Abstract Immune mechanisms contribute to the neuropathology of Alzheimer’s disease (AD) but the role of adaptive immune cells is unclear. Here we show that the brain CD8^+ T cell compartment is dysregulated in AD patients and in the 3xTg-AD mouse model, accumulating activated CD103^– tissue-resident memory T cells that produce large amounts of granzyme K (GrK). These CD103^–CD8^+ T cells originate from the circulation and migrate into the brain using LFA-1 integrin. Ablation of brain CD103^–CD8^+ T cells in 3xTg-AD mice ameliorates cognitive decline and reduces neuropathology. GrK induces neuronal dysfunction and tau hyperphosphorylation in human and mouse cells via protease-activated receptor-1 (PAR-1), which is expressed at higher levels in the AD brain, revealing a key immune-mediated neurotoxic axis. We conclude that communication between CD8^+ T cells and the nervous system is altered in AD, paving the way for therapies targeting T cell-dependent neurotoxic inflammation. Subject terms: Neuroimmunology, Alzheimer's disease __________________________________________________________________ Neuroimmune communication in Alzheimer’s disease (AD) is poorly understood. Here, the authors show that CD103–CD8+ resident memory T cells induce neurotoxicity and neuroinflammation in AD via the granzyme K–PAR-1 axis. Introduction The dysregulation of innate and adaptive immunity is a driving force in the development of Alzheimer’s disease (AD), the most common form of dementia, affecting more than 32 million people worldwide^[66]1. Classical AD neuropathology is characterized by β-amyloid (Aβ) deposition, tau hyperphosphorylation, and the loss of neurons and synapses, but more recent evidence shows that chronic inflammation promoted by local and peripheral immune cells is also a hallmark of AD^[67]2,[68]3. CD8^+ T cells are one of the most intriguing components of the AD immunological landscape. During immune responses, these cytotoxic cells produce classical granzymes (such as GrA and GrB), perforin, and a plethora of cytokines^[69]4. Most CD8^+ T cells in healthy non-lymphoid organs have a tissue-resident memory (Trm) phenotype, fulfill a local protective role, and can rapidly mount immune responses^[70]5. However, CD8^+ T cells also infiltrate the brains of AD patients and equivalent animal models^[71]6–[72]13. Aging is the main risk factor for AD and recent studies have shown that CD8^+ T cells also accumulate as the brain ages, promoting axonal degeneration by releasing GrB^[73]14. But although CD8^+ T cells are found in close proximity to neuronal structures in AD, it is unclear whether this communication between adaptive immune cells and neural cells promotes disease development^[74]9. Furthermore, recent studies suggest that the meningeal compartment, which is highly enriched in immune cell populations, may also contribute to AD, but the role of meningeal CD8^+ T cells in disease development is unclear^[75]15. In most previous studies of AD brains, CD8^+ T cells have been considered as a single homogeneous population^[76]6–[77]8,[78]10. However, there is clear evidence in other tissues to support the presence of CD8^+ T cell subsets with district roles during homeostasis and immune responses^[79]4. The population of CD8^+CD45RA^+CCR7^- T effector memory (T[EMRA]) cells expands in the peripheral blood of individuals with mild cognitive impairment (MCI) and AD, and in the cerebrospinal fluid (CSF) following infection with Epstein–Barr virus (EBV), suggesting particular CD8^+ T cell phenotypes may also play a role in AD^[80]9. Furthermore, CD69^+CD103^+ and CD69^+CD103^–CD8^+ Trm cells have been observed in the human brain^[81]16, but the findings involved a heterogeneous group of subjects with multiple sclerosis (MS), various types of dementia and bipolar disorders, as well as controls with no brain disease. AD patients formed a minority in this mixed group, so it was not possible to determine how CD8^+ Trm cell populations are represented in the AD brain. The presence of CD8^+ T cell subsets has also been proposed in transgenic animal models of AD. However, these studies involved mice with separate Aβ or tau pathology and showed that CD8^+ T cells may have protective or deleterious effects depending on the disease stage and type of pathology^[82]7,[83]10–[84]13,[85]17. In APP/PS1 mice with late-stage Aβ pathology, the abundance of CD103^+CD8^+ T cells increased, and their transcriptomic profile was similar to wild-type (WT) mice, potentially explaining why the depletion of CD8^+ T cells does not affect amyloid pathology^[86]10,[87]11. The Trm marker CD103 (αE integrin) is also more abundant in the 5xFAD mouse model, which develops an early and aggressive amyloid pathology, together with higher levels of CXCR6 and PD-1 compared to WT controls, and this CD8^+ T cell subset has a protective anti-Aβ role suppressing the activation of microglia^[88]13. Although CD8^+ T cells generally seem to be deleterious in a model of pure tauopathy^[89]17, the role of specific subsets is less clear. P301S mice expressing APOE4 accumulate activated CD11c^+, KLRE1^+ and ISG15^+ CD8^+ T cells while the pool of TOX^+PDCD1^+ exhausted CD8^+ T cells is depleted. However, P301S mice lacking APOE show no increase in the abundance of brain CD8^+ T cells despite tauopathy, suggesting additional signals are needed for CD8^+ T cell responses in AD^[90]12. Aβ and tau pathologies have a synergistic effect in AD and diagnosis requires the presence of both hallmarks^[91]18–[92]20. Here we performed single-cell RNA sequencing (scRNAseq) in the 3xTg-AD mouse model, which develops both amyloid and tau pathologies, to investigate how brain and meningeal CD8^+ T cell subsets may shape the neuropathology of AD. We found that CD8^+ Trm cells are strongly dysregulated in 3xTg-AD mice, with the number of activated CD103^– cells increasing as the CD103 expression level declines in the CD8^+ T cell population. We showed that the brain is the main site of CD8^+ T cell dysregulation and found that CD103^–CD8^+ T cells originate from the circulation and invade the brain using a mechanism dependent on integrin LFA-1. We also observed higher levels of granzyme K (GrK) in the CD103^–CD8^+ T cells of 3xTg-AD mice and patients with AD compared to controls and discovered a role for GrK in the induction of neuronal dysfunction through the activation of protease-activated receptor 1 (PAR-1). Finally, we demonstrated that GrK–PAR-1 interaction induces tau hyperphosphorylation, revealing a critical immune-mediated neurotoxic axis. Together, our data show that dysfunctional communication between the immune system and central nervous system (CNS) mediated by CD103^–CD8^+ T cells and GrK–PAR-1 signaling contributes to the development of AD, identifying key molecular mechanisms that can be targeted to prevent immune-mediated neurotoxic inflammation. Results CD103^–CD8^+ Trm cells accumulate in the brains of 3xTg-AD mice Previous studies suggested that CD8^+ T cells play a detrimental role in AD, but the subsets that drive the disease and the underlying mechanisms that contribute to disease development are still unclear^[93]6–[94]12. We therefore compared the phenotypes and functions of CD8^+ T cells in WT and 3xTg-AD transgenic mice, the latter developing amyloid and tau pathologies representing neuropathological characteristics of human AD patients. Single-cell RNA sequencing (scRNAseq) of CD45^HIGH leukocytes isolated from the meninges and brains of 3xTg-AD (n = 8) mice as well as sex and age-matched WT controls (n = 8) at the onset of cognitive deficit^[95]21,[96]22 (Fig. [97]1a) revealed 13 cell types, including CD8^+ and CD4^+ T cells, neutrophils, B cells, natural killer (NK) cells, innate lymphoid cells (ILCs), macrophages, and microglia (Fig. [98]1b; Supp. Fig. [99]1a-c). Clustering analysis applied to the whole CD8^+ T cell population (3,098 cells representing 19.06% of all CD45^HIGH leukocytes; Supp. Fig. [100]1c) revealed the presence of five subsets (Fig. [101]1c), each characterized by the strong expression of known marker genes (Fig. [102]1d-f; Supp. Data [103]1): (i) T central memory (Tcm) cells (4.52%) expressing Sell, Ccr7, Nsg2, Dapl1 and Cmah^[104]23,[105]24; (ii) T effector cells (11.43%) expressing Gzma, S1pr5, Cx3cr1, Zeb2, Klrg1 and Klf2^[106]24,[107]25; (iii) CD103^– Trm cells (64.59%) expressing Gzmk, Cxcr6, Cxcr3, Ltb, Xcl1, Tnf and Eomes^[108]26,[109]27; (iv) proliferating Trm (Trm^PROL) cells (4.29%) expressing Chek1, Stmn1, Cdk1, Mki67, Mcm2, Mcm5, Mcm7, Tfdp1 and Pola2^[110]28,[111]29 and (v) CD103^+ Trm cells (15.24%) expressing Gstp3, Foxo1, Il10, Il17a and Il2ra, as well as Itgae, encoding CD103, which is related to the tissue residence phenotype^[112]30–[113]32. Cd69, previously associated with the tissue residence phenotype but also with the activation of T cells^[114]5, was expressed by most of the CD8^+ T cells in both 3xTg-AD and WT mice (Supp. Fig. [115]1d). Flow cytometry experiments confirmed this phenotypic characterization of CD8^+ T cells showing that: (i) Tcm highly expressed CD62L, encoded by Sell gene, and were negative for CD69; (ii) T effector highly expressed CX3CR1, granzyme A (GrA) and granzyme B (GrB), and were negative for CD69; (iii) CD103^– Trm cells were positive for CD69^+ and expressed high levels of Eomes; and (iv) CD103^+ Trm cells were positive for CD69^+ and expressed low levels of Eomes (Fig. [116]2a, b; Supp. Fig. [117]5f–h). Very few Trm^PROL cells were found in the scRNAseq dataset and these cells were not characterized by flow cytometry. Fig. 1. Characterization of CD8 T cell phenotype in 3xTg-AD and WT mice. [118]Fig. 1 [119]Open in a new tab a Graphical overview of the scRNAseq experimental design created in BioRender. Terrabuio, E. (2025) [120]https://BioRender.com/f3lmkb2. b UMAP plot showing CD45^HIGH leukocytes detected in the brains and meninges of wild-type (WT; n = 8) and 3xTg-AD (n = 8) mice. CD8^+ T cells (n = 3,098) are shown in red. c UMAP plots showing the five subsets of CD8^+ T cells detected in the brains and meninges of WT (n = 8) and 3xTg-AD (n = 8) mice. d Bubble plot reporting the phenotypic marker genes for each CD8^+ T cell subset. Transcript levels are color-coded. e Normalized expression of known marker genes on UMAP plot. Transcript levels are color-coded. f Violin plots showing the expression of marker genes for each CD8^+ T cell subset. White dashed line indicates the median expression level. Fig. 2. The CD8^+ Trm compartment is altered in the brains of 3xTg-AD mice. [121]Fig. 2 [122]Open in a new tab a, b Violin plots (g) and bubble plot (h) showing the expression of phenotypic markers detected by flow cytometry in the brains and meninges of WT (n = 3 pools of two organs each) and 3xTg-AD (n = 3 pools of two organs each) mice. White dashed line indicates the median expression level in violin plots (g). MFI expressions in the bubble plot (h) are scaled and color-coded. P-values are based on two-way ANOVA - multiple comparisons. Source data are provided as a Source Data file. c Donut plots indicating the distribution of CD8^+ T cell subsets in the brains of WT (left) and 3xTg-AD (right) mice d Heat map showing the expression of residency genes in the brains and meninges of WT and 3xTg-AD mice. Transcript levels are color-coded. e Violin plots showing gene expression in the brains of WT (n = 8) and 3xTg-AD (n = 8) mice. f Heat map showing the expression in CD45^HIGH leukocytes of genes in brains of WT (n = 8) and 3xTg- AD (n = 8) mice. Transcript levels are color-coded. To better understand differences in the CD8^+ population between 3xTg-AD and WT mice, we separately analyzed the phenotypic changes in the brain and meningeal compartments. Notably, scRNAseq analysis revealed an almost three-fold reduction in the abundance of CD103^+CD8^+ Trm cells in the brains of 3xTg-AD mice (WT = 27.44%, 3xTg-AD = 9.8%) with a parallel increase in the abundance of CD103^–CD8^+ Trm cells (WT = 45.9%, 3xTg-AD = 73.69%) (Fig. [123]2c). Notably, the probability to observe CD103^–CD8^+ Trm cells was significantly higher (OR = 3.30; P-value = 0) in the brain of 3xTg-AD mice compared to those of WT controls (Data source Fig. [124]2). In contrast, the probability to observe CD103^+CD8^+ Trm cells was significantly lower (OR = 0.29; P-value = 0.0038) in the brain of 3xTg-AD mice compared to those of WT controls (Data source Fig. [125]2). However, these changes were not evident in the meninges, where there was no significant accumulation of CD103^– cells (OR = 0.72; P-value = 0.09) and the proportion of CD103^+ cells (WT = 11.97%, 3xTg- AD = 19.01%; OR = 1.72; P-value = 0) increased only slightly (Supp. Fig. [126]1e; Data source Supp. Fig.[127]1). These data suggest that the major alterations in the CD8^+ Trm cell compartment occur in the brain, as corroborated by data showing the downregulation of several transcription factor (TF) genes related to the CD103^+ tissue residence phenotype (Fabp5, Stat1, Stat3, Stat4, Stat5a, Stat5b, Smad4, Smad6, Smad7 and Smad9) and the suppression of genes encoding key cytokines involved in the differentiation and maintenance of CD103^+ Trm cells (Il15, Il7, Tgfb3, Tgfb2, Tgfb1, Il21, Tnf and Il33; Fig. [128]2d, f). Moreover, the expression level of Itgae was reduced, whereas the expression of Eomes and S1pr1, which inhibits the CD103^+CD8^+ T cell phenotype^[129]5,[130]31, was upregulated in CD8^+ T cells from the brains of 3xTg-AD mice compared to WT controls (Fig. [131]2e). These data highlight the dysregulation of brain CD8^+ T cells and the depletion of the CD103^+ Trm population, presumably affecting CNS immunity and contributing to disease development. The results above were supported by unbiased cell fate trajectory analysis showing that CD8^+ T cell differentiation followed a tightly organized trajectory along with the pseudo-time progression (Fig. [132]3a), starting from a common root and ending with two differentiation states distinguished by Itgae (Arm A) and Eomes (Arm B) expression (Fig. [133]3b). Only slight differences were observed in the meninges whereas there were clear differences in the brain (Fig. [134]3c, Supp. Fig. [135]1f–l), where Arm A of the trajectory plot was populated by a lower proportion of CD103^+CD8^+ Trm cells in 3xTg-AD mice compared to WT controls (WT = 26.1%, 3xTg-AD = 14.9%) accompanied by a higher percentage of CD103^–CD8^+ Trm cells in Arm B (WT = 54.5%, 3xTg-AD = 75.8%; Fig. [136]3c; Supp. Fig. [137]1h-l). This supports the hypothesis that the brain is a fundamental point of CD8^+ T cell dysregulation in 3xTg-AD mice. Fig. 3. GrK expression is upregulated in brain CD103^–CD8^+ Trm cells of 3xTg-AD mice. [138]Fig. 3 [139]Open in a new tab a Pseudo-time ordered trajectory plot of CD8^+ T cells in the brains and meninges of WT and 3xTg- AD mice. b Expression of genes in the trajectory plots. Transcript levels are color-coded. c Trajectory plot indicating the distribution of brain CD103^– and CD103^+CD8^+ Trm cells in WT and 3xTg-AD mice. d, e Abundances of brain CD103^– (d) and CD103^+ (e) CD8^+ Trm cells in WT (n = 4) and 3xTg-AD (n = 4) mice. Data are means ± SD. P-values based on two-tailed Student’s t-test. f Scatter plot showing KEGG pathway enrichment analysis for CD103^– and CD103^+ CD8^+ Trm cells in the brains of 3xTg-AD mice (PD = “Parkinson disease”, ALS = “Amyotrophic lateral sclerosis”). g Bubble plot showing BP-GO GSEA analysis for brain CD103^–CD8^+ Trm cells in 3xTg-AD mice. h Normalized gene expression on UMAP plot of brain and meningeal CD8^+ T cells. Transcript levels are color-coded. i Violin plots showing gene expression in CD103^– and CD103^+CD8^+ Trm cells in the brain and meninges. j Violin plot showing gene expression in CD8^+ T cells in the brains of WT and 3xTg-AD mice. k Representative flow cytometry plot showing GrK expression in brain CD8^+ T cells in WT and 3xTg-AD mice. l Bar plots showing the percentage of GrK^–, GrK^MED, and GrK^HIGH CD103^– CD8^+ Trm cells in the brains of WT (n = 5) and 3xTg-AD (n = 5) mice. Data are means ± SD. P-values based on two-tailed Student’s t-test. m Scatter plot showing MFIs of CD103 and GrK in CD8^+ T cells in the brains of WT (n = 5) and 3xTg-AD (n = 5) mice. n, o Representative histograms (n) and violin plots (o) showing GrK MFI for brain CD103^– (red) and CD103^+ (blue) CD8^+ Trm cells in 3xTg-AD mice. White dashed lines indicate median expressions. P-values based on two-tailed Student’s t-test. p CD8^+ T cells (squares) in the brains of WT and 3xTg-AD mice detected by immunofluorescence microscopy. Scale bar = 5 µm or 2 µm for zoomed images. Flow cytometry showed that most brain CD8^+ T cells were CD69^+ (Supp. Fig. [140]1m) and clearly validated the presence of immune dysregulation in the brain CD8^+ Trm cell compartment. We observed a dramatic increase in both the proportion and absolute numbers of CD69^+CD103^– Trm cells in the brains of 6-month-old 3xTg-AD mice compared to sex/age-matched WT controls (WT proportion = 48.025%, 3xTg-AD = 88.08%; WT number = 6,224.72, 3xTg-AD = 12,760.5), with CD69^+CD103^+ Trm cells showing the opposite profile (Fig. [141]3d, e). However, we detected no significant differences between genotypes in the numbers of CD103^–CD8^+ Trm cells populating the meninges (WT number = 9,018.66, 3xTg-AD = 8,895.18), highlighting the brain as a key site for pathological changes in 3xTg-AD mice (Supp. Fig. [142]1n, o). Brain CD103^–CD8^+ T cells show an activated phenotype strongly associated with AD Gene set enrichment analysis (GSEA) using the Kyoto Encyclopedia of Genes and Genomes (KEGG) database reported “pathway of neurodegeneration – multiple diseases”, “Alzheimer’s disease” (AD), and “Huntington disease” (HD) as the three most enriched terms in the brains of 3xTg-AD mice (Supp. Fig. [143]1p), whereas these pathways were not enriched in the meninges (Supp. Fig. [144]1q). Importantly, whereas CD103^+CD8^+ Trm cells were negatively associated with the AD pathway suggesting a protective role, CD103^–CD8^+ Trm cells infiltrating the brains of 3xTg-AD mice showed a strong positive association with this pathway (Supp. Data [145]1), further suggesting these cells may be specifically involved in AD (Fig. [146]3f). Moreover, GSEA applied to the biological process Gene Ontology (BP-GO) database revealed an activated phenotype for CD103^–CD8^+ Trm cells populating the brains of 3xTg-AD mice compared to WT controls. Indeed, the five most enriched biological processes included “Immune system process” (GO:0002376) and “Regulation of immune system process” (GO:0002682), indicating pathways involved in the development of immune responses, “Nitrogen compound transport” (GO:0071705), which is related to T cell activation^[147]33, and “Hydrolase activity” (GO:0016787), which mediates the secretion of cytotoxic granules, suggesting a potential pro-inflammatory and cytotoxic role for CD103^–CD8^+ T cells in AD^[148]34 (Fig. [149]3g). Collectively, our data show a profound dysregulation of the CD8^+ Trm cell compartment in the brains of 3xTg-AD mice, with a loss of CD103^+ cells and the accumulation of activated CD103^– cells potentially contributing to neurotoxicity and AD development. CD103^–CD8^+ Trm cells produce granzyme K in the brains of 3xTg-AD mice We next sought to identify the pathological mechanisms mediated by activated CD103^– cells under AD-like conditions and found that Gzmk, encoding GrK, was one of the most strongly expressed marker genes in the global CD103^–CD8^+ Trm cell population (brain and meninges; Fig. [150]3h, i; Supp. Fig. [151]2a; Supp. Data [152]1). Interestingly, the genes encoding other granzymes (Gzma and Gzmb) and perforin-1 (Prf1) were not found in the list of marker genes in CD103^–CD8^+ Trm cells compared to other subsets (Supp. Data [153]1). Flow cytometry experiments validated these data confirming the low expression of GrA and GrB but high expression of GrK in the CD103^– CD8^+ Trm population (Fig. [154]2a, b), suggesting GrK may have a selective functional role in the CD103^– CD8^+ Trm cell subpopulation in AD. Importantly, Gzmk expression was upregulated in the brains but not in the meninges of 3xTg-AD mice compared to WT controls (Fig. [155]3j; Supp. Fig. [156]2b). Flow cytometry confirmed the scRNAseq data, showing a dramatic increase in the percentage of GrK-producing CD103^–CD8^+ Trm cells (both GrK^MED and GrK^HIGH) infiltrating the brain, but not the meninges, of 3xTg-AD mice compared to WT controls, whereas no differences were observed for brain-infiltrating CD103^–GrK^– CD8^+ T cells (Fig. [157]3k–m; Supp. Fig. [158]2c). Importantly, we also found that the strong increase in intracellular GrK expression was selective for the CD103^–CD8^+ Trm cell population (86% of which produced GrK) when compared to the CD103^+ population (Fig. [159]3n, o), suggesting the production of GrK is a key pathological mechanism of CD103^–CD8^+ Trm cells in the brains of 3xTg-AD mice. GrK^+CD103^–CD8^+ Trm cells induce neuronal dysfunction via the GrK–PAR-1 axis GrK is associated with pro-inflammatory and cytotoxic activities^[160]35, suggesting that GrK produced by CD103^–CD8^+ T cells may induce neuronal dysfunction in AD. Immunofluorescence staining revealed the presence of GrK^+CD8^+ T cells in close proximity to hippocampal neurons in the brains of 3xTg- AD mice, whereas CD8^+ T cells with lower GrK expression were predominantly detected in WT brains (Fig. [161]3p; Supp. Fig. [162]2j). These results indicate that GrK^+CD103^–CD8^+ Trm cells may contribute to neuronal alterations in 3xTg-AD mice. To confirm that GrK^+CD103^–CD8^+ Trm cells directly induce neuronal dysfunction in AD, we used wide-field high-resolution microscopy for the live imaging of primary hippocampal neurons isolated from 3xTg-AD mice co-cultured with CD103^– or CD103^+ CD8^+ Trm cells obtained from the livers of 3xTg-AD mice, an abundant source of CD8^+ Trm cells^[163]36 (Fig. [164]4a). Immunofluorescence staining confirmed the high density of GrK granules inside CD103^–CD8^+ Trm cells compared to CD103^+ counterparts (Fig. [165]4b). Time-lapse fluorescence microscopy revealed that primary neurons in contact with GrK^+CD103^–CD8^+ Trm cells, but not those in contact with CD103^+CD8^+ Trm cells, showed significantly higher cytoplasmic calcium (Ca^2+) levels compared to the negative control, clearly indicating that GrK^+CD103^–CD8^+ Trm cells induce intracellular Ca^2+ dysregulation (Fig. [166]4c), which is associated with neuronal alterations^[167]37. Importantly, purified active GrK directly induced intracellular Ca^2+ release in a dose-dependent manner, unequivocally demonstrating the detrimental role of this enzyme in the context of neuronal dysfunction (Fig. [168]4d). Fig. 4. GrK^+CD103^–CD8^+ Trm cells induce neuronal alterations by engaging PAR-1. [169]Fig. 4 [170]Open in a new tab a Graphical overview of the experimental design. Created in BioRender. Terrabuio, E. (2025) [171]https://BioRender.com/ir5h5rf. b Immunofluorescence staining showing GrK granules inside CD103^+ and CD103^– CD8^+ Trm cells. Scale bar = 3 µm. c Intracellular Ca^2+ release in neurons co-cultured with CD103^+ or CD103^– CD8^+ Trm cells. The last time point is shown in the right panel. Data are means ± SD from four independent experiments. P-values based on two-way ANOVA-multiple comparisons. Ctrl^– = neurons alone. Ctrl^+ = ionomycin-stimulated neurons (10 µM). Source data are provided as a Source Data file. d Intracellular Ca^2+ release in neurons treated with purified active GrK or vehicle. The last time point is shown in the right panel. Data are means ± SD from three independent experiments. P-values based on two-way ANOVA-multiple comparisons. Source data are provided as a Source Data file. e Intracellular Ca^2+ release in neurons co-cultured for 5 h with CD103^–CD8^+ Trm cells in the presence/absence of the PAR-1 inhibitor [172]SCH79797 (100 nM). The last time point (300 min) is shown in the right panel. Data are means ± SD from four independent experiments. P-values based on two-way ANOVA-multiple comparisons. Ctrl^– = neurons alone. Ctrl^+ = ionomycin-stimulated neurons (10 µM). Source data are provided as a Source Data file. f, g Intracellular Ca^2+ release in neurons cultured for 5 h with purified active GrK alone (150 nM) or with [173]SCH79797 (50 nM, or 100 nM). Ctrl^– = neurons alone. The last time point is shown in the right panel. Data are means ± SD from three independent experiments. P-values based on two-way ANOVA-multiple comparisons. Representative images are shown in (g). Red = intracellular Ca^2+ release. Scale bar = 20 µm. Source data are provided as a Source Data file. h Immunofluorescence microscopy showing GrK^+CD103^–CD8^+ Trm cells near the soma (cell I) and dendrites (cell II) of a PAR-1^+ neuron. Cell morphology was visualized by wide-field imaging using a DIC filter. Scale bar = 5 µm (or 2 µm for zoomed images I, and II). GrK can bind the thrombin receptor PAR-1^[174]38, which is implicated in synaptic plasticity and memory formation in healthy mice, but also plays a negative role in some brain pathologies^[175]39–[176]41. Importantly, immunofluorescence staining revealed a significant increase in both the number and area of PAR-1^+ neurons in the hippocampus of 3xTg-AD mice compared to WT controls, suggesting that signaling via PAR-1, a G-protein coupled receptor (GPCR), is dysregulated in AD (Supp. Fig. [177]2g, h). Importantly, wide-field time-lapse microscopy showed that the PAR-1 inhibitor [178]SCH79797 strongly reduced intra-neuronal Ca^2+ release in the presence of GrK^+CD103^–CD8^+ Trm cells (Fig. [179]4e). Moreover, our data clearly showed the strong, dose-dependent inhibition of intracellular Ca^2+ release in neurons cultured with 150 nM purified active GrK in the presence of [180]SCH79797 (Fig. [181]4f, g; Supplementary Movies [182]1-[183]3). Immunofluorescence staining confirmed the presence of GrK^+CD103^–CD8^+ Trm cells in close contact with PAR-1^+ neurons in in vitro cultures (Fig. [184]4h), and of GrK^+ CD8^+ T cells in the hippocampus of 3xTg-AD mice (Supp. Fig. [185]2i), further supporting a role for GrK–PAR-1 binding in the neuronal dysfunction underlying AD. These data demonstrate that the GrK–PAR-1 axis is a key immune-mediated pathological mechanism promoting neuronal dysfunction in AD. Brain CD103^–CD8^+ Trm cells originate from the circulation and are detrimental in 3xTg-AD mice The intraperitoneal (ip) treatment of mice with an anti-CD8 antibody depletes circulating CD8^+ T cells leaving the brain Trm compartment unaltered^[186]42,[187]43. To determine the origin of CD103^– T cells, we therefore used this method to deplete circulating CD8^+ T cells in 3xTg-AD mice and sex-matched WT controls (Supp. Fig. [188]3a-h). The anti-CD8 treatment significantly reduced the abundance of brain CD103^–CD8^+ Trm cells compared to mice treated with an isotype control antibody, reaching the levels of the control groups (Fig. [189]5a), suggesting this cell subset is replenished from the blood. However, we detected no differences in the abundance of CD103^+CD8^+ Trm cells after CD8^+ T cell depletion, in 3xTg-AD mice and WT controls (Fig. [190]5a), in line with previous data showing these cells are spared by systemic depletion^[191]43. Importantly, the ablation of CD103^–CD8^+ Trm cells in the brains of 3xTg-AD mice was paralleled by an amelioration of cognitive functions in behavioral tests, confirming that CD103^–CD8^+ Trm cells contribute to memory decline in 3xTg-AD mice. Particularly, the Morris water maze (MWM) test showed improved hippocampal-dependent learning after CD8^+ T cell depletion in 3xTg-AD mice compared to controls, as indicated by the learning curve recorded during the training days of the test (Fig. [192]5b). In addition, the probe test showed that 3xTg-AD mice devoid of circulating CD8^+ T cells actively searched the platform, maintaining the memory of the original platform position and achieving better escape performance, as also shown by the greater path efficiency and significant reduction in body rotations (Fig. [193]5c–e). The contextual fear conditioning (CFC) associative learning task revealed an amelioration of associative memory following the depletion of circulating CD8^+ T cells in 3xTg-AD mice, as shown by the significant increase in the percentage of freezing responses compared to control animals treated with an isotype control (Fig. [194]5f). Fig. 5. Detrimental CD103^–CD8^+ Trm cells in the brain originate from the circulation and their depletion ameliorates disease in 3xTg-AD mice. [195]Fig. 5 [196]Open in a new tab a Flow cytometry showing the percentage of brain CD103^– and CD103^+CD8^+ Trm cells after anti-CD8 antibody treatment. Anti-CD8 (WT, n = 6; 3xTg-AD, n = 9), isotype-control (WT, n = 6; 3xTg-AD, n = 9). Data are means ± SD from two independent experiments and P-values are based on one-way ANOVA-multiple comparisons was used. b Time spent during training (MWM test). Anti-CD8 (WT, n = 10; 3xTg-AD, n = 21), isotype-control (WT, n = 22; 3xTg-AD, n = 18). Data are means ± SD from two-independent experiments (two-way ANOVA-multiple comparisons). c Representative tracking of three mice/group (MWM test). d, e Violin plots showing path efficiency (d) and number of body rotations (e) (MWM test). Data are from two-independent experiments one-way ANOVA-multiple comparisons). f Bar plot showing the percentage of freezing during the CFC test after anti-CD8 treatment. Anti-CD8 (WT, n = 9; 3xTg-AD, n = 16), isotype-control (WT, n = 21; 3xTg-AD, n = 13). Data are means ± SD from two-independent experiments. One-way ANOVA-multiple comparisons was used. g–i Immunohistochemical staining of the hippocampus after CD8 T cell depletion showing Aβ-load (g), the levels of hyperphosphorylated (h) and total (i) tau protein. Anti-CD8 (n = 3), isotype-control (n = 3). Scale bar = 20 µm. Data are means ± SEM. Two-tailed Student’s t-test was used. ROI = 624.7 µm×501.22 µm. j–l ELISA showing Aβ 1−40 and Aβ 1−42 (j) levels of tau hyperphosphorylation (k) and total tau (l) in soluble and insoluble fractions of brain homogenates after anti-CD8 treatment. Anti-CD8 (n = 4), isotype control (n = 4). Data from three-independent experiments are means ± SEM (two-tailed Student’s t-test). m Dot blot showing insoluble oligomeric and fibrillar forms of Aβ in brain homogenates after anti-CD8 treatment. Anti-CD8 (n = 4), isotype-control (n = 4). Data from three-independent experiments are shown as means ± SEM. P-values based on two-tailed Student’s t-test. n Immunofluorescence staining showing hippocampal oligomeric and fibrillar Aβ. Scale bar = 10 µm. To confirm whether the amelioration of cognitive deficits by anti-CD8a antibody treatment correlates with the loss of neuropathological hallmarks of AD, we stained coronal murine brain sections with anti-Aβ (6E10), anti-phospho-tau (AT180), and anti-total-tau (HT7) antibodies. We observed a significant decrease in both the Aβ load and levels of tau hyperphosphorylation in the hippocampus of 3xTg-AD mice following the depletion of circulating CD8^+ T cells compared to animals treated with an isotype control (Fig. [197]5g, h), whereas the levels of total tau were unchanged (Fig. [198]5i). These data were confirmed by ELISA experiments, showing significantly less Aβ1-40 and Aβ1-42 deposition, and tau hyperphosphorylation (pT231, AT180) in the soluble and insoluble fractions of brain homogenates from 3xTg-AD mice depleted of CD8 + T cells compared to isotype controls (Fig. [199]5j, k), while no significant differences were observed in the levels of total tau (Fig. [200]5l). Similarly, we observed significantly lower levels of insoluble oligomeric (A11 antibody) and fibrillar (OC antibody) Aβ forms in the brain of 3xTg-AD mice depleted of CD8 + T cells compared to isotype controls (Fig. [201]5m, Supp. Fig. [202]3j, Supp. Data 5-8). Importantly, our immunofluorescence staining showed that the oligomeric and fibrillar forms of Aβ were located at the intraneuronal level in the hippocampus of 9 9-month-old 3xTg-AD mice, and both co-localized with the 6E10 signal (Fig. [203]5n). Collectively, these results demonstrate that CD103^–CD8^+ Trm cells originate from the circulation, infiltrate the brain, and promote memory decline and neuropathological changes that characterize AD, strongly supporting their role in AD pathogenesis. LFA-1 integrin mediates the infiltration of CD103^–CD8^+ T cells into the brain Next, we investigated the molecular mechanisms that allow circulating CD8^+ T cells to infiltrate the brains of 3xTg-AD mice. Our recent work suggests that α4-integrins are not involved in CD8^+ T cell extravasation in 3xTg-AD mice^[204]44. However, our flow cytometry data showed a significant increase in both the percentage and mean fluorescence intensity (MFI) of LFA-1^+ circulating CD8^+ T cells in the blood of 3xTg-AD mice compared to age-matched WT controls (Fig. [205]6a). These results, together with our earlier data showing the upregulation of brain endothelial intracellular adhesion molecule 1 (ICAM-1), the counterligand of LFA-1^[206]22, suggest a role for LFA-1 integrin in the trafficking of CD8^+ T cells into the brain. To understand the role of LFA-1 in more detail, we crossed 3xTg-AD mice with Itgal^–/– mice lacking functional LFA-1 integrin and observed a significant decrease in the percentage of CD103^–CD8^+ Trm cells in the brains of 3xTg-AD/Itgal^–/– mice compared to 3xTg-AD animals (Fig. [207]6b, c). Neuropathological studies showed a significantly lower Aβ load and significantly less tau hyperphosphorylation (AT180) in the brains of 3xTg-AD/Itgal^–/– mice compared to sex- and age-matched 3xTg-AD controls, but no significant difference in the levels of total tau (Fig. [208]6d–f). Overall, these data suggest a pivotal role for LFA-1 in the accumulation of CD103^–CD8^+ Trm cells in the brain, and further supports the origin of detrimental CD103^–CD8^+ Trm cells in the circulation. Fig. 6. LFA-1 integrin controls the accumulation of CD8^+CD103^– cells in the brains of 3xTg- AD mice. [209]Fig. 6 [210]Open in a new tab a Flow cytometry showing the percentage of LFA-1^+ CD8^+ T cells (left) and corresponding MFIs (right) in the blood of WT (n = 6) and 3xTg-AD (n = 6) mice. Data are means ± SD, with P-values based on two-tailed Student’s t-test. b Flow cytometry showing the percentage of CD45+ leukocytes among live cells. Data are means ± SEM, with P-values based on two-tailed Student’s t-test. c Flow cytometry showing the percentage of CD103^– (left) and CD103^+ (right) CD8^+ Trm cells in the brains of 3xTg-AD (n = 6) and 3xTg-AD/Itgal^–/– (n = 6) mice. Data are means ± SEM, with P-values based on two-tailed Student’s t-test. d–f Representative immunohistochemical staining of the hippocampus in 3xTg-AD (n = 3) and 3xTg-AD/Itgal^–/– (n = 3) mice, showing the Aβ load (c), and the levels of hyperphosphorylated (d) and total (e) tau protein. Scale bar = 20 µm. Data are means ± SEM, with P-values based on two-tailed Student’s t-test. g UMAP plot showing immune cell populations in human blood (n = 3 AD patients and n = 2 negative controls, NCs) by scRNAseq. h Normalized expression of CD3G, CD3D and CD3E on a UMAP plot. Transcript levels are color- coded i CD3^+ T cell cluster subsets: UMAP plot showing CD8^+ and CD4^+ T cell subpopulations in human blood (n = 3 AD patients and n = 2 NCs). j Normalized expression of CD8A, CD8B and CD4 on a UMAP plot. Transcript levels are color- coded. k CD8^+ T cell cluster subsets: UMAP plot showing early active, Tem, and naïve-like CD8^+ T cells in human blood (n = 3 AD patients and n = 2 NCs). l Radar plot showing AUCell score using known genes. m Heat map reporting the expression values (mean calculated using the Hurdle model) of ITGAL in AD patients (n = 3) and NCs (n = 2). Transcript levels are color-coded. Circulating CD8^+ T cells express more LFA-1 in AD patients To understand the translational potential of our data, we also investigated the role of LFA-1 in the trafficking of circulating CD8^+ T cells in AD patients. We analyzed a published scRNAseq dataset (accession [211]GSE181279) of CD45^+ leukocytes isolated from the blood of three AD patients and two negative controls (NCs)^[212]45 (Fig. [213]6g–m). We used known gene signatures to characterize CD8^+ T cells (n = 10,162) in the dataset (Fig. [214]6i, j), identifying: (i) naïve; (ii) early activated; and (iii) CD8^+ Tem cells^[215]46 (Fig. [216]6k, l; Supp. Data [217]2). Interestingly, these results showed that ITGAL, encoding the CD11a subunit of LFA-1 integrin, was upregulated in the whole population of circulating CD8^+ T cells in AD patients 1 and 3, compared to controls (Fig. [218]6m). Particularly, ITGAL expression was upregulated in the Tem subset of circulating CD8^+ T cells in AD patients compared to controls (Fig. [219]6m) but was not differentially expressed in the early activated or naïve CD8^+ T cell populations (Fig. [220]6m). These results agree with our data showing a higher percentage of circulating LFA- 1^+CD44^+KLRG1^+CD8^+ Tem cells and the stronger expression of LFA-1 on Tem cells in 3xTg-AD mice compared to WT controls (Supp. Fig. [221]4a), whereas no significant differences were observed in the KLRG1^–CD44^+ and KLRG1^–CD44^– subpopulations of circulating CD8^+ T cells (Supp. Fig. [222]4b, c). Collectively, these data confirm that circulating activated CD8^+ T cells, particularly the Tem subset, are more activated and have a greater LFA-1-dependent migration capacity in AD patients, as well as 3xTg-AD mice. GrK-producing CD8^+ T cells accumulate in the CSF and brains of AD patients We also analyzed a published scRNAseq dataset of CSF immune cells (accession [223]GSE134578)^[224]9 (Fig. [225]7a, b), revealing two subsets of CD8^+ T cells (Fig. [226]7c, d): (i) CD69^+ and PRDM1^+ Trm cells^[227]24,[228]47, and (ii) KLF2-expressing CD8^+ T cells (Supp. Data [229]3). The CD8^+ Trm population could be divided into two further subpopulations (Fig. [230]7e; Supp. Data [231]3): (i) CD103^–CD8^+ Trm cells characterized by the expression of GZMK, EOMES, and S1PR1, and (ii) ITGAE-expressing CD103^+ CD8^+ Trm cells characterized by the expression of CD7, ITGA1, KLRB1, and CCR6^[232]32. We observed an increase in the abundance of CD103^–CD8^+ Trm cells during disease progression, whereas the number of CD103^+CD8^+ Trm cells declined, supporting our data from 3xTg-AD mice (Fig. [233]7f). Importantly, we also observed a significant increase in GZMK expression in CD103^–CD8^+ Trm cells in AD patients compared to healthy controls and patients with MCI (Fig. [234]7 g, h), strongly suggesting that GrK-producing CD103^–CD8^+ Trm cells also play a detrimental role in the development of AD in humans. Fig. 7. GrK^+CD103^–CD8^+ Trm cells accumulate in the CSF of AD patients. [235]Fig. 7 [236]Open in a new tab a UMAP plot showing leukocyte populations detected in human CSF (n = 4 AD, n = 5 MCI, and n = 9 healthy controls, HCs) by scRNAseq. b Normalized expression of CD3E, CD8B and CD4 in the UMAP plot. Transcript levels are color-coded. c CD8^+ T cell cluster subsets: UMAP plot showing KLF2^+ and Trm cell subpopulations in human CSF (n = 4 AD, n = 5 MCI, and n = 9 HCs). d Normalized expression of KLF2, CD69 and PRDM1 in a UMAP plot. Transcript levels are color-coded. e Trm cell cluster subsets: UMAP plot showing CD103^– and CD103^+ Trm cells in human CSF (n = 4 AD, n = 5 MCI, and n = 9 HCs). f Percentage of CD103^– and CD103^+ CD8^+ cells in human CSF (n = 4 AD, n = 5 MCI, and n = 9 HCs). Data are means ± SD. Source data are provided as a Source Data file. g Violin plot reporting GZMK expression in Trm cells of n = 4 AD patients, n = 5 MCI subjects, and n = 9 HCs. The white dot represents median expression. h Normalized expression of GZMK in a UMAP plot of all Trm cells (panel 1) and Trm cells from HCs (panel 2), MCI subjects (panel 3), and AD patients (panel 4). We used immunofluorescence staining to determine whether GrK-producing CD103^–CD8^+ Trm cells also accumulate in the human brain during AD (Supp. Table [237]1). Importantly, whereas GrK^–CD103^+ CD8^+ T cells were predominantly detected in the hippocampus of controls, we observed the presence of both intraparenchymal and intravascular GrK^+CD103^–CD8^+ T cells near the hippocampal neurons of AD patients (Fig. [238]8a). These observations match our 3xTg-AD mice, strongly supporting a role for GrK-producing cells in AD. In further agreement with our mouse data, we observed a significant increase in the abundance of intraparenchymal CD103^–CD8^+ T cells in the hippocampus of AD patients but a lower percentage of CD103^+CD8^+ T cells (Fig. [239]8b). Although not statistically significant, we observed a slight increase in the percentage of both CD103^–CD8^+ and GrK^+CD103^– CD8^+ T cells at the intravascular level in the hippocampus of AD patients compared to controls (Fig. [240]8c). Importantly, our quantifications showed a significant increase in the number of intraparenchymal GrK^+CD103^–CD8^+ T cells in the hippocampus of AD patients compared to age-matched controls, suggesting a role for GrK in the neuronal dysfunction associated with AD in humans (Fig. [241]8b). Fig. 8. GrK^+CD103^–CD8^+ Trm cells accumulate in the brains of AD patients. [242]Fig. 8 [243]Open in a new tab a Immunofluorescence staining of control (CTRL) and AD human brain tissues. Scale bar = 40 µm, or 15 µm in the zoomed images. b Bar plots showing the percentage of intraparenchymal CD103^–, CD103^+, and GrK^+CD103^– CD8^+ T cells in the hippocampus of HCs (n = 3) and AD patients (n = 3). Data are means ± SD (two-tailed Student’s t-test). Source data are provided as a Source Data file. c Bar plots showing the percentage of intravascular CD103^–and GrK^+CD103^– CD8^+ T cells in the hippocampus of HCs (n = 3) and AD patients (n = 3). Data are means ± SD (two-tailed Student’s t-test). Source data are provided as a Source Data file. GrK-PAR-1 signaling induces functional alterations and tau hyperphosphorylation in human neuronal cells Finally, we assessed whether GrK can induce neuronal alterations via PAR-1 in human cells by measuring intracellular Ca^2+ released by differentiated SH-SY5Y human neuroblastoma cells treated with purified GrK in the presence or absence of the PAR-1 inhibitor [244]SCH79797 (Fig. [245]9a–c). We observed a significant increase in the release of intracellular Ca^2+ by GrK alone, whereas cells cultured with GrK in the presence of [246]SCH79797 showed intracellular Ca^2+ levels comparable to the negative control (Fig. [247]9b, c). Next, we showed that recombinant GrK alone significantly increased the hyperphosphorylation of tau on serine residues (pS199 and pS396), but not threonine (pT231), in differentiated SH-SY5Y cells, whereas tau hyperphosphorylation on serine residues was prevented in the presence of [248]SCH79797^[249]48 (Fig. [250]10a–d; Supp. Fig. [251]4f–h;). No differences were observed in total tau protein levels, suggesting GrK has a specific effect on the signaling machinery leading to tau hyperphosphorylation (Supp. Fig. [252]4e, i). Fig. 9. GrK induces functional alterations in human neuronal cells via PAR-1. [253]Fig. 9 [254]Open in a new tab a Immunofluorescence staining for βIII-tubulin, MAP-2, pNF-H, Nestin and PAR-1 in undifferentiated and differentiated SH-SY5Y human neuroblastoma cells. b Intracellular Ca^2+ release in differentiated SH-SY5Y cells cultured in the presence of 150 nM GrK, or 150 nM GrK and 100 nM [255]SCH79797. The last time point is shown in the bottom panel. Ctrl^– = neurons alone. Ctrl^+ = ionomycin-stimulated neurons (10 µM). Data are means ± SEM from three independent experiments. P-values based on two-way ANOVA-multiple comparisons. c Representative images of differentiated SH-SY5Y cells at different time points cultured without GrK (Ctrl^–), in the presence of 150 nM active GrK, or in the presence of 150 nM active GrK plus 100 nM [256]SCH79797. The red signal shows intracellular Ca^2+ release. Scale bar = 20 µm. Fig. 10. GrK induces tau hyperphosphorylation and alteration of signaling pathways in human neuronal cells via PAR-1. [257]Fig. 10 [258]Open in a new tab a–c Bar plots and representative images showing tau phosphorylation levels on serine and threonine (AT8, AT100) (d, e) and only threonine (AT180) (f) residues in differentiated SH-SY5Y cells cultured alone (Ctrl^–), in the presence of 100 nM [259]SCH79797, in the presence of 150 nM active GrK alone, and the presence of both. Data are means ± SD of three independent experiments (two-way ANOVA-multiple comparisons). Scale bar = 5 µm. d ELISA showing the levels of phosphorylation on the pS199, pS396, and pT231 residues of tau protein in differentiated SH-SY5Y cells cultured alone (Ctrl^–), in the presence of 100 nM [260]SCH79797, in the presence of 150 nM active GrK alone, and in the presence of both. Data are means ± SD of three independent experiments. P-values are based on two-way ANOVA multiple comparisons. e, f Protein enrichment analysis from three independent proteomic experiments showing the best enriched clusters of terms (h) and terms belonging to the “Alzheimer’s/neurodegeneration” and “kinase” pathways (i) in SH-SY5Y human neuroblastoma cells cultured in the absence of GrK and [261]SCH79797 (Ctrl^–), in the presence of 100 nM [262]SCH79797 alone, in the presence of 150 nM active GrK alone, and in the presence of both. Source data are provided as a Source Data file. To understand downstream signaling pathways induced by GrK, we analyzed the proteome of differentiated human neuroblastoma SH-SY5Y cells (treated with recombinant GrK or untreated) in the presence or absence of the PAR-1 inhibitor [263]SCH79797 (Fig. [264]10e). We found that GrK significantly increased the abundance of 84 proteins, including proteins involved in the development of AD (Supporting Data [265]4). Pathway enrichment analysis applied to upregulated proteins revealed that the “Alzheimer’s/Neurodegeneration” and “Kinase pathway” clusters were the most enriched (Fig. [266]10e). In the “Alzheimer’s/Neurodegeneration” cluster, the most significantly enriched pathways included “amyloid fiber formation”, “neurodegenerative diseases”, and “deregulated CDK5 triggers multiple neurodegeneration” (Fig. [267]10f, Supporting Data [268]4). The “Kinase pathway” cluster of terms was characterized by the upregulation of pathways related to the activation of MAP kinases^[269]49, which mediate tau hyperphosphorylation, and the “Post-translational protein phosphorylation pathway” (Fig. [270]10f). We also found that GrK induced the upregulation of “NF-κB signaling”, which in neurons is associated with the modulation of ion channel protein expression, supporting Ca^2+ increases at intraneuronal levels, and with a higher Aβ load^[271]50. Notably, the induction of all these pathways was prevented when GrK treatment took place in the presence of [272]SCH79797, leading to the significant upregulation of only 12 proteins (Supporting Data [273]4), whereas enrichment analysis detected no pathways significantly related to these proteins. Together, these data clearly support our results in 3xTg-AD mice, suggesting that CD103^–CD8^+ T cells play a negative role in AD and that blocking GrK activity and its interaction with PAR-1 may have therapeutic value to reduce neuronal dysfunction and cognitive decline in AD. Discussion CD8^+ Trm cells form a defensive line against infections and cancer but may also play a role in the development of chronic inflammatory conditions^[274]5. Human and mouse brains contain CD103^+CD8^+ and CD103^–CD8^+ Trm cells^[275]11,[276]13,[277]16,[278]42,[279]51, but their role in AD pathogenesis is unclear. Our scRNAseq analysis revealed an altered brain CD8^+ T cell compartment with more abundant CD103^– Trm cells and the inhibition of signals promoting CD103 expression in the 3xTg-AD mouse model, which develops the diagnostic neuropathological features of human AD (amyloid and tau pathologies). We showed that CD103^–CD8^+ T cells originate from the circulation, use LFA-1 integrin to extravasate into the brain and selectively produce larger quantities of GrK in 3xTg-AD mice and human AD patients compared to controls. GrK induced neuronal dysfunction and tau hyperphosphorylation in mouse and human cells via PAR-1, revealing a key immune mechanism that promotes neurotoxic inflammation and brain damage in AD (Supp. Fig. [280]6). Previous studies in transgenic mice with separate Aβ or tau pathology showed the existence of CD8^+ T cell subsets with protective or deleterious effects depending on the disease stage and pathological hallmarks^[281]7,[282]10–[283]13,[284]17. Particularly, CD8^+ T cells in APP/PS1 mice with advanced β-amyloid pathology show a transcriptomic profile similar to old WT animals and higher levels of CD103 were detected by flow cytometry^[285]11. Brain CD8^+ T cells of 5xFAD mice with aggressive late-stage Aβ pathology had higher CD103 abundance together with higher expression of CXCR6 and PD-1 (compared to WT mice), inhibiting Aβ accumulation by suppressing the activation of microglia^[286]13. Previous studies using models of tau pathology suggested brain-infiltrating CD8^+ T cells promote disease development, although additional signals appear to be essential for CD8^+ T cell accumulation in the CNS of such mice^[287]12,[288]17. Particularly, old P301S mice lacking APOE do not accumulate more brain CD8^+ T cells despite tau pathology, suggesting that CD8^+ T cell infiltration into the CNS requires APOE4^[289]12. In this context, our data show that the co-occurrence of Aβ and tau pathologies significantly reduces the abundance of intraparenchymal CD103^+CD8^+ Trm cells in the brains of 3xTg-AD mice compared to WT animals due to the downregulation of genes encoding transcription factors and cytokines involved in the differentiation and maintenance of CD103^+ Trm cells^[290]31. Particularly, our results indicate strong suppression of the homeostatic and anti-inflammatory cytokine IL-33, which is required for the generation and survival of brain CD103^+CD8^+ Trm cells^[291]52, and are in line with previous studies showing the loss of IL-33 in AD patients^[292]53. Our results also show that CD103^–CD8^+ Trm cells are more abundant in the brains of 3xTg-AD mice than WT controls. The accumulation of these cells (and the loss of CD103^+ cells) was also observed in the brains of MS patients^[293]54 and in primary Sjögren’s syndrome^[294]55 suggesting common dysregulation of the CD8^+ T cell compartment in these diseases. Similarly, CD103^–CD8^+ Trm cells accumulated in the guts of patients with inflammatory bowel disease (IBD), and the rebalancing of CD103^–CD8^+ and CD103^+CD8^+ Trm cells was associated with disease remission^[295]56, suggesting CD103^– cells promote disease pathogenesis. Indeed, our data demonstrate that CD103^–CD8^+ T cells are detrimental in 3xTg-AD mice and their depletion in the brain ameliorates neuropathology and cognitive deficits. Importantly, our data from AD patients show an increase in the abundance of intraparenchymal CD103^–CD8^+ T cells in the hippocampus while the percentage of CD103^+CD8^+ T cells declines, suggesting CD103^– cells may also promote disease development in human AD patients. We also found that brain CD103^–CD8^+ T cells originate from the circulation in 3xTg-AD mice. Previous studies have shown that intratissutal cytotoxic CD103^–CD8^+ Trm cells originate from circulating CXCR3^+ “exKLRG1” effector cells^[296]26 and our single-cell data showed that Cxcr3 was one of the best marker genes for the CD103^–CD8^+ Trm cell population, providing additional evidence that these cells migrate from the blood into the brain. CXCR3 was recently shown to regulate CD8^+ T cell infiltration and neuronal damage in a human 3D model of neuro-immune interactions, and CXCL10 (the chemokine ligand of CXCR3) accumulates in AD, suggesting that the trafficking of detrimental CD8^+ T cells in the AD brain is mediated by CXCR3–CXCL10 interaction^[297]57. Our data also show the massive depletion of CD103^–CD8^+ T cells in the brains of 3xTg-AD/Itgal^–/– mice lacking LFA-1, suggesting a role for this integrin in the migration of detrimental CD8^+ T cells into the brain during AD. Interestingly, an LFA-1 blockade in 3xTg-AD mice reduces neutrophil migration into the CNS and mitigates disease symptoms, suggesting a key role for this integrin in the migration of circulating leukocytes into the AD brain^[298]22. Our results also showed that the percentage of circulating LFA-1^+CD44^+KLRG1^+CD8^+ Tem cells increased (along with higher LFA-1 levels) in 3xTg-AD mice compared to WT controls, suggesting that circulating CD8^+ T cells are stickier and more prone to migrate into the AD brain. This is also supported by the higher expression of ICAM- 1, the vascular ligand of LFA-1, on the brain endothelial cells of AD mouse models and human AD patients, indicating that vascular inflammation may promote the recruitment of circulating activated CD8^+ T cells into the brain. Finally, our analysis of a published dataset showed that ITGAL (encoding the CD11a subunit of LFA-1) was upregulated in the whole population of circulating CD8^+ T cells in AD, suggesting that the LFA-1–ICAM-1 and CXCR3–CXCL10 axes may act in concert to promote the invasion of the brain by peripheral CD8^+ T cells in AD. We found that most CD103^+CD8^+ and CD103^–CD8^+ T cells expressed CD69, a marker of both “residency” and T cell activation, suggesting CNS Trm cells may have effector activities^[299]5. However, CD103^+CD8^+ and CD103^–CD8^+ T cells have distinct transcriptional phenotypes and functions in the normal intestine or during pathological conditions, including a model of brain viral infection^[300]11,[301]26,[302]32,[303]51,[304]56. The analysis of our datasets from 3xTg-AD and WT mice are consistent with these data and unbiased cell fate trajectory analysis clearly showed that brain CD8^+ T cell differentiation occurred on a tightly organized trajectory starting from a common root and ending with two differentiation states distinguished by the expression of Itgae and Eomes. However, only CD103^– (not CD103^+) CD8^+ Trm cells infiltrating the brains of 3xTg-AD mice were strongly associated with the “Alzheimer’s disease” pathway, suggesting these cells have distinct pathological functions specifically promoting AD. Importantly, CD103^–CD8^+ Trm cells accumulating in the brains of AD patients and 3xTg-AD mice were characterized by no significant increase in Gzmb and Prf1 expression combined with the strong expression of Gzmk and high levels of GrK protein (but low levels of GrA and GrB proteins, a distinct phenotype for CD8^+ Trm cells in AD. Indeed, recent studies performed in other contexts have shown that GrK is an atypical cytotoxic molecule that can be produced and released independently of GrA and GrB and can act in the absence of perforin^[305]58,[306]59, thus supporting the phenotype of the CD103^–GrK^+CD8^+ T cells we found in AD. GrK^+ T cells may respond to cytokine stimuli alone, given that TCR stimulation can downregulate Gzmk expression, suggesting that GrK^+ T cells, despite their activated phenotype, are not antigen-stimulated T cells in AD^[307]59. Higher numbers of GrK^+CD8^+ T cells have been reported in several pathologies, including immune aging, cancer and autoimmune diseases^[308]55,[309]58–[310]61, suggesting AD pathogenesis may share common immune mechanisms with other inflammatory conditions. In a heterogeneous group of subjects, GrK was more abundant in brain CD103^–CD69^+CD8^+ T cells than CD103^+CD69^+CD8^+ cells populations, but most brain donors had no neurological disease or were patients with MS or Parkinson’s diseases and no data could be correlated with AD^[311]16. A recent study found an increased percentage of CD69^+CD103^+ cells in the CSF of a small group of AD patients, relative to the total population of memory CD45RA^–CD8^+ T cells^[312]62. However, these authors did not apply their CSF analysis to Trm cells (the majority of T cells in the brain parenchyma) and did not consider that CSF cells contain a significant population of KLF2^+ cells with a circulation origin, as shown by our data^[313]62. CD8^+ T cells have also been found in the leptomeninges of AD patients^[314]9, and despite recent studies showing the abundance of meningeal immune cells and the role of CNS borders during neurodegenerative and neuroinflammatory diseases^[315]15, our results reveal that the number of CD103^–CD8^+ Trm cells does not change significantly and GrK secretion does not increase in the meninges, suggesting the brain is the main site of CD8^+ Trm cell dysregulation. How CD8^+ T cells communicate with neuronal cells and potentially induce deleterious effects in AD is unclear. The accumulation of CD8^+ T cells in the CNS of aged mice was recently found to promote axonal degeneration and age-related cognitive and motor decline through the release of GrB^[316]14, which we did not find in our studies of 3xTg-AD mice. In addition, age-related alteration of brain functions has also been associated with clonally expanded CD8^+ T cells producing INF-γ, which infiltrate old neurogenic niches, inhibiting the proliferation of neural stem cells^[317]63. The ablation of circulating CD8^+ T cells in APP/PS1 mice does not reduce the Aβ load or cognitive decline but increases the expression of neuronal genes such as Arc and Npas4, suggesting that brain CD8^+ T cells may modulate synaptic plasticity^[318]10. Nevertheless, CD8^+ T cells were found close to neurons in AD patients, but their ability to exert direct neurotoxic functions has not been shown^[319]9. Our data fill this knowledge gap and demonstrate that CD103^–CD8^+ Trm cells induce neuronal dysfunction by releasing GrK. Indeed, our high-resolution live imaging studies showed that neurons from 3xTg-AD mice cultured in the presence of GrK^+CD103^–CD8^+ Trm cells undergo profound functional changes. Notably, purified active GrK also induced neuronal dysfunction, confirming the neurotoxic role of this molecule. Our proteomic data on human neuronal cells further supported these observations, showing that GrK-PAR-1 interactions activate several intracellular pathways involved in neurodegeneration and AD. Moreover, we show that GrK^+CD8^+ T cells accumulated in the parenchyma near hippocampal neurons in AD patients and 3xTg-AD mice, whereas GrK^–CD8^+ T cells were preferentially located in the healthy hippocampus, further suggesting a neurotoxic role for GrK. Our data thus suggest that GrK is the basis of a key immune mechanism that induces brain neurotoxicity with a selective role in the mediation of CD103^–CD8^+ Trm cell-dependent neuronal changes in AD. We found that GrK released by CD103^–CD8^+ Trm cells activates PAR-1 (the classical thrombin receptor) on primary AD neurons, leading to functional alterations and potentially affecting neuronal networks^[320]64. GrK may also activate PAR-1-expressing glial cells, such as astrocytes, which release glutamate following PAR-1 engagement, further contributing to neuronal alterations^[321]65. Previous studies have shown that GrK cleavage and PAR-1 activation also induce the phosphorylation of ERK1/2 and MAPK, triggering the secretion of pro-inflammatory cytokines such as IL-1β, MCP-1, IL-6, and IL-8, as well as upregulating the expression of adhesion molecules on monocytes, fibroblasts and cells of epithelial origin^[322]35. Considering these pro-inflammatory activities of GrK, and our data showing that the ablation of brain CD103^–CD8^+ T cells (the majority expressing GrK) mitigates AD in mouse models, we propose that GrK–PAR-1 interactions promote chronic neuroinflammation and neurodegeneration in AD. PAR-1 has a negative role in several neuropathological conditions and its inhibition reduces brain damage and inflammation in animal models of brain ischemia^[323]66. Although not related to CD8^+ T cells, PAR-1 inhibition also ameliorates cognitive performance in rats injected intracerebrally with Aβ peptide 1–42^[324]40. However, PAR-1 engagement can also promote learning and synaptic plasticity under physiological conditions^[325]39. Notably, our data show an increase in neuronal PAR-1 expression in 3xTg-AD mice, suggesting that chronic alterations in PAR-1 signaling promote immune-driven neuropathology in AD. Importantly, our data demonstrate that neuronal PAR-1 activation by GrK in mice and humans induces tau hyperphosphorylation in vitro^[326]48 and these results are in line with previous studies showing that the number of CD8^+ T cells in the brains of AD patients correlates with tau pathology and Braak staging^[327]10,[328]17. Also, the percentage of T cells in the brains of mice with tau pathology was higher than that detected in animals with amyloidosis^[329]12. Systemic CD8^+ T cell ablation has no effect on behavioral changes or neuropathology in APP-PS1 mice^[330]10, further highlighting the pathological link between tau and CD8^+ T cells. Along with our work, this evidence may explain why an increase in the abundance of CD103^–CD8^+ Trm cells was not observed in mouse models of amyloidosis^[331]11,[332]13. However, we found that the ablation of brain CD103^–CD8^+ T cells (86% of which produce GrK) in 3xTg-AD mice reduced both amyloid and tau pathologies. The effect of CD8^+ T cell inhibition on amyloidosis is supported by the fact that Aβ and tau pathologies have a synergistic effect in AD and tau pathology itself may favor Aβ deposition^[333]19. Together, these data indicate that CD8^+ T cells can directly sustain tau pathology during AD, mechanistically linking the invasion of the brain by CD8^+ T cells and neuroinflammation with the development of AD neuropathology. In conclusion, we show that dysregulated CD103^–CD8^+ T cells and GrK–PAR-1 signaling contribute to changes in communication between the immune system and CNS, leading to neuroinflammation and neuronal dysfunction in AD. GrK inhibitors have already been developed to treat inflammatory diseases, including myocardial infarction^[334]35, whereas drugs targeting PAR-1 are already used to block platelet aggregation^[335]67, suggesting that the manipulation of GrK–PAR-1 interactions may also benefit AD patients. Methods All chemicals, kits, and antibodies are listed in Supp. Table [336]2. Ethics statement Formalin-fixed paraffin-embedded (FFPE) hippocampal sections of controls and Alzheimer’s disease (AD) cases were obtained from the Medical Research Council (MRC) London Neurodegenerative Disease Brain Bank. All material was collected from donors (females aged 51-69 at time of death as listed in Supp. Table [337]1) from whom written informed consent for brain autopsy and the use of the material and clinical information for research purposes had been obtained by MRC London Neurodegenerative Disease Brain Bank, approved by the NRES Committee London – City & East (REC references: 08/H0704/128 + 5). Our