 (2008).
25
A. Serrano-Pozo, M. P. Frosch, E. Masliah, B. T. Hyman, Neuropathological alterations in Alzheimer disease. Cold Spring Harb. Perspect. Med. 1, a006189 (2011).
26
R. J. Douglas, in “The development of hippocampal function: implications for theory and therapy” in The Hippocampus: Neurophysiology and Behavior, R. L. Isaacson, K. H. Pribram, Eds. (Plenum, New York, NY, 1975), vol. 2, pp. 327–361.
27
R. J. Kirkby, A maturation factor in spontaneous alteration. Nature 215, 784 (1967).
28
Q. L. Ma et al., Beta-amyloid oligomers induce phosphorylation of tau and inactivation of insulin receptor substrate via c-Jun N-terminal kinase signaling: Suppression by omega-3 fatty acids and curcumin. J Neurosci. 29, 9078–9089 (2009).
29
M. M. Karabeg et al., 5-HTT deficiency affects neuroplasticity and increases stress sensitivity resulting in altered spatial learning performance in the Morris water maze but not in the Barnes maze. PloS ONE 8, e78238 (2013).
30
M. R. Brier et al., Tau and Abeta imaging, CSF measures, and cognition in Alzheimer’s disease. Sci. Translational Med. 8, 338ra366 (2016).
31
L. M. Wakim et al., The molecular signature of tissue resident memory CD8 T cells isolated from the brain. J. Immunol. 189, 3462–3471 (2012).
32
C. Cebrián et al., MHC-I expression renders catecholaminergic neurons susceptible to T-cell-mediated degeneration. Nat. Commun. 5, 3633 (2014).
33
K. Lin et al., MHC class I H2-Kb negatively regulates neural progenitor cell proliferation by inhibiting FGFR signaling. PLoS Biol 19, e3001311 (2021).
34
R. Mukhopadhyay et al., DAPK-ZIPK-L13a axis constitutes a negative-feedback module regulating inflammatory gene expression. Mol. Cell 32, 371–382 (2008).
35
C. Bellenguez et al., New insights into the genetic etiology of Alzheimer’s disease and related dementias. Nat. Genet. 54, 412–436 (2022).
36
N. Ciesielska et al., Is the Montreal Cognitive Assessment (MoCA) test better suited than the Mini-Mental State Examination (MMSE) in mild cognitive impairment (MCI) detection among people aged over 60? Meta-analysis. Psychiatr. Pol. 50, 1039–1052 (2016).
37
S. Palmqvist et al., Discriminative accuracy of plasma phospho-tau217 for Alzheimer disease vs other neurodegenerative disorders. JAMA 324, 772–781 (2020).
38
C. Schwab, M. Hosokawa, P. L. McGeer, Transgenic mice overexpressing amyloid beta protein are an incomplete model of Alzheimer disease. Exp. Neurol. 188, 52–64 (2004).
39
S. Capsoni et al., Alzheimer-like neurodegeneration in aged antinerve growth factor transgenic mice. Proc. Natl. Acad. Sci. U.S.A. 97, 6826–6831 (2000).
40
S. E. Stepp et al., Perforin gene defects in familial hemophagocytic lymphohistiocytosis. Science 286, 1957–1959 (1999).
41
T. C. Browne et al., IFN-gamma Production by amyloid beta-specific Th1 cells promotes microglial activation and increases plaque burden in a mouse model of Alzheimer’s disease. J. Immunol. 190, 2241–2251 (2013).
42
G. Jacquemin et al., Granzyme B-induced mitochondrial ROS are required for apoptosis. Cell Death Differ. 22, 862–874 (2015).
43
D. Martinvalet, ROS signaling during granzyme B-mediated apoptosis. Mol. Cell Oncol. 2, e992639 (2015).
44
E. Lana et al., Perforin promotes amyloid beta internalisation in neurons. Mol. Neurobiol. 54, 874–887 (2017).
45
D. Sredni-Kenigsbuch, TH1/TH2 cytokines in the central nervous system. Int. J. Neurosci. 112, 665–703 (2002).
46
D. Ferreira, A. Nordberg, E. Westman, Biological subtypes of Alzheimer disease: A systematic review and meta-analysis. Neurology 94, 436–448 (2020).
47
P. Theofilas, S. Dunlop, H. Heinsen, L. T. Grinberg, Turning on the light within: Subcortical nuclei of the isodentritic core and their role in Alzheimer’s disease pathogenesis. J. Alzheimer’s Dis. 46, 17–34 (2015).
48
S. H. Choi, R. E. Tanzi, Is Alzheimer’s disease a neurogenesis disorder? Cell stem. cell 25, 7–8 (2019).
49
W. A. Banks, S. M. Robinson, S. Verma, J. E. Morley, Efflux of human and mouse amyloid beta proteins 1–40 and 1–42 from brain: Impairment in a mouse model of Alzheimer’s disease. Neuroscience 121, 487–492 (2003).
50
R. W. Shin et al., Amyloid beta-protein (Abeta) 1–40 but not Abeta1-42 contributes to the experimental formation of Alzheimer disease amyloid fibrils in rat brain. J. Neurosci. 17, 8187–8193 (1997).
51
T. Uchihara, M. Hara, A. Nakamura, K. Hirokawa, Tangle evolution linked to differential 3- and 4-repeat tau isoform deposition: A double immunofluorolabeling study using two monoclonal antibodies. Histochem. Cell Biol. 137, 261–267 (2012).
52
V. Ilievski et al., Chronic oral application of a periodontal pathogen results in brain inflammation, neurodegeneration and amyloid beta production in wild type mice. PloS ONE 13, e0204941 (2018).
53
B. Rocha, N. Dautigny, P. Pereira, Peripheral T lymphocytes: Expansion potential and homeostatic regulation of pool sizes and CD4/CD8 ratios in vivo. Eur. J. Immunol. 19, 905–911 (1989).
54
B. Ernst, D. S. Lee, J. M. Chang, J. Sprent, C. D. Surh, The peptide ligands mediating positive selection in the thymus control T cell survival and homeostatic proliferation in the periphery. Immunity 11, 173–181 (1999).
55
F. R. Petry et al., Specificity of anti-tau antibodies when analyzing mice models of Alzheimer’s disease: Problems and solutions. PloS ONE 9, e94251 (2014).
56
M. J. West, L. Slomianka, H. J. G. Gundersen, Unbiased stereological estimation of the total number of neurons in the subdivisions of the rat hippocampus using the optical fractionator. Anat. Rec. 231, 482–497 (1991).
57
M. W. Pitts, Barnes maze procedure for spatial learning and memory in mice. Bio-Protocol 8, e2744 (2018).
58
S. Engelborghs et al., Diagnostic performance of a CSF-biomarker panel in autopsy-confirmed dementia. Neurobiol. Aging 29, 1143–1159 (2008).
59
R. C. Petersen, Mild cognitive impairment as a diagnostic entity. J. Inter. Med. 256, 183–194 (2004).
60
G. M. McKhann et al., The diagnosis of dementia due to Alzheimer’s disease: Recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimer’s Dementia 7, 263–269 (2011).
61
B. Dubois et al., Advancing research diagnostic criteria for Alzheimer’s disease: The IWG-2 criteria. Lancet Neurol. 13, 614–629 (2014).
62
A. Panwar et al., Panwar, Rentsendorj, Jhun et al., PNAS 2024. Open Science Framework. https://doi.org/10.17605/OSF.IO/354JS. Deposited 8 May 2024.

Information & Authors

Information

Published in

The cover image for PNAS Vol.121; No.29
Proceedings of the National Academy of Sciences
Vol. 121 | No. 29
July 16, 2024
PubMed: 38995966

Classifications

Data, Materials, and Software Availability

RNAseq, Western blot quantitation, behavioral test data, flow cytometry data have been deposited in Open Science Framework (https://doi.org/10.17605/OSF.IO/354JS) (62). Model Organisms and/or the means to generate them will be made generally available for research (non-commercial) use.

Submission history

Received: February 5, 2024
Accepted: May 23, 2024
Published online: July 12, 2024
Published in issue: July 16, 2024

Keywords

  1. Alzheimer’s disease
  2. T cell
  3. mouse model
  4. neuroscience
  5. biomarker

Acknowledgments

We gratefully acknowledge the patients and families who provided tissue and blood for analysis; the Cedars-Sinai Research Institute Biobehavioral Core for conducting mouse behavioral tests; Dr. Igor Antoshechkin for RNAseq performance; Dr. Jeremy Sanders and Dr. Sol Katzman for guidance in gene expression analysis; Ms. Hannah Schubloom and Mia Oviatt for excellent administrative support and editing; and Dr. Kristina Trujillo for editing and proofreading. NIH Grant (UC Davis Alzheimer’s Disease Center) P30AG10129 (L.-W.J.); NIH Grant R21NSO54162 (C.J.W.); NIH Grant R21AG033394 (R.M.C.); Cedars-Sinai Medical Center Biobehavioral Core (R.N.P.); Joseph Drown Foundation (C.J.W.); Maxine Dunitz Neurosurgical Institute (C.J.W.); and Maxine Dunitz Neurosurgical Institute (D.K.I.).
Author contributions
R.C., R.N.P., J.A.R., D.K.I., B.A.W., and C.J.W. designed research; A.P., A.R., M.J., R.M.C., R.C., N.G., R.N.P., G.D., A.M., D.G., L.-W.J., D.V.D., Y.V., H.D.R., P.P.D.D., D.K.I., B.A.W., and C.J.W. performed research; A.R., R.C., R.N.P., L.-W.J., Y.V., H.D.R., P.P.D.D., K.L.B., D.K.I., B.A.W., and C.J.W. contributed new reagents/analytic tools; A.P., A.R., M.J., R.M.C., R.C., N.G., R.N.P., G.D., A.M., H.S., D.V.D., Y.V., H.D.R., P.P.D.D., D.K.I., B.A.W., and C.J.W. analyzed data; R.N.P., L.-W.J., and P.P.D.D. advised on paper framing & design; J.A.R. facilitated collaborations, advised on paper framing & design; K.L.B. and C.J.W. facilitated collaborations, supervised facilities and personnel; and A.P., A.R., R.C., and C.J.W. wrote the paper.
Competing interests
C.J.W. is the author of patents PCT/US2016/049598, WO2017/040594, and PCT/US2019/017879. R.C. and K.L.B. are co-authors on patent PCT/US2019/017879. PCT/US2016/049598 and WO 2017/040594 are licensed by Cedars-Sinai Medical Center to T-Neuro Pharma, Inc. C.J.W. has received salary and ownership interest in T-Neuro Pharma, Inc.

Notes

This article is a PNAS Direct Submission.

Authors

Affiliations

Akanksha Panwar1
Department Neurosurgery, Maxine Dunitz Neurosurgical Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
Department Neurosurgery, Maxine Dunitz Neurosurgical Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
Michelle Jhun1
Department Neurosurgery, Maxine Dunitz Neurosurgical Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
Robert M. Cohen
Department Psychiatry & Behavioral Sciences and Neuroscience Program, Graduate Division of Biological and Biomedical Sciences (GDBBS), Emory University, Atlanta, GA 30322
Ryan Cordner
Department Neurosurgery, Maxine Dunitz Neurosurgical Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
Department Biomedical & Translational Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
Present address: Department of Microbiology & Molecular Biology, Brigham Young University, Provo, UT 84604.
Department Biomedical & Translational Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
Robert N. Pechnick
Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific Western University of Health Sciences, Pomona, CA 91766
Gretchen Duvall
Department Neurosurgery, Maxine Dunitz Neurosurgical Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
Armen Mardiros
Department Neurosurgery, Maxine Dunitz Neurosurgical Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
Present address: Department of Translational Science, A2 Biotherapeutics, Inc., Agoura Hills, CA 91301.
David Golchian
Department Neurosurgery, Maxine Dunitz Neurosurgical Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
Hannah Schubloom
Department Neurosurgery, Maxine Dunitz Neurosurgical Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
Lee-Way Jin
Department Medical Pathology and Laboratory Medicine, Laboratory Medicine, Medical Investigation of Neurodevelopmental Disorders (M.I.N.D.) Institute, University of California, Davis, Sacramento, CA 95817
Department of Biomedical Sciences, Institute Born-Bunge, Laboratory of Neurochemistry and Behavior, University of Antwerp, Antwerp 2610, Belgium
Department of Neurology and Alzheimer Research Center, University of Groningen and University Medical Center Groningen, Groningen AB 9700, Netherlands
Department of Biomedical Sciences, Institute Born-Bunge, Laboratory of Neurochemistry and Behavior, University of Antwerp, Antwerp 2610, Belgium
Faculty of Medicine & Health Sciences, Department of Translational Neurosciences, University of Antwerp, Antwerp 2610, Belgium
Division of Human Nutrition and Health, Chair Group of Nutritional Biology, Wageningen University & Research, Wageningen AA 6700, The Netherlands
Faculty of Medicine and Health Sciences, Vaccine and Infectious Disease Institute, Laboratory of Experimental Hematology, University of Antwerp, Antwerp 2610, Belgium
Peter Paul De Deyn
Department of Biomedical Sciences, Institute Born-Bunge, Laboratory of Neurochemistry and Behavior, University of Antwerp, Antwerp 2610, Belgium
Department of Neurology and Alzheimer Research Center, University of Groningen and University Medical Center Groningen, Groningen AB 9700, Netherlands
Department of Neurology, Memory Clinic of Hospital Network Antwerp, Middelheim and Hoge Beuken, Antwerp BE-2660, Belgium
Department of Chemistry & Biochemistry, University of California, Santa Cruz, CA 95064
Jevgenij A. Raskatov
Department of Chemistry & Biochemistry, University of California, Santa Cruz, CA 95064
Department Neurosurgery, Maxine Dunitz Neurosurgical Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
Department Neurosurgery, Maxine Dunitz Neurosurgical Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
NovAccess Global and StemVax LLC, Cleveland, OH 44023
Brian A. Williams
Transcriptome Function and Technology Program, Department of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA 91125
Department Neurosurgery, Maxine Dunitz Neurosurgical Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
Department of Chemistry & Biochemistry, University of California, Santa Cruz, CA 95064
NovAccess Global and StemVax LLC, Cleveland, OH 44023
Society for Brain Mapping & Therapeutics, World Brain Mapping Foundation, Pacific Palisades, CA 90272
T-Neuro Pharma, Inc., Albuquerque, NM 87123

Notes

4
To whom correspondence may be addressed. Email: [email protected].
1
A.P., A.R., and M.J. contributed equally to this work.

Metrics & Citations

Metrics

Note: The article usage is presented with a three- to four-day delay and will update daily once available. Due to ths delay, usage data will not appear immediately following publication. Citation information is sourced from Crossref Cited-by service.


Altmetrics




Citations

Export the article citation data by selecting a format from the list below and clicking Export.

Cited by

    Loading...

    View Options

    View options

    PDF format

    Download this article as a PDF file

    DOWNLOAD PDF

    Login options

    Check if you have access through your login credentials or your institution to get full access on this article.

    Personal login Institutional Login

    Recommend to a librarian

    Recommend PNAS to a Librarian

    Purchase options

    Purchase this article to access the full text.

    Single Article Purchase

    Antigen-specific age-related memory CD8 T cells induce and track Alzheimer’s-like neurodegeneration
    Proceedings of the National Academy of Sciences
    • Vol. 121
    • No. 29

    Figures

    Tables

    Media

    Share

    Share

    Share article link

    Share on social media