Abstract
The role of microglia in the amyloid cascade of Alzheimerâs disease (AD) is debated due to conflicting findings. Using a genetic and a pharmacological approach we demonstrate that depletion of microglia before amyloid-β (Aβ) plaque deposition, leads to a reduction in plaque numbers and neuritic dystrophy, confirming their role in plaque initiation. Transplanting human microglia restores Aβ plaque formation. While microglia depletion reduces insoluble Aβ levels, soluble Aβ concentrations stay consistent, challenging the view that microglia clear Aβ. In later stages, microglial depletion decreases plaque compaction and increases neuritic dystrophy, suggesting a protective role. Human microglia with the TREM2R47H/R47H mutation exacerbate plaque pathology, emphasizing the importance of non-reactive microglia in the initiation of the amyloid cascade. Adaptive immune depletion (Rag2-/-) does not affect microgliaâs impact on plaque formation. These findings clarify conflicting reports, identifying microglia as key drivers of amyloid pathology, and raise questions about optimal therapeutic strategies for AD.
Similar content being viewed by others
Introduction
Alzheimerâs disease (AD) is a neurodegenerative disorder characterized by several pathological features including the aggregation of amyloid-beta (Aβ) peptides in both brain parenchyma and vasculature, aggregation of hyper-phosphorylated TAU protein within neurons, neuroinflammation, synaptic dysfunction, and progressive neuronal loss. The classical model of AD progression suggests that the disease begins with the accumulation of misfolded proteins, predominantly the amyloid-beta peptide (Aβ), forming extracellular plaques1. This phase, also referred to as the biochemical phase2, involves the build-up of amyloid plaques leading to complex responses by astrocytes and microglia, neuronal pathology, and network changes, called the cellular phase of AD2. Finally, after decades of silent pathology, AD enters the clinical phase wherein cognitive and functional symptoms manifest, typically around twenty years after amyloid plaques can first be detected. The amyloid cascade hypothesis is supported by genetic evidence that autosomal dominant mutations causing early-onset AD affect the amyloid precursor protein or its processing enzymes, causing early plaque accumulation3. Recent progress with passive immunotherapy demonstrated clinical benefits by reducing amyloid plaques from the brains of patients4,5. However, the vast majority of AD cases are not caused by autosomal dominant mutations. Still, the genetic contribution to sporadic AD risk is estimated to be about 70%6. This genetic risk for late-onset AD is largely associated with microglia, indicating a crucial role of these cells in modulating the disease7,8.
Genome-wide association studies have identified over 80 risk variants associated with AD6. These variants are predominantly or highly expressed in microglia7,9,10. However, the precise role of microglia in AD pathogenesis remains elusive. On one hand, microglia are believed to facilitate amyloid clearance through phagocytosis, potentially limiting plaque pathology11,12,13. On the other hand, microglia may also drive chronic neuroinflammation, exacerbating neuronal damage14 and even promote amyloid plaque formation and spreading12,15,16. In response to amyloid plaque accumulation, microglia cluster around amyloid plaques, adapt an activated morphology and a distinct transcriptional signature, referred to as disease-associated microglia (DAM), microglial neurodegenerative phenotype (MGnD), or activated response microglia (ARM)17,18,19. The response in human microglia encompasses in addition cytokine (CRM) and HLA responses20. The genetic risk of AD, assessed by the expression of associated risk genes, is spread over all different cell states of microglia, including homeostatic microglia20,21. Despite the well-documented microglial cellular states, their molecular functions remain unclear.
Studies investigating microglial depletion through genetic (Csf1rÎFIRE/ÎFIRE)22 or pharmacological inhibition of the colony-stimulating factor 1 receptor (CSF1R) signaling23 have yielded conflicting results regarding their role in amyloid plaque pathology. While some studies suggest that microglia depletion can inhibit plaque formation24,25, others report no change26,27 or even an increase in plaque size11,28 (Supplementary Table 1). These conflicting studies underscore the need for further research to clarify whether microglia are beneficial or detrimental to AD pathology29 and whether they act primarily during the early biochemical phase of amyloid deposition or play a more prominent role in the later cellular phase, potentially modulating the progression of AD pathology or contributing to synaptic and neuronal damage2.
In addition to microglia, the adaptive immune system has also been implicated in AD progression30. Dysregulation of T cells has been observed in the blood and CSF of AD patients31,32. CD8+ T cells were found in proximity to Aβ plaques and neuronal processes33. However, research on the interactions between adaptive immunity and microglia in the context of amyloid pathology is still limited. Notably, studies using Rag2-/-, which results in an impaired adaptive immune system in mice34 have produced conflicting results with increased amyloid plaque pathology in Rag2â/â; 5xFAD mice35, and reduced pathology and decreased brain Aβ levels in Rag2â/â; PSAPP mice36. Hence, the role of adaptive immunity in amyloid plaque formation in AD remains controversial.
Given these complexities, we hypothesized that microglia may perform distinct functions at different stages of amyloidosis, influenced by their dynamic cellular states. To test this, we employed PLX339723 mediated microglial depletion both before and after amyloid plaque deposition in AppNL-G-F mice. Additionally, we explored the role of peripheral adaptive immunity in the microglia-amyloid interaction by depleting microglia in AppNL-G-F; Rag2-/- mice. To validate our findings, we utilized a genetic model of microglia depletion (Csf1rÎFIRE/ÎFIRE)22 and investigated the effects of human microglia transplantation, including those carrying the TREM2R47H/R47H AD risk variant, which fail to respond to amyloid plaque pathology. Our findings highlight the complex disease stage- and cell-state-dependent functions of microglia in AD. Microglia are critical in the initiation of AD, specifically in the formation of amyloid plaques, challenging the prevailing view that microglia primarily serve to clear Aβ. In more advanced stages, activated microglia compact amyloid plaques and limit their toxic effects on neurons.
Results
Microglia seed early amyloid plaques
To understand the role of microglia in the early stages of amyloid formation, we depleted microglia in AppNL-G-F mice by targeting the CSF1R using a pharmacological antagonist. AppNL-G-F mice were treated with PLX3397 in chow from 1 month (before plaque pathology) until 4 months of age (Fig. 1a). The amyloid burden and associated neuritic dystrophy were evaluated. Microglia depletion was greater than 80%, as evaluated by IBA1 staining (Supplementary Fig. 1a). We used two readouts for amyloid plaque formation at 4 months of age, X34 is a dye that reacts with β-sheeted amyloids, and 82E1 is a monoclonal antibody against the Aβ peptide recognizing all conformations of Aβ plaques. Both X34-positive amyloid fibrils and 82E1-immunoreactive Aβ deposits were reduced in the PLX3397-treated group. Microglia depletion reduced the area covered by amyloid plaques and the number of plaques but not their average size (Fig. 1bâd). ELISA measurements of soluble and insoluble Aβ brain fractions from the cortex confirmed this conclusion: soluble Aβ levels remained stable (apart from a minor decrease in Aβ38), while significant reductions of Aβ38, Aβ40, and Aβ42 were observed in the insoluble fraction (Fig. 1e). The reduction in amyloid plaques was accompanied by a reduction in LAMP1+ dystrophic neurites (Fig. 1f, g). Thus microglia are seeding new plaques during the early amyloidosis phase in AD and cause increased neuritic dystrophy. The observation that soluble Aβ levels remain stable when microglia are depleted, suggests that Aβ turnover and clearance is largely independent of microglia.
Microglia compact late amyloid plaques
To investigate the role of microglia in more advanced stages of amyloidosis, AppNL-G-F mice were treated with PLX3397 after plaques had been generated, beginning at 3 months until 7 months of age (Fig. 2a). In line with previous findings that activated microglia are less dependent on CSF1R signaling19,37, microglia depletion was less effective, however, IBA1 staining area was reduced by more than 60% (Supplementary Fig. 1b). 82E1-positive amyloid plaques increased significantly in area and size, but not in number (Fig. 2bâd). The levels of soluble and insoluble Aβ in the brain were unchanged, except for a reduction in insoluble Aβ38 (Fig. 2e). Notably, insoluble Aβ42 levels in the cortex of AppNL-G-F mice are 10-fold higher than insoluble Aβ38 levels, hence the reduction in Aβ38 may be negligible. Similar to the 82E1-positive amyloid plaque staining, the LAMP1-staining for dystrophic neurites was increased in area and size (Fig. 2f, g). 82E1 and LAMP1 staining areas across the early and late treatment cohorts and controls were highly correlated (Supplementary Fig. 1c). This indicates that the decrease in dystrophic neurite pathology is likely not a direct effect of the microglia on the neurons, but the indirect consequence of plaque compaction, decreasing the exposure of the neurons to amyloid plaques. In conclusion, microglia do not alter the overall Aβ load in the brain but compact amyloid plaques and reduce dystrophic neurites in the late stage of amyloid plaque accumulation.
Sustained microglia depletion from before the onset of amyloid plaques (1 month) until 7 months of age (Supplementary Fig. 1dâh), resulted in a combination of the early seeding and late compacting effects of microglia on amyloid plaques. The total number of plaques was reduced due to reduced seeding, while the average plaque size was increased due to reduced compaction. Overall, this treatment regimen resulted in reduced amyloid area and insoluble Aβ levels in the brain (Supplementary Fig. 1eâg).
The adaptive immune system does not alter microglia-mediated modulation of amyloid plaque pathology
To test the role of adaptive immunity in modulating amyloid plaques and microglia-plaque interactions, we depleted microglia with PLX3397 in immunodeficient AppNL-G-F; Rag2-/- mice. As in the immune-competent mice (Fig. 1), we found a reduction of amyloid plaque size, number, and insoluble Aβ levels when microglia were depleted early in amyloid plaque deposition (1â4âM) in immunodeficient AppNL-G-F; Rag2-/- mice compared to AppNL-G-F; Rag2-/- mice with intact microglia (Fig. 3aâc). When microglia were depleted at more advanced stages of amyloidosis in immunodeficient mice (3â7âM), amyloid plaque area and size were increased (Fig. 3dâf), again matching the effects in immunocompetent mice (Fig. 2). Soluble and insoluble Aβ levels were unchanged except for a decrease in insoluble Aβ38. Sustained microglia depletion also resulted in similar effects as observed in AppNL-G-F mice. Plaque numbers were reduced, while the average plaque size increased, resulting in a net decrease in plaque area and insoluble Aβ levels (Supplementary Fig. 2câg). Finally, the overall soluble and insoluble Aβ concentrations were similar in AppNL-G-F; Rag2-/- mice compared to AppNL-G-F mice across all treatments. This indicates that adaptive immunity does not alter the effect of microglia on amyloid plaque load or total amyloid levels in the AppNL-G-F model.
FIRE mice fail to initiate amyloid plaques which are restored by transplantation of human microglia
Microglia depletion with pharmacological inhibitors is incomplete. To exclude effects by PLX3397-resistant microglia or off-target effects of the drug, we sought an independent way to confirm the effects of microglia depletion on amyloid plaque pathology. We generated a deletion of the fms-intronic regulatory element (FIRE) enhancer in the Csf1r gene (Csf1rÎFIRE/ÎFIRE) of AppNL-G-F; Rag2â/â; IL2rgâ/â; hCSF1KI mice. These mice develop amyloid plaques, allow for the transplantation of human microglia20,38,39, and completely lack microglia (Fig. 4a)22, hereafter referred to as FIRE mice. We xenotransplanted human-derived microglial progenitors20,39 into these mice at P4, and collected the brains 3 or 6 months after transplantation. Human transplanted microglia populate the entire mouse brain (Supplementary Fig. 3a). Additionally, staining with the DAM-marker CD9 is sparse and of low intensity in human microglia in 3-month-old FIRE mice (Supplementary Fig. 3b). By the age of 6 months, however CD9-staining is widespread and much more intense, where human CD9+ microglia cluster around amyloid plaques (Supplementary Fig. 3c). This indicates that human microglia display a delayed response to amyloid plaque deposition.
AppNL-G-F; Rag2â/â; IL2rgâ/â; hCSF1KI mice (control, with intact mouse microglia) show amyloid plaque deposition already at 6 weeks of age as demonstrated by 82E1 and X34 staining, which is not seen in FIRE mice (Fig. 4a). This result was confirmed by a 77% reduction in insoluble Aβ42 in the absence of microglia at the same age (Fig. 4b). Apolipoprotein E (ApoE), a major genetic risk factor for AD, has been shown to play an important role in amyloid plaque formation40. ApoE is upregulated once microglia become activated by exposure to amyloid plaques17,19,41. However, in his early stage, microglia are not activated yet, which was confirmed by ApoE staining (Supplementary Fig. 4) showing that at this stage of pathology, ApoE is mainly expressed by astrocytes41,42. By 3 months of age, FIRE mice develop some amyloid plaques, but to a much lower extent than control mice (Fig. 4câe), while grafting of human microglia significantly restored plaque burden in the FIRE mice (Fig. 4câe). Although the restoration is not complete, the result confirms the important contribution of microglia to amyloid plaque seeding at 3 months of age. By 6 months, the differences in amyloid load and dystrophic neurites between the control and FIRE mice groups are less pronounced but FIRE mice still have significantly reduced plaque load, insoluble Aβ levels (Supplementary Fig. 5aâc), and LAMP1+ dystrophic neurites (Supplementary Fig. 5d, e) compared to control mice. In contrast, levels of insoluble Aβ42 in xenotransplanted FIRE mice have reached similar levels as in control mice (Supplementary Fig. 5c). In conclusion, these results independently confirm the crucial role of microglia in initiating amyloid plaques in early disease stages.
Human TREM2 R47H/R47H microglia exacerbate amyloid and dystrophic neurite pathology
The experiments above demonstrate the dual interaction of microglia with amyloid plaque load. Our data suggest that microglia mediate the seeding of plaques even when they are in their homeostatic state, while compaction of amyloid plaques is performed by microglia that become activated after exposure to amyloid plaques. To test this further we grafted H9-derived WT human microglia and TREM2R47H/R47H microglia43 into the FIRE mice. The triggering receptor expressed on myeloid cells 2 (TREM2) is a transmembrane receptor abundantly expressed on microglia. Loss of function mutations and polymorphisms of the TREM2 gene (e.g., TREM2 R47H) are strong risk factors for AD10,44,45 and impair the microglia response to plaques20,46,47,48,49.
At 6-7 weeks of age, both WT and TREM2R47H/R47H xenotransplanted human microglia seeded amyloid plaques in FIRE mice (Supplementary Fig. 6), which were not present in non-grafted FIRE mice (Fig. 4a, b), confirming that non-reactive microglia are capable to seed amyloid plaques. The plaque load at that age was very low and not significantly different between WT and TREM2R47H/R47H microglia but the observation nevertheless reinforces the importance of microglia for the initial seeding of the amyloid plaques. Interestingly, at 3 months, insoluble Aβ42 levels were significantly increased in TREM2R47H/R47H microglia-grafted FIRE brains compared to WT microglia-grafted brains, suggesting increased seeding capacity of these microglia (Fig. 5c). Furthermore, the size of X34+ and 82E1+ plaques was increased as well, indicating that the late protective function in plaque compaction is also affected in the TREM2R47H/R47H microglia (Fig. 5a, b). This was accompanied by an increase in the LAMP1+ dystrophic neurite area (Fig. 5dâf). Thus TREM2R47H/R47H mutations, which prevent the activation of microglia in response to amyloid plaques, exacerbate amyloid pathology both by increasing the seeding of plaques in early amyloid deposition, and not being able to build the protective response once the plaques are formed.
Discussion
The role of microglia in AD is multifaceted. In this study, we examined microgliaâs involvement in the amyloid cascade of AD, revealing a dual-phase response. We showed that microglia operate both upstream and downstream of amyloid plaque formation, assuming two distinct cell states: homeostatic microglia facilitate plaque formation, while activated microglia compact plaques in later stages of the disease and limit neuritic dystrophy.
Early microglia depletion, prior to plaque onset in the AppNL-G-F model, led to reduced levels of insoluble Aβ and fewer plaques without affecting plaque size. This supports the notion that microglia are key in initiating plaque formation. These results were validated using an independent model of microglia depletion (FIRE mice), where human microglia transplantation restored amyloid plaque seeding, demonstrating microgliaâs direct involvement in early plaque initiation. Once plaques were formed, microglia depletion did not alter Aβ levels or plaque numbers but did impact plaque morphology, indicating that microglia also play a pivotal role in compacting plaques during advanced stages of disease. The close correlation between dystrophic neurite and amyloid staining suggests that the effects of microglia on neurites are indirect by modifying the amyloid plaque exposure to neurons. Plaque compaction by microglia appears therefore protective.
We further confirmed our findings through two pathology-relevant approaches. First, transplantation of human microglia with the TREM2R47H/R47H risk variant, known to prevent microglial activation20,46,47,48,49, exacerbated amyloid plaque and neuritic pathology compared to WT human microglia. This indicates that microglia activation is beneficial both during the early plaque formation and later plaque compaction stages of AD, although neuroinflammation may still harm other aspects of the disease. Second, we explored whether microglial expression of ApoE, a key genetic risk factor for AD, is involved in amyloid plaque formation40. Immunostaining in 6-week-old mice (Supplementary Fig. 4) confirmed previous findings that under homeostatic conditions, astrocytes are the primary providers of ApoE in the CNS41,42. ApoE expression in microglia is only detected later in the disease when they adopt an inflammatory state17,19,41. Our observations are in line with work by Henningfield et al. who showed that a microglia-specific ApoE deletion does not alter plaque formation50. However, microglia may take up APOE from their surrounding51, which serves as a co-factor for plaque formation52.
Studies before have implicated microglia in the formation of amyloid plaques12,15,16, but the consensus in the field is that microglia clear Aβ from the brain12,13. Our study resolves many of the contradictions in the previous literature (Supplementary Table 1), where microglia were variably described as either promoting or mitigating amyloid pathology. By carefully considering disease stages and microglial cell states, we demonstrate that early-stage microglia initiate plaque formation, while later-stage microglia perform plaque compaction.
Importantly, the seeding and compaction of amyloid plaques by microglia are independent of the adaptive immune system, as similar effects were observed in immunodeficient AppNL-G-F; Rag2-/- mice. This indicates that while adaptive immunity may modulate other aspects of AD30,35, microgliaâs role in amyloid plaque dynamics operates independently. This is in line with our previous RNA sequencing data analyzing the effect of Rag2-/- deficiency on transcriptomic profiles in the brain of amyloid plaque AD mice53.
Interestingly, despite significant changes in insoluble Aβ levels, which reflect amyloid plaque accumulation, soluble Aβ levels remained relatively unchanged across different ages and experimental conditions. This indicates that Aβ turnover is only marginally dependent on microglia and that the amyloid plaque accumulation is driven by microglia presence rather than increasing soluble Aβ concentration.
Our discovery that homeostatic microglia initiate amyloid plaque seeding has critical implications, as it suggests that the genetics of sporadic AD may partially converge on amyloid initiation similarly to familial AD. Homeostatic microglia uptake extracellular Aβ and concentrate it within endosomes and lysosomes at an acidic pH54,55, which could generate intracellular high molecular weight species56,57. In the interstitial and cerebrospinal fluid, the pH is neutral, and Aβ concentrations are comparatively low, which does not promote Aβ aggregation58,59. Studies have shown that microglia can uptake and release Aβ60, produce amyloid fibrils52,61 or propagate amyloid plaques15,62. These processes may be accelerated by AD risk genes, many of which are implicated in endocytosis, phagocytosis or the lysosomal system7,8. The release of seeds may be mediated by microglial cell death15,57,62 but further studies are needed. This mirrors the behavior of macrophages in other amyloidogenic diseases63,64, suggesting that the mechanism has broad biomedical relevance.
In summary, our data place microglia at the very origin of the amyloid cascade, and future studies should explore how AD risk genes expressed in homeostatic microglia20, may prime these cells for early plaque formation. Moreover, the therapeutic potential of microglia activation63,64 in promoting plaque clearance is supported by recent antibody trial results65, suggesting that activation of microglia may counteract amyloid pathology. Previously it was thought that activated microglia were detrimental to AD66,67.
In conclusion, we demonstrate that non-reactive microglia seed plaques early in AD, while activated microglia later compact plaques, limiting their toxicity. Our findings clarify previous contradictory reports on microglial function and highlight the need to consider disease stages and microglial states when developing AD therapies.
Methods
Mice
All rodent experiments were approved by the Ethical Committee for Animal Experimentation (ECD) of KU Leuven and were executed in compliance with the ethical regulations for animal research. AppNL-G-F mice (Apptm3.1Tcs+; Takaomi Saido)68 express amyloid precursor proteins (APP) at endogenous levels but contain the humanized Aβ sequence, as well as Swedish (NL; K670_M671delinsNL), Arctic (G; E693G), and Iberian (F; I716F) familial Alzheimerâs disease-causing mutations in C57BL/6 background. This strain was crossed with homozygous Rag2 knockout mice (Rag2tm1.1Cgn; Jackson Laboratory, strain 008309) to generate AppNL-G-F/Rag2-/- mice. Homozygous Rag2tm1.1Flv; Csf1tm1(CSF1)Flv; Il2rgtm1.1Flv mice (Jackson Laboratory, strain 017708) were crossed with Apptm3.1Tcs+ to generate the Rag2tm1.1Flv; Csf1tm1(CSF1)Flv; Il2rgtm1.1Flv; Apptm3.1Tcs strain. To generate the FIRE mice, homozygous mouse oocytes from Rag2tm1.1Flv; Csf1tm1(CSF1)Flv; Il2rgtm1.1Flv; Apptm3.1Tcs crosses15 in a mixed C57Bl6; BALB/c; 129S4 background were microinjected with reagents targeting the fms-intronic regulatory sequence (FIRE sequence) in the intron 2 of the mouse Csf1R gene as previously described by David Hume and Clare Pridans22. Ribonucleoproteins containing 0.3âμM purified Cas9HiFi protein 0.3âμM crRNA (5âGTCCCTCAGTGTGTGAGA3â and 5âCAATGAGTCTGTACTGGAGC3â) and 0.3âμM trans-activating crRNA (Integrated DNA Technologies) were injected into the pronucleus of 120 embryos by the CBD Mouse Expertize Unit of KU Leuven. One female founder with the expected 428âbp deletion was selected and crossed with a Rag2tm1.1Flv; Csf1tm1(CSF1)Flv; Il2rgtm1.1Flv;Apptm3.1Tcs male and the progeny were interbred to obtain a Rag2tm1.1Flv; Csf1tm1(CSF1)Flv; Il2rgtm1.1Flv; Apptm3.1Tcs; Csf1Rem1Bdes. For maintaining the colony Rag2-/-; Csf1CSF1/CSF1; Il2rg-/y; AppNL-G-F/NL-G-F; Csf1RÎFIRE/ÎFIRE males were crossed with Rag2-/-; Csf1CSF1/CSF1; Il2rg-/-; AppNL-G-F/NL-G-F; Csf1RÎFIRE/WT females as the 5 times homozygous females tend to take less care of their progeny. Mice were randomized, and both sexes were used for all experiments. Mice were housed in groups of two to five per cage with ad libitum access to food and water and a 14âh light/10âh dark cycle at 21â°C and 32% humidity. PLX3397 (Pexidartinib PLX3397, Asclepia MedChem Solutions), a CSF1R antagonist, was mixed in the mouse chow (600âmg/kg), which was replaced two times per week during the treatment period.
Immunofluorescence and Imaging
Mice were sacrificed with an overdose of sodium pentobarbital and immediately perfused with ice-cold 1âÃâDPBS (Gibco, Cat.#14190-144). After perfusion, one hemisphere was postfixed for 24âh at 4â°C in 4% formaldehyde solution (PFA) for histology. The second hemisphere was dissected on ice. The olfactory bulb and cerebellum were removed, and the hippocampus, cortex, and midbrain were individually collected. The dissected brain regions were snap-frozen and stored at â80â°C until use for biochemistry. For sectioning, the PFA fixed hemisphere was embedded in 4% Top Vision Low Melting Point Agarose (Thermo Scientific) and sectioned coronally into 35â40âµm sections using a vibrating microtome (Leica). Brain sections were collected under free-floating conditions and stored in cryoprotectant solution (40% PBS, 30% ethylene glycol, 30% glycerol) at â20â°C.
For the APOE staining, sections were washed with 1x DPBS, and antigen retrieval was performed by incubation for 10âmin in 10âmM sodium citrate at 95â°C, followed by 30âmin incubation with RNAscope® Protease III (Advanced Cell Diagnostics, Cat.#322337) at 37â°C. All other stainings were performed without antigen retrieval and sections were immediately permeabilized for 30âmin at room temperature in PBS with 0.2% Triton. After permeabilization, sections were stained with X-34 staining solution (10âμM X-34 (Sigma-Aldrich), 20âmM NaOH (Sigma-Aldrich), and 40% ethanol) for 20âmin at room temperature. X34 is a derivative of Congo red and is a fluorescent beta-sheet specific amyloid dye. Sections were washed three times with 40% ethanol for 2âmin and twice with PBSâ+â0.2% Triton for 5âmin. Sections were blocked with 5% normal donkey serum in PBSâ+â0.2% Triton X-100 for 1âh at room temperature. Primary antibody incubation was done overnight at 4â°C. The 82E1 antibody reacts with the N-terminal of amyloid Aβ peptides and, therefore, also stains diffuse Aβ-aggregates in the brain. The next day, sections were washed 3x with PBS and incubated with secondary antibodies for 2âh at RT. Sections were washed and finally mounted with Glycergel mounting medium (Agilent). The details of all antibodies used for immunofluorescence are listed in Table 1. Confocal images were obtained using a Nikon AX microscope with at Ã4 (NA 0.2), Ã20 (NA 0,75), x40 (NA 1.25), and x60 (NA 1.42). All images were acquired using similar acquisition parameters such as 16-bit, 1024âÃâ1024 quality, and images were processed in the FIJI/Image J software. All the images of Z-series stacks were then converted to Fiji/ImageJ maximum intensity projections.
For quantification of amyloid plaques (X34 and 82E1) and dystrophic neurites (LAMP1) in the whole brain hemisphere, large images with roughly 4â6 z-stacks were obtained with a 4x (NA 0.2) objective. Microscope settings were kept the same for all samples that were compared. Z-stacks were then converted to Fiji/ImageJ maximum intensity projections. The brain hemisphere was manually outlined with the polygon selection tool and added to the ROI manager. An intensity threshold for amyloid plaque quantification was selected manually and used for all the sections. In some sections, the threshold had to be adjusted, e.g., due to increased background. In those cases, a second threshold was determined which was applied for all the sections showing increased background. After thresholding, particles were analyzed and the relative area, number, and average size of particles were recorded. Two sections per brain were analyzed and the average values per brain were calculated. The same untreated 7-month-old mice were used as the control for the respective 3 to 7 months and the 1 to 7 months PLX3397 treatment. A few samples were excluded due to damaged sections or imaging artifacts. Excluded data is indicated in the source data.
For the quantification of microglia depletion, three 20x images of different cortical areas were acquired per mouse. Z-stacks were converted to maximum intensity projections and IBA1 staining was automatically thresholded (Otsu dark). Particles were analyzed to determine IBA1+ area.
Isolation of soluble and insoluble brain extracts and Aβ ELISA
Soluble and insoluble brain extracts were isolated from snap-frozen cortex samples. Samples were randomized and 8 samples across different cohorts were processed together. Snap-frozen cortices were thawed on ice and tissue weights were recorded. Ten volumes (w/v) P-TER buffer (Thermo Fisher, Cat.#78510) supplemented with cOmplete⢠Protease Inhibitor Cocktail (Roche, Cat.#5056489001) was added and the tissue was homogenized in Fast prep tubes (MP Biomedicals) for 45âs at 6.5âm/s. Samples were centrifuged for 5âmin at 5000âÃâg to remove debris. Subsequently, the supernatant was centrifuged for 1âh at 55,000ârpm at 4â°C in an Optima Ultracentrifuge using a TLA110 rotor (~125,000âÃâg) to pellet the insoluble brain fraction. The supernatant (â=âsoluble fraction) was collected and stored at -80â°C and the pellet was used for guanidine extraction.
Pellets were resuspended in 2âµl/mg tissue 6âM GuHCl solution (6âM GuHCl/50âmM Tris-HCl, protease inhibitor cocktail, pH 7.6). Samples were sonicated with a micro-tip for 30âs at 10% amplitude, vortexed for 5âmin, and incubated on a shaker for 1âh at 25â°C and 450ârpm. Samples were ultracentrifuged for 20âmin at 70,000ârpm and 4â°C (~230,000âÃâg). The supernatant, containing guanidine-soluble Aβ fractions (insoluble Aβ) were transferred into a new tube, diluted 12 times with GuHCl diluent (20âmM phosphate, 0.4âM NaCl, 2âmM EDTA, 10% Block Ace, 0.2% BSA, 0.05% NaN3, 0.075% CHAPS, protease inhibitor cocktail, pH 7.0), and stored at â80â°C until use.
Aβ38, Aβ40, and Aβ42 levels in the soluble and insoluble brain extracts were quantified by Meso Scale Discovery (MSD). Standard 96-well SECTOR plates (MSD, Cat.#L15XA-3) were coated with 1.5âµg/ml LTDA_38, LDA_40, or LTDA_Aβ42 capture antibody (homemade mouse monoclonal against Aβ38, Aβ40 or Aβ42 neoepitope respectively) in PBS, pH 7.4 at 4â°C overnight. Plates were washed 5x with PBS-T (PBSâ+â0.05% Tween 20) and blocked with 0.1% casein in PBS for 1.5âh at room temperature. Aβ standard curves were prepared with human Aβ1-38 (rPeptide Cat.#A-1078-1), Aβ1-40 (Cat.#A-1153-1), or Aβ1-42 (Cat.#A-1163-1). Samples were diluted according to previously determined concentrations. Diluted samples and standards were mixed 1:1 with LTDA_hAβN labeled with a sulfo-TAG detection antibody (homemade mouse monoclonal against the N-terminal sequence of human Aβ, in collaboration with Maarten Dewilde), in 0.1% casein in PBS, loaded on the blocked MSD plate, and incubated overnight at 4â°C. Plates were washed 5x with PBS-T and 150âµl MSD GOLD Read Buffer A (Cat.#R92TG-2) was added to the wells. Plates were read with an MSD Sector Imager 2400âA. Outliers were identified in GraphPad using ROUT (Qâ=â1%) and excluded from analysis. Outliers were likely caused by technical issues, e.g., air bubbles or bad coating of individual wells. All data is available in the source data including excluded measures and reasons for exclusion.
Differentiation of microglial progenitors and transplantation
All experiments involving human stem cells have been approved by the UZ Leuven Ethical Committee and Biobank under study protocol S62888. H9-WT (WA09) human embryonic stem cells and H9 cells with TREM2R47H/R47H mutation were differentiated into microglia progenitors and transplanted into the brains of AppNL-G-F; Rag2â/â; IL2rgâ/â; hCSF1KI; Csf1rÎFIRE/ÎFIRE mice following our published MIGRATE protocol39. H9 (WA09) cells were obtained from WiCell Research Institute and TREM2R47H/R47H cells were acquired from the lab of Catherine M. Verfaillie at KU Leuven and have been generated from H9-WT (WA09) human embryonic stem cells as described by Claes et al43.
In brief, control and TREM2R47H/R47H H9-WA09 stem cells were plated and maintained on human Matrigel-coated six-well plates in E8 flex media until reaching ~70â80% confluence. Once confluent, stem cell colonies were dissociated into single cells using Accutase (Sigma-Aldrich) and plated into U-bottom 96-well plates at a density of ~10,000 per well in mTeSR1 medium with BMP4 (50ângâmlâ1), VEGF (50ângâmlâ1) and SCF (20ângâmlâ1) for 4 days and allowed them to self-aggregate into embryoid bodies. On day 4, embryoid bodies were transferred into six-well plates (~20 embryoid bodies per well) in X-VIVO (LO BE02-060F, Westburg) (â+âsupplements) medium supplemented with SCF (50ângâmlâ1), M-CSF (50ângâmlâ1), IL-3 (50ângâmlâ1), FLT3 (50ângâmlâ1) and TPO (5ângâmlâ1) for 7âdays. A full medium change was performed on day 8. On day 11, the differentiation medium was replaced with X-VIVO (â+âsupplements) with FLT3 (50ângâmlâ1), M-CSF (50ângâmlâ1) and GM-CSF (25ângâmlâ1). On day 18, floating human microglial precursors were collected from the supernatant and engrafted into P4 mouse brains (0.5âmillion cells per pup) by bilateral injection with a Hamilton syringe as previously described39. Heterozygous AppNL-G-F; Rag2â/â; IL2rgâ/â; hCSF1KI; Csf1r+/ÎFIRE females and homozygous AppNL-G-F; Rag2â/â; IL2rgâ/â; hCSF1KI; Csf1rÎFIRE/ÎFIRE males were used for breeding, or pups were fostered with CD1 mothers when breeding with AppNL-G-F; Rag2â/â; IL2rgâ/â; hCSF1KI; Csf1rÎFIRE/ÎFIRE homozygous females to facilitate the survival.
Statistics and reproducibility
Statistical tests and data visualization were performed using GraphPad Prism10. Each data point represents one mouse and data is presented as meanâ±âSD. Animals were randomly assigned to conditions to account for potential ordering effects. For all the ELISA data, statistical outliers (caused by technical errors) were identified using the ROUT test in Prism10 (Qâ=â1%) and excluded from further analysis. 2â3 brain sections per mouse were analyzed and averaged for histological analysis. To avoid litter bias in the mouse experiments, experimental groups were composed of animals from different litters randomly distributed. Analysis was performed semi-automated or fully automated (when possible) using FIJI/Image J. Normality of residuals was checked with the ShapiroâWilk tests and homoscedasticity was checked with the F-test or Brown-Forsythe test (for three groups). Comparisons between two groups following a normal distribution were analyzed using a two-tailed unpaired t-test, when homoscedasticity requirements were not met Welchâs correction was applied. Comparisons between two groups not following a normal distribution were analyzed with the Mann-Whitney test. When three groups were compared and data were normally distributed and homoscedastic, ordinary one-way ANOVA was used; when significant, it was followed by Tukeyâs multiple comparisons test. When data were normally distributed but not homoscedastic, Welchâs ANOVA was used; when significant, it was followed by Dunnettâs multiple comparison. When data were not normally distributed, ranks were compared with Kruskal-Wallis followed by Dunnâs multiple comparisons test. Statistical significance was set at pâ<â0.05. All data necessary for the conclusions of the study are available in the main text, figures, and Supplementary Figs. Source data, including excluded data and reasons for exclusion, and detailed statistical analysis are provided with this paper.
Reporting summary
Further information on research design is available in the Nature Portfolio Reporting Summary linked to this article.
Data availability
All data necessary for the conclusions of the study are available in the main text, figures and Supplementary Figures. Source data are provided as a Source Data file. Source data are provided with this paper.
References
Hardy, J. & Selkoe, D. J. The amyloid hypothesis of Alzheimerâs disease: progress and problems on the road to therapeutics. Science 297, 353â356 (2002).
De Strooper, B. & Karran, E. The cellular phase of Alzheimerâs disease. Cell 164, 603â615 (2016).
De Strooper, B. & Karran, E. New precision medicine avenues to the prevention of Alzheimerâs disease from insights into the structure and function of γ-secretases. EMBO J. 43, 887â903 (2024).
van Dyck, C. H. et al. Lecanemab in early Alzheimerâs Disease. N Eng. J. Med. 388, 9-21 (2023).
Sims, J. R. et al. Donanemab in early symptomatic Alzheimer disease: the TRAILBLAZER-ALZ 2 randomized clinical trial. JAMA 330, 512â527 (2023).
Karlsson, I. K. et al. Measuring heritable contributions to Alzheimerâs disease: polygenic risk score analysis with twins. Brain Commun. 4, fcab308 (2022).
Sierksma, A., Escott-Price, V. & De Strooper, B. Translating genetic risk of Alzheimerâs disease into mechanistic insight and drug targets. Science 370, 61â66 (2020).
Efthymiou, A. G. & Goate, A. M. Late onset Alzheimerâs disease genetics implicates microglial pathways in disease risk. Mol. Neurodegener. 12, 1â12 (2017).
Deczkowska, A. et al. Disease-associated microglia: a universal immune sensor of neurodegeneration. Cell 173, 1073â1081 (2018).
Karch, C. M. & Goate, A. M. Alzheimerâs disease risk genes and mechanisms of disease pathogenesis. Biol. Psychiatry 77, 43â51 (2015).
Zhao, R., Hu, W., Tsai, J., Li, W. & Gan, W. B. Microglia limit the expansion of β-amyloid plaques in a mouse model of Alzheimerâs disease. Mol. Neurodegener. 12, 1â11 (2017).
Fruhwürth, S., Zetterberg, H. & Paludan, S. R. Microglia and amyloid plaque formation in Alzheimerâs disease â Evidence, possible mechanisms, and future challenges. J. Neuroimmunol. 390, 578342 (2024).
Hickman, S. E., Allison, E. K. & El Khoury, J. Microglial dysfunction and defective β-amyloid clearance pathways in aging alzheimerâs disease mice. J. Neurosci. 28, 8354â8360 (2008).
Block, M. L., Zecca, L. & Hong, J. S. Microglia-mediated neurotoxicity: uncovering the molecular mechanisms. Nat. Rev. Neurosci. 8, 57â69 (2007).
dâErrico, P. et al. Microglia contribute to the propagation of Aβ into unaffected brain tissue. Nat. Neurosci. 25, 20â25 (2022).
Spangenberg, E. et al. Sustained microglial depletion with CSF1R inhibitor impairs parenchymal plaque development in an Alzheimerâs disease model. Nat. Commun. 10, 1â21 (2019).
Keren-Shaul, H. et al. A unique microglia type associated with restricting development of Alzheimerâs Disease. Cell 169, 1276â1290.e17 (2017).
Sala Frigerio, C. et al. The major risk factors for alzheimerâs disease: age, sex, and genes modulate the microglia response to Aβ Plaques. Cell Rep. 27, 1293â1306.e6 (2019).
Krasemann, S. et al. The TREM2-APOE pathway drives the transcriptional phenotype of dysfunctional microglia in neurodegenerative diseases. Immunity 47, 566â581.e9 (2017).
Mancuso, R. et al. Xenografted human microglia display diverse transcriptomic states in response to Alzheimerâs disease-related amyloid-β pathology. Nat. Neurosci. 27, 886â900 (2024).
Sun, N. et al. Human microglial state dynamics in Alzheimerâs disease progression. Cell 186, 4386â4403.e29 (2023).
Rojo, R. et al. Deletion of a Csf1r enhancer selectively impacts CSF1R expression and development of tissue macrophage populations. Nat. Commun. 10, 1â17 (2019).
Elmore, M. R. P. et al. Colony-stimulating factor 1 receptor signaling is necessary for microglia viability, unmasking a microglia progenitor cell in the adult brain. Neuron 82, 380â397 (2014).
Sosna, J. et al. Early long-term administration of the CSF1R inhibitor PLX3397 ablates microglia and reduces accumulation of intraneuronal amyloid, neuritic plaque deposition and pre-fibrillar oligomers in 5XFAD mouse model of Alzheimerâs disease. Mol. Neurodegener. 13, 1â11 (2018).
Kiani Shabestari, S. et al. Absence of microglia promotes diverse pathologies and early lethality in Alzheimerâs disease mice. Cell Rep. 39, 1â20 (2022).
Spangenberg, E. E. et al. Eliminating microglia in Alzheimerâs mice prevents neuronal loss without modulating amyloid-β pathology. Brain 139, 1265â1281 (2016).
Olmos-Alonso, A. et al. Pharmacological targeting of CSF1R inhibits microglial proliferation and prevents the progression of Alzheimerâs-like pathology. Brain 139, 891â907 (2016).
Clayton, K. et al. Plaque associated microglia hyper-secrete extracellular vesicles and accelerate tau propagation in a humanized APP mouse model. Mol. Neurodegener. 16, 1â16 (2021).
Romero-Molina, C. et al. Should we open fire on microglia? Depletion models as tools to elucidate microglial role in health and alzheimerâs disease. Int. J. Mol. Sci. 22, 9734 (2021).
Chen, X. & Holtzman, D. M. Emerging roles of innate and adaptive immunity in Alzheimerâs disease. Immunity 55, 2236â2254 (2022).
Larbi, A. et al. Dramatic shifts in circulating CD4 but not CD8 T cell subsets in mild alzheimerâs disease. J. Alzheimerâs Dis. 17, 91â103 (2009).
Lueg, G. et al. Clinical relevance of specific T-cell activation in the blood and cerebrospinal fluid of patients with mild Alzheimerâs disease. Neurobiol. Aging 36, 81â89 (2015).
Gate, D. et al. Clonally expanded CD8 T cells patrol the cerebrospinal fluid in Alzheimerâs disease. Nature 577, 399â404 (2020).
Shinkai, Y. et al. RAG-2-deficient mice lack mature lymphocytes owing to inability to initiate V(D)J rearrangement. Cell 68, 855â867 (1992).
Marsh, S. E. et al. The adaptive immune system restrains Alzheimerâs disease pathogenesis by modulating microglial function. Proc. Natl Acad. Sci. USA 113, E1316âE1325 (2016).
Späni, C. et al. Reduced β-amyloid pathology in an APP transgenic mouse model of Alzheimerâs disease lacking functional B and T cells. Acta Neuropathol. Commun. 3, 71 (2015).
Shi, Y. et al. Microglia drive APOE-dependent neurodegeneration in a tauopathy mouse model. J. Exp. Med. 216, 2546â2561 (2019).
Mancuso, R. et al. Stem-cell-derived human microglia transplanted in mouse brain to study human disease. Nat. Neurosci. 22, 2111â2116 (2019).
Fattorelli, N. et al. Stem-cell-derived human microglia transplanted into mouse brain to study human disease. Nat. Protoc. 16, 1013â1033 (2021).
Bales, K. R. et al. Apolipoprotein E is essential for amyloid deposition in the APP V717F transgenic mouse model of Alzheimerâs disease. Proc. Natl Acad. Sci. USA 96, 15233â15238 (1999).
Eskandari-Sedighi, G. & Blurton-Jones, M. Microglial APOE4: more is less and less is more. Mol. Neurodegener. 18, 99 (2023).
Xu, Q. et al. Profile and regulation of apolipoprotein E (ApoE) expression in the CNS in mice with targeting of green fluorescent protein gene to the ApoE locus. J. Neurosci. 26, 4985â4994 (2006).
Claes, C. et al. Human stem cellâderived monocytes and microglia-like cells reveal impaired amyloid plaque clearance upon heterozygous or homozygous loss of TREM2. Alzheimerâs. Dementia 15, 453â464 (2019).
Jonsson, T. et al. Variant of TREM2 Associated with the Risk of Alzheimerâs Disease. N. Engl. J. Med. 368, 107â116 (2013).
Guerreiro, R. et al. TREM2 variants in AD. N. Engl. J. Med. 368, 117â127 (2013).
Wang, Y. et al. TREM2 lipid sensing sustains the microglial response in an Alzheimerâs disease model. Cell 160, 1061â1071 (2015).
Song, W. M. et al. Humanized TREM2 mice reveal microglia-intrinsic and -extrinsic effects of R47H polymorphism. J. Exp. Med. 215, 745â760 (2018).
Wood, J. I. et al. Plaque contact and unimpaired Trem2 is required for the microglial response to amyloid pathology. Cell Rep. 41, 1â13 (2022).
Zhou, Y. et al. Human and mouse single-nucleus transcriptomics reveal TREM2-dependent and TREM2-independent cellular responses in Alzheimerâs disease. Nat. Med. 26, 131â142 (2020).
Henningfield, C. M., Arreola, M. A., Soni, N., Spangenberg, E. E. & Green, K. N. Microglia-specific ApoE knock-out does not alter Alzheimerâs disease plaque pathogenesis or gene expression. Glia 70, 287â302 (2022).
Preman, P. et al. APOE from astrocytes restores Alzheimerâs Aβ-pathology and DAM-like responses in APOE deficient microglia. EMBO Mol. Med. (online ahead of print) 1â29 (2024).
Kaji, S. et al. Apolipoprotein E aggregation in microglia initiates Alzheimerâs disease pathology by seeding β-amyloidosis. Immunity 57, 2651â2668 (2024).
Balusu, S. et al. MEG3 activates necroptosis in human neuron xenografts modeling Alzheimerâs disease. Science 381, 1176â1182 (2023).
Lomakin, A., Teplow, D. B., Kirschner, D. A. & Benedeki, G. B. Kinetic theory of fibrillogenesis of amyloid β-protein. Proc. Natl Acad. Sci. USA 94, 7942â7947 (1997).
Harper, J. D., Wong, S. S., Lieber, C. M. & Lansbury, P. T. Assembly of Aβ amyloid protofibrils: an in vitro model for a possible early event in Alzheimerâs disease. Biochemistry 38, 8972â8980 (1999).
Hu, X. et al. Amyloid seeds formed by cellular uptake, concentration, and aggregation of the amyloid-beta peptide. Proc. Natl Acad. Sci. USA 106, 20324â20329 (2009).
Friedrich, R. P. et al. Mechanism of amyloid plaque formation suggests an intracellular basis of Aβ pathogenicity. Proc. Natl Acad. Sci. USA 107, 1942â1947 (2010).
Andreasen, N. et al. Cerebrospinal Fluid-Amyloid (1-42) in Alzheimer disease differences between early-and late-onset Alzheimer disease and stability during the course of disease. Arch. Neurol. 56, 673â680 (1999).
Strozyk, D., Blennow, K., White, L. R. & Launer, L. J. CSF AÃ 42 levels correlate with amyloid-neuropathology in a population-based autopsy study. Neurology 60, 652â656 (2003).
Chung, H., Brazil, M. I., Thwe Soe, T. & Maxfield, F. R. Uptake, degradation, and release of fibrillar and soluble forms of Alzheimerâs amyloid-peptide by microglial cells. J. Biol. Chem. 274, 32301â32308 (1999).
Frackowiak, J. et al. Ultrastructure of the microglia that phagocytose amyloid and the microglia that produce β-amyloid fibrils. Acta Neuropathol. 84, 225â233 .
Baik, S. H., Kang, S., Son, S. M. & Mook-Jung, I. Microglia contributes to plaque growth by cell death due to uptake of amyloid β in the brain of Alzheimerâs disease mouse model. Glia 64, 2274â2290 (2016).
Claus, S. et al. Cellular mechanism of fibril formation from serum amyloid A1 protein. EMBO Rep. 18, 1352â1366 (2017).
Magy, N., Benson, M. D., Liepnieks, J. J. & Kluve-Beckerman, B. Cellular events associated with the initial phase of AA amyloidogenesis: Insights from a human monocyte model. Amyloid 14, 51â63 (2007).
Cadiz, M. P. et al. Aducanumab anti-amyloid immunotherapy induces sustained microglial and immune alterations. J. Exp. Med. 221, e20231363 (2024).
Spangenberg, E. E. & Green, K. N. Inflammation in Alzheimerâs disease: Lessons learned from microglia-depletion models. Brain Behav. Immun. 61, 1â11 (2017).
Leng, F. & Edison, P. Neuroinflammation and microglial activation in Alzheimer disease: where do we go from here? Nat. Rev. Neurol. 17, 157â172 (2021).
Saito, T. et al. Single App knock-in mouse models of Alzheimerâs disease. Nat. Neurosci. 17, 661â663 (2014).
Acknowledgements
The authors thank Prof. Catherine Verfaillie for the TREM2R47H/R47H cells, Prof. Maarten Dewilde for the antibodies used for Aβ ELISAâs. An Snellinx for help with the transplantation work. Amber Claes and Veronique Hendricks for breeding and taking care of the mice and the VIB Mouse Expertize Unit for generation of the FIRE mice. This work was funded by a Medical Research Grant (MR/Y014847/1) awarded to B.D.S., the European Research Council (ERC) under the European Unionâs Horizon 2020 Research and Innovation Programme (grant agreement no. ERC-834682 CELLPHASE_AD). The Flanders Institute for Biotechnology (VIB vzw), a Methusalem grant from KU Leuven and the Flemish Government, the Fonds voor Wetenschappelijk Onderzoek, KU Leuven, The Queen Elisabeth Medical Foundation for Neurosciences, the Opening the Future campaign of the Leuven Universitair Fonds, The Belgian Alzheimer Research Foundation (SAO-FRA) and the Alzheimerâs Association USA. B.D.S. holds the Bax-Vanluffelen Chair for Alzheimerâs Disease. N.B. is the recipient of a PhD fellowship from Fonds voor Wetenschappelijk Onderzoek (fellowship no. 11PSA24N). Schemes in Figs. 1a and 2a, and Supplementary Figs. 1d and 2c were created in BioRender.
Author information
Authors and Affiliations
Contributions
N.B., S.B., and B.D.S conceived and designed the study and wrote the manuscript. N.B. performed experiments and analyzed data. G.A. and S.C.B. performed experiments involving microglia transplantation. L.S. generated the FIRE mouse line with the assistance of CP. All authors discussed the results and commented on the manuscript.
Corresponding author
Ethics declarations
Competing interests
B.D.S. has been a consultant for Eli Lilly, Biogen, Janssen Pharmaceutica, Eisai, AbbVie and other companies and is now consultant to Muna Therapeutics. B.D.S is a scientific founder of Augustine Therapeutics and a scientific founder and stockholder of Muna Therapeutics. The remaining authors declare no competing interests.
Peer review
Peer review information
Nature Communications thanks the anonymous reviewers for their contribution to the peer review of this work. A peer review file is available.
Additional information
Publisherâs note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Supplementary information
Source data
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the articleâs Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.
About this article
Cite this article
Baligács, N., Albertini, G., Borrie, S.C. et al. Homeostatic microglia initially seed and activated microglia later reshape amyloid plaques in Alzheimerâs Disease. Nat Commun 15, 10634 (2024). https://doi.org/10.1038/s41467-024-54779-w
Received:
Accepted:
Published:
DOI: https://doi.org/10.1038/s41467-024-54779-w