Abstract Monoclonal antibody immunotherapy targeting the clearance of amyloid-β (Aβ) has shown promise in Alzheimer’s disease (AD). However, current antibody treatments trigger Fc receptors and induce proinflammatory responses, in turn exacerbating neuronal damage. Here, we report a synthetic efferocytic receptor (SER) integrating Aβ-targeting scFv, efferocytosis receptor backbone based on TIM4 and downstream signal for microglia (MG) reprogramming, which enabled selective elimination of Aβ without inducing an inflammatory response. Specifically, our in-house–customized MG-editing mRNA lipid nanoparticles (MERLINs) efficiently introduced SER mRNA into MG to generate Aβ-specific SER-MG in situ. SER-MG exhibited robust Aβ-specific phagocytosis and stimulated anti-inflammatory efferocytosis typical signaling in vitro. In a mouse model of AD, SER expression in the MG markedly increased the clearance of Aβ and dampened inflammation, resulting in improved behavioral outcomes along with substantially reduced synapse elimination. Our findings establish that AD-associated aberrant MG can be in situ reprogrammed with SER for Aβ clearance in an anti-inflammatory manner, with broad application in other inflammation-related diseases. __________________________________________________________________ Microglia with a synthetic receptor show enhanced Aβ clearance without inflammation, opening pathways for AD treatment. INTRODUCTION Neuroinflammatory responses mediated by abnormal amyloid deposition and dysregulated immune cell activation are critical causative factors in neurodegenerative diseases, including Alzheimer’s disease (AD) ([70]1, [71]2). Characterized by the accumulation of amyloid-β (Aβ) plaques and neurofibrillary tangles of hyperphosphorylated tau proteins, AD manifests mainly as progressive cognitive decline and memory impairment, and currently affects millions of individuals without an available cure ([72]3–[73]6). Existing treatments, such as the monoclonal antibody aducanumab, are shown to selectively bind Aβ aggregates and substantially reduce Aβ burden in AD patients’ brains ([74]7–[75]9). Despite promoting amyloid clearance, these antibody-based treatments only offer modest symptomatic relief in patients at the early and middle stages of AD, accompanied by several side effects, including brain edema and microhemorrhage associated with Fc receptor activation and complement cascade reactions ([76]10–[77]12). Furthermore, antibody-induced inflammatory responses within the central nervous system (CNS) could result in increased neuronal damage and inhibited neurogenesis, potentially exacerbating cognitive impairment ([78]7, [79]12–[80]14). Therapies that can effectively eliminate Aβ and inhibit the inflammatory response are urgently needed, but such approaches remain unexplored. Microglia (MG), the resident immune cells of the brain, play a crucial role in maintaining CNS homeostasis by clearing protein aggregates and cellular debris ([81]15–[82]17). This function is achieved mainly through efferocytosis, a process by which MG eliminate dying cells while triggering signals that inhibit the inflammatory response. As a classical efferocytic receptor, T cell immunoglobulin mucin protein 4 (TIM4) is predominant in maintaining immune homeostasis and preventing autoimmune responses ([83]18, [84]19). TIM4-derived chimeric receptor for efferocytosis (CHEF) generated by fusing the signaling domain of cytoplasmic adapter protein ELMO1 to the cytoplasmic tail of TIM4 displays a marked increase in efferocytosis and dampening of inflammation in multiple models of tissue injury ([85]18). In the AD microenvironment, MG exhibit impaired phagocytosis and efferocytosis due to a disturbed local microenvironment, leading to the accumulation of toxic debris and fostering a proinflammatory environment that exacerbates neurodegeneration ([86]20–[87]22). Thus, reprogramming AD-associated aberrant MG in situ through constructing synthetic efferocytic receptor (SER) based on CHEF could restore MG phagocytosis capacity while simultaneously attenuating multiple inflammatory insults, thus eliminating Aβ and mitigating AD progression. Here, we introduce a therapeutic strategy in which MG-editing mRNA lipid nanoparticles (MERLINs) are used to generate SER-modified MG (SER-MG) in situ. MERLINs encapsulating modified SER-mRNA are formulated with custom ionizable lipids, 1,2-dimyristoyl-rac-glycero-3-methoxypolyethylene glycol (DMG-PEG)–Mannose for MG in situ targeting and lysophosphatidylcholine-oleate (LPC-oleate) as a helper lipid to achieve high-efficiency meningeal and ventricular penetration after intrathecal injection. The SER, a variant of CHEF, consists of a single-chain variable fragment (scFv) derived from a monoclonal antibody targeting Aβ and the backbone of the TIM4 receptor, combining the effective signaling domain of the cytoplasmic adapter protein ELMO1 to trigger the downstream efferocytic signaling pathway ([88]18). The engineered MG are designed to specifically recognize and engulf Aβ aggregates, alleviate neurotoxicity, and thereby mitigate AD. Compared with aducanumab, the generated SER-MG manifests strong efferocytosis, promotes the clearance of Aβ without inflammation, and substantially reduces neuronal death and synaptic loss, thus leading to improved behavioral outcomes. Collectively, our work provides a reversible and broadly applicable treatment strategy for AD and potentially for other neurodegenerative diseases. This work highlights the critical need for synthetic biological approaches that could efficiently target and clear toxic aggregates without triggering a concomitant inflammatory response for AD treatment. By leveraging mRNA-based therapeutics, we offer a compelling strategy that could transform the landscape of AD therapy and provide synthetic receptor design strategies to treat inflammation-related diseases. RESULTS The SER mediates noninflammatory efferocytosis of Aβ TIM4 is a PtdSer-binding receptor that mediates efferocytosis and is expressed exclusively by phagocytes. During efferocytosis, phagocytes take up debris while processing the ingested materials in an anti-inflammatory manner, thereby maintaining homeostasis of the microenvironment ([89]23). Here, we describe a SER that combines an scFv targeting Aβ, the TIM4 efferocytic receptor backbone, and the effective structural domain of downstream ELMO1, which is expressed on the surfaces of MG through proper design, modification, and transfection of mRNA ([90]Fig. 1, A and B). On the basis of the mRNA sequence, we first predicted the spatial structure of the SER and modeled the molecular docking of Aβ with the anti-Aβ scFv via AlphaFold2 (fig. S1) ([91]24). The analysis revealed that the SER has a TIM4-like backbone along with other functional structural domains, including a green fluorescent protein (GFP) tag ([92]Fig. 1C and fig. S2). Then, we applied lipid nanoparticle (LNP) technology to deliver mRNA encoding the SER to BV2 cells and found that transient expression of the SER could be achieved ([93]Fig. 1D). To validate the structure of the SER, further analysis demonstrated that the transfection efficiency of the anti-Aβ scFv with the G4S linker reached approximately 38.27% and 32.99% in BV2 cells and primary MGs, respectively ([94]Fig. 1E and fig. S3). Fig. 1. SER-MG mediate the phagocytosis of Aβ without inducing proinflammatory responses. [95]Fig. 1. [96]Open in a new tab (A) Schematic of SER mRNA design. (B) Schematic illustrating structural design of SER and its variants. (C) Structural simulation of TIM4 and SER with AlphaFold2. (D) Immunofluorescence images of BV2 cells expressing the SER with a GFP tag. Blue, DAPI; green, SER. Scale bar, 10 μm. (E) Flow cytometry profiles of SER expression in BV2 cells and primary MG with a G4S linker of anti-Aβ scFv. (F) Immunofluorescence results for SER-MG incubated with Aβ for 1 hour (left) and corresponding linear scan of fluorescence intensity along white dotted line (right). Blue, DAPI; green, SER; red, dock180. Scale bar, 2 μm. (G) Immunofluorescence results (left) and enlarged images (right) for SER-MG incubated with Aβ for 1 hour. Blue, DAPI; green, phalloidin; red, Aβ. Scale bars, 2 μm (left) and 0.5 μm (right). (H and I) Immunofluorescence results (H) and calculated phagocytic index (I) for BV2 cells in different treatment groups after phagocyting Aβ plaques for 2 hours. Blue, DAPI; red, Aβ; green, WGA. Scale bar, 1 μm (n = 10). (J) Fluorescence images of BV2 cells after incubation with Aβ for 6 hours (left) and corresponding linear scan of fluorescence intensity along white dotted line (right). Blue, DAPI; green, LysoTracker; red, Aβ. Scale bar, 5 μm. (K) Quantitative analysis of secreted TNF-α, IL-6, and IL-1β in Aβ-coincubated MG with different treatments (n = 9). (L) Quantitative analysis of secreted IL-10 and TGF-β in Aβ-coincubated MG with different treatments (n = 6). The data are presented as the means ± SD. The statistical comparisons in (I), (K), and (L) were performed via one-way ANOVA (ns = not significant, *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001). (D), (F), (G), (H), and (J) show representative images of the corresponding independent biological samples. Furthermore, two SER variants, SER [anti–fluorescein isothiocyanate (FITC)] and SER (6M), were designed for subsequent functionality validation and mechanism exploration. As shown in [97]Fig. 1B, the anti-Aβ scFv was replaced with anti-FITC scFv in SER (anti-FITC), and six active sites of ELMO1 C terminus in SER were mutated for constructing SER (6M) ([98]18). In the presence of Aβ, immunofluorescence staining showed tangible colocalization of SER and dock180, while not observed in SER(6M)-MG, followed by cytoskeletal deformation of SER-MG for Aβ endocytosis ([99]Fig. 1, F and G, and figs. S4 to S6) ([100]25). This finding indicated that ELMO1 domain endowed SER with the ability to mimic the Rac activation and actin rearrangement activity of the natural ELMO/Dock180 complex and promote cytoskeleton reorganization in efferocytosis. Coincubation of the SER-MG with Aβ revealed that SER-MG had more robust phagocytotic effects than those of the monoclonal antibody targeting Aβ (aducanumab), yet SER (anti-FITC)–MG and SER (6M)–MG showed the deficiency in Aβ phagocytic capacity, indicating that both anti-Aβ scFv and ELMO1 domain were indispensable for Aβ-targeting phagocytosis induced by SER ([101]Fig. 1, H and I). Moreover, the phagocytosed Aβ was degraded within the lysosomes of BV2 cells ([102]Fig. 1J). Further analysis revealed that aducanumab substantially increased the secretion of inflammatory factors, whereas SER-MG demonstrated reduced inflammatory factor secretion as well as increased anti-inflammatory factor production after the phagocytosis of Aβ ([103]Fig. 1, K and L). The lower expression of proinflammatory factors [tumor necrosis factor–α (TNF-α), interleukin-6 (IL-6), and IL-1β] and the upper expression of anti-inflammatory factors [IL-10 and transforming growth factor–β (TGF-β)] in the SER-treated group than that in the aducanumab-treated group also indicated a robust anti-inflammatory effect of SER-MG (fig. S7). Additionally, the impact of SER on native phagocytic pathways of MG was also investigated. When coincubated with apoptotic cells, SER-MG exhibited comparable phagocytosis efficiency to untransfected MG, demonstrating a low risk of interfering with MG’s normal physiological function (fig. S8). Collectively, our designed and synthesized SER-MG have a specific structure and demonstrate a superior capacity to enhance the phagocytosis of Aβ while concurrently mitigating inflammatory responses, establishing the basis for subsequent cognitive improvement in vivo. Synthesis and characterization of LNPs targeting Mfsd2a Given that major facilitator superfamily domain-containing protein 2a (Mfsd2a) is selectively expressed on the cerebral vascular endothelium, meninges, and ventricles ([104]Fig. 2A), where it transports common LPCs carrying long-chain fatty acids, including LPC-oleate and LPC–docosahexaenoic acid (DHA) ([105]26–[106]28), we propose that substituting helper lipids in conventional LNP components with LPC-oleate may achieve meningeal and ventricular penetration to deliver RNA drugs effectively following intrathecal injection. First, to screen and obtain suitable carbon chains with desirable transfection efficiency, we modified LPC-oleate with saturated fatty acids, including 10-carbon, 12-carbon, 14-carbon, 16-carbon, and 18-carbon fatty acids, and their structures were confirmed by ^1H nuclear magnetic resonance ([107]Fig. 2B and fig. S9). Next, our house-synthesized thymine ionizable lipid T12 was applied to encapsulate the mRNA, and then a microfluidic device was used to formulate different LNPs ([108]Fig. 2C and fig. S10). As shown in [109]Fig. 2 (D and E), the longer the carbon chain of LPC-oleate was, the larger the resulting particle size. However, there was no discernible effect on the surface charge of the LNPs ([110]Fig. 2F). We next tested the mRNA delivery efficacy of LNPs after the replacement of helper lipids with LPC-oleate, and flow cytometry analysis revealed that the desirable transfection efficiency of LNPs was achieved ([111]Fig. 2, G and H). LNP-14 loaded with mRNA encoding GFP exhibited outstanding meninges-penetrating efficiency (approximately 65.4%) and achieved the transfection of target cells in a transwell model (fig. S11). The in vivo luciferase expression distribution following intrathecal injection ([112]Fig. 2J) also confirmed that LNP-14 had better cerebral delivery efficiency, which were 8.91, 2.94, 2.04, and 1.43 times greater than those of LNP-10, LNP-12, LNP-16, and LNP-18, respectively ([113]Fig. 2, I and K). Furthermore, we designed a C14-based formulation optimization strategy and found that the cells were transfected with the highest efficiency when the lipid ratios used in regimen 2 were used; therefore, this regimen was adopted for subsequent in vivo and in vitro studies (table S1 and fig. S12). In summary, these results indicate that LNPs synthesized with LPC-oleate display superior performance and have the potential for RNA delivery to generate SER-MG in situ. Fig. 2. Construction and characterization of brain-penetrating LNPs. [114]Fig. 2. [115]Open in a new tab (A) Expression of Mfsd2a in the meninges and ventricles according to immunofluorescence staining. The white dashed line indicates the surface of the cerebral parenchyma; blue, DAPI; green, Mfsd2a. Scale bar, 25 μm. (B) LPC-oleate library. (C) Schematic of LNP preparation. (D) Particle diameter distribution of LNPs using LPC-oleate equipped with saturated fatty acid chain ranging from C10 to C18 as helper lipids. (E) Average particle diameter of LNP-10, LNP-12, LNP-14, LNP-16, and LNP-18 (n = 3). (F) Zeta potentials of LNP-10, LNP-12, LNP-14, LNP-16, and LNP-18 (n = 3). (G) Flow cytometry results after the transfection of GFP-mRNA with LNPs based on different LPC-oleates. (H) Quantitative fluorescence assay of cells transfected with different LNPs (n = 3). (I to K) Scheme for intrathecal injection in mice (J), visualization of luciferase expression distribution (I), and quantitative analysis of brain bioluminescence intensity (BLI) (K) after the injection of different luciferase mRNA-laden LNPs into nude mice (n = 3). The data are presented as the means ± SD. The statistical comparisons in (H) and (K) were performed via one-way ANOVA (*P < 0.05, ****P < 0.0001). Targeting MG for the cellular delivery of SER-mRNAs CD206, also known as the macrophage mannose receptor, is a membrane protein expressed on the surfaces of macrophages that is intimately implicated in the phagocytosis and immune phenotypes of MG. CD206-expressing MG generally exhibit a phenotype associated with anti-inflammatory responses and tissue repair ([116]29, [117]30). Before confirming the targeting of mouse MG, we found that CD206 was expressed on the surface of MG after immunofluorescence staining of AD mouse brain sections (fig. S13). To endow LNPs with the ability to target MG, mannose moieties capable of strongly binding to the CD206 receptor were covalently modified to the end of the DMG-PEG. LNP-14, in which DMG-PEG2000 was replaced with DMG-PEG-Mannose, was designated as MERLIN ([118]Fig. 3A) The formulated MERLIN solution showed a typical Tyndall effect after exposure to a laser at 532 nm ([119]Fig. 3B). The images of MERLINs captured by transmission electron microscopy (TEM) revealed a spherical structure with an approximate size of 150 nm, consistent with the dynamic light scattering (DLS) measurements ([120]Fig. 2C). Further analysis of the stability after 15 days of storage at 4°C, as shown in fig. S14, revealed that no obvious mRNA leakage was detected in MERLINs for up to 15 days, with negligible changes in size and zeta potential. Fig. 3. MERLINs selectively target MG and achieve intracellular mRNA release. [121]Fig. 3. [122]Open in a new tab (A) Schematic diagram of the process by which MGs internalize MERLINs and express the SER on the surface of the cell membrane for the clearance of Aβ. (B) Transmission of LNPs detected by a 532-nm laser. (C) TEM image showing the morphology of the constructed LNPs. (D and E) Fluorescence images (D) and corresponding intensity (E) of BV2 cells after treatment with PBS, LNP-Cy7, and MERLIN-Cy7 (n = 3). Scale bar, 20 μm. (F and G) Flow cytometry profiles (F) and corresponding quantitative analysis (G) of GFP fluorescence intensity after administration of GFP-mRNA, LNP, and MERLINs encapsulating GFP-mRNA to BV2, C8-D1A, and bEnd3 cells, respectively (n = 3). (H) Representative immunofluorescence images of BV2 cells after 2, 4, and 6 hours of incubation with MERLINs. Blue, DAPI; green, LysoTracker; red, Cy5-mRNA. Scale bars, 1 μm (left) and 2 μm (right). (I) Immunofluorescence showing the distribution of MERLIN-Cy7 and LNP-Cy7 within the cerebral parenchyma (white arrows, colocalization between MG and MERLIN-Cy7). Blue, DAPI; green, IBA1; red, MERLINs/LNPs. Scale bars, 100 μm (top) and 10 μm (bottom). (J) Quantitative analysis of MG internalizing MERLINs or LNPs (n = 10). The data are presented as the means ± SD. The statistical comparisons in (E) and (G) were performed via one-way ANOVA and two-way ANOVA, respectively. The statistical comparison in (J) was calculated using Student’s t test (**P < 0.01, ****P < 0.0001). (D), (H), and (I) show representative images of the corresponding independent biological samples. We next used flow cytometry and confocal laser scanning microscopy (CLSM) for MG-targeting delivery assessment of MERLIN and found that LNP modified with mannose (MERLIN) substantially increased the uptake efficiency of MG ([123]Fig. 3, D and E). Compared with that of mouse astrocyte C8-D1A cells and cerebral endothelial bEnd3 cells, the efficiency of GFP-mRNA transfection of MERLINs in BV2 cells was notably higher, suggesting that MERLINs are capable of selectively delivering mRNA to MG in the CNS ([124]Fig. 3, F and G). SER expression was then monitored after MERLIN transfection, and it peaked at 48 hours and almost returned to baseline at 96 hours (fig. S15). In addition, we examined the lysosomal escape ability of MERLINs via CLSM and revealed that MERLIN@CY5-mRNA colocalized mainly with LysoTracker Green–stained organelles after 2 hours of incubation. The separation of the green and red fluorescence spots was more notable after 6 hours, suggesting that MERLIN@CY5-mRNA efficiently escaped from the endosomes or early lysosomes to the cytoplasm ([125]Fig. 3H and fig. S16). Immunofluorescence staining of whole-brain sections after drug administration revealed substantial infiltration of MERLINs within the brain parenchyma, with predominant colocalization with MG, but LNPs permeated the brain parenchyma nonspecifically ([126]Fig. 3, I and J). Together, these results demonstrate that the constructed MERLINs could effectively penetrate the cerebral parenchyma, target MG, and achieve intracellular mRNA release and expression, demonstrating great feasibility for MG reengineering in vivo. SER-MG facilitates Aβ clearance and attenuates AD in vivo To evaluate the capability of SER-MG to eliminate Aβ and mitigate cognitive impairment in vivo, 9-month-old APP/PS1 mice were selected and divided into three groups ([127]31). Wild-type mice were also included. The mice were intrathecally injected with phosphate-buffered saline (PBS), aducanumab, or MERLINs every 3 days for a total of 10 injections ([128]Fig. 4A). Aβ deposition, as demonstrated by thioflavin T staining of the brain tissues of mice, was reduced at the end of treatment, with a reduction of approximately 57.2% in the MERLIN-treated group and approximately 40.9% in the aducanumab-treated group compared with the control group. A sustainable Aβ reduction was observed in the MERLIN-treated group, indicating that SER-MG could effectively phagocyte and degrade Aβ, and ultimately alleviate the Aβ burden in the brains of AD mice ([129]Fig. 4, B and C). Moreover, cerebral Aβ deposition was monitored for 4 weeks following MERLIN treatment, and Aβ levels remained relatively low for 3 weeks but rebounded in the fourth week (fig. S17). Subsequently, two variants of SER were also used for further mechanism exploration of SER-induced Aβ clearance. Compared with the aducanumab-treated group, MERLIN encapsulating SER mRNA inhibited the abnormal deposition of Aβ and reduced the damage to neurons manifested by LAMP1 expression ([130]Fig. 4, D and E), which may be attributed to the construction of SER-MG in situ, mimicking the traditional efferocytosis signaling pathway and mediating Aβ phagocytosis in an anti-inflammatory manner ([131]Fig. 4, F and G, and fig. S18). In contrast, neither Aβ endocytosis nor reduced neuron damage was observed in two variant groups, indicating that the anti-Aβ scFv and ELMO1 motif were the key elements contributing to Aβ clearance and neuronal repair in SER. Mice were then tested for novel object/location recognition, and the discrimination index of mice treated with MERLINs was substantially improved, indicating that SER-MG could improve the cognition and memory of AD model mice ([132]Fig. 4, H to M, and fig. S19). In additional Morris water maze tests, mice in MERLIN treatment groups exhibited similar learning function to wild-type mice and determined the location of the platform more precisely than that in the control group after training for 5 days, demonstrating the therapeutic impact of SER on learning, cognition, and memory (fig. S20). Thus, we speculate that, compared with aducanumab treatment, the SER-MG generated by synthesized MERLINs can notably increase the phagocytosis and clearance of Aβ by MG in an anti-inflammatory manner, inhibit neuronal damage, and thus improve cognitive conditions in AD model mice. Fig. 4. SER-MG mediates Aβ clearance and behavior rescue in APP/PS1 mice. [133]Fig. 4. [134]Open in a new tab (A) Schematic illustration of the experimental design. WT, wild type. (B and C) Thioflavin T staining (B) and statistical analysis of fluorescence intensity (C) of brain sections from normal and APP/PS1 mice after various treatments. Scale bar, 200 μm (n = 3). (D and E) Representative immunofluorescence images (D) and statistical analysis (E) of Aβ burden and neuron damage in wild-type or APP/PS1 mice from different groups. Blue, DAPI; green, LAMP1; red, Aβ. Scale bars, 100 μm (left) and 30 μm (right) (n = 3). (F and G) Immunofluorescence staining of wild-type and AD mouse brain sections (F) from different treatment groups to observe Aβ phagocytosis by MG and quantitative analysis of fluorescence intensity (G). Scale bars, 50 μm (left) and 10 μm (right) (n = 20). (H to J) Graphical illustration of the novel object location (NOL) test (H) and representative pictures of the locomotor trajectories of mice in different treatment groups (I) with statistical analysis (J) (n = 6). (K to M) Graphical illustration of the novel object recognition (NOR) test (K) and representative pictures of the movement trajectories of mice in different treatment groups (L) with statistical analysis (M) (n = 6). The data are presented as the means ± SD. The statistical comparisons in (C), (E), (G), (J), and (M) were performed via one-way ANOVA (ns = not significant, *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001). (B), (D), (F), (I), and (L) show representative images of the corresponding independent biological samples. Single-cell RNA sequencing reveals the neuronal improvement effects of SER-MG To further elucidate the mechanism of the therapeutic strategy of generating SER-MG via MERLINs in vivo, we performed single-cell sequencing of the hippocampi from AD mice treated with PBS, aducanumab, or MERLINs via the same procedure described above ([135]Fig. 4A). The data revealed that editing MG with MERLINs increased the number of MG in the hippocampal region, which is critical for the elimination of Aβ ([136]Fig. 5, A and B, and fig. S21). The number of Cajal-Retzius cells is higher in the brains of MERLIN-treated AD mice than in those of aducanumab-treated mice, which contributes to neuronal remodeling in the brain and underlies the improvement in cognition in AD mice ([137]32, [138]33). Fig. 5. Single-cell sequencing analysis reveals the mechanism of SER-MG treatment in APP/PS1 mice. [139]Fig. 5. [140]Open in a new tab (A) Single-cell sequencing analyses of the brain hippocampal region of AD mice in the PBS, aducanumab, and MERLIN treatment groups and the corresponding UMAP plots of cells depicting separation into nine clusters. (B) Cluster fractions of cells from the hippocampus of each treatment group. (C and D) GSEA comparing MERLIN and aducanumab treatment. Gene set terms are indicated as follows: FDR, false discovery rate; NeS, normalized enrichment score. (E) KEGG enrichment analysis of signaling pathway enrichment of neurons in the MERLIN versus aducanumab treatment groups. (F to H) GO enrichment analysis of neurons based on sequencing results from different treatment groups. MG from different treatment groups were further segregated for differential expression gene (DEG) analysis. Consistent with the in vitro data, Fc-mediated endocytosis was down-regulated in MERLIN-treated mice, and the activation of inflammation-related signaling pathways, such as the mammalian target of rapamycin (mTOR) pathway, was inhibited according to gene set enrichment analysis (GSEA) comparing MERLIN and aducanumab treatment, confirming that SER-MG activate efferocytosis rather than FcR-mediated Aβ phagocytosis ([141]Fig. 5, C and D). Further analysis of the scRNA-seq data explored the molecular mechanisms by which SER-MG induced an anti-inflammatory phenotype. We identified 235 genes up-regulated and 174 genes down-regulated in the MERLIN group when compared to the aducanumab group (fig. S22A). Quantitative heat map analysis of the inflammation-related genes highlighted in the volcano plot showed substantial up-regulation of anti-inflammatory genes and down-regulation of inflammatory genes including Tnf and Il6, which was consistent with the enzyme-linked immunosorbent assay (ELISA) results (fig. S22B). Gene ontology (GO) annotation of differentially overexpressed genes in MG revealed that differentiational genes are enriched in pathways associated with mitosis and cytoskeletal remodeling, indicating that the construction of SER facilitates microglial proliferation and promotes cytoskeletal reorganization, thereby enhancing their capacity to phagocytose amyloid proteins (fig. S22, C and D). To elucidate the influence of the construction of SER-MG on CNS, neurons from different treatment groups were segregated for DEG analysis similarly. The pathway enrichment analysis based on the Kyoto Encyclopedia of Genes and Genomes (KEGG) was then used to further explore the mechanism of SER’s improving effect on neuronal repair and cognition elevation ([142]34). Compared with those in the aducanumab-treated group, expression of genes related to axon guidance (Slit1, Slit2, Nrp1, Epha6, and Unc5c) and synapse formation (Prkca, Cacna1c, Kcnq5, and Grik4) was up-regulated in AD mice treated with MERLINs. Additionally, genes with up-regulated expression were enriched in the oxytocin signaling pathway, which is thought to be associated with neuroplasticity and the modulation of social behavior ([143]35, [144]36) ([145]Fig. 5E and fig. S23A). Further analysis by GO annotation revealed notable enrichment of genes related to axon generation or guidance in biological process (BP), synaptic formation in cellular component (CC), and signal transduction activities in molecular function (MF). Overall, these results collectively elucidate the mechanism by which MERLIN treatment improved cognitive function in AD mice compared with aducanumab treatment ([146]Fig. 5, F to H, and fig. S23, B to D), suggesting its potential for application in other neurodegenerative diseases. Collectively, we developed a reengineering strategy by using MERLINs to selectively deliver SER-mRNAs into MG and generate Aβ-specific SER-MG in situ, which effectively eliminate Aβ and suppress the intracranial inflammatory response more effectively than aducanumab treatment does, thus promoting neural repair and improving behavioral outcomes in AD model mice ([147]Fig. 6). Fig. 6. Intracranial generation of SER-MG for anti-inflammatory clearance of Aβ. [148]Fig. 6. [149]Open in a new tab (A) Schematic of MERLIN-enabled in situ generation of SER-MG for alleviating AD via Aβ clearance and inflammation resolution. (B) Transport of MERLINs penetrating the meninges or ventricles. (C) MG selectively internalize MERLINs via CD206 and express the SER on their surface, subsequently activating downstream signals to reorganize the cytoskeleton and mediate anti-inflammatory responses. DISCUSSION Aberrant deposition of Aβ plaques in the brain extracellular space is a key hallmark of AD, and brain Aβ accumulation is commonly denoted as the initiating event in AD’s pathogenesis, resulting in subsequent neurofibrillary tangle formation, synapse loss, and neuronal cell death ([150]37–[151]39). Current treatments for AD involve Aβ clearance through multiple means, including the use of monoclonal antibodies. Aducanumab, the first antibody therapy approved by the U.S. Food and Drug Administration for AD treatment, has effectively reduced Aβ burdens within the brains of AD patients. However, owing to side effects such as Fc receptor activation and complement cascade reactions, such therapies may activate local immune cells while inducing neuroinflammation, further exacerbating cognitive and memory deficits in AD patients ([152]7, [153]9). Eliminating Aβ while restricting the inflammatory response is therefore of utmost importance in AD treatment ([154]40). The SER-MG described in this study could facilitate Aβ elimination and simultaneously suppress neuroinflammatory responses ([155]Figs. 1 and [156]4). Further single-cell RNA sequencing of the hippocampus elucidated the neural improvement effects of SER-MG in vivo, revealing up-regulated neuronal axon guidance and synapse restoration signaling pathways, indicating the potential for mitigating AD in patients ([157]Fig. 5). MG, the predominant type of immune cell in the brain, maintain homeostasis in the CNS through the phagocytic clearance of protein aggregates and cellular debris. However, MG in the AD brain display signs of impaired phagocytosis, and dysfunction in MG’s efferocytosis could lead to the accumulation of such debris and promote a proinflammatory and anti-repair microenvironment that may result in further brain tissue dysbiosis and neurodegeneration ([158]41). Thus, restoring the phagocytic functions of aberrant MG represents a promising therapeutic strategy. Here, we first reported a SER that enables MG to specifically recognize and phagocytose Aβ in a noninflammatory manner through the fusion of an scFv targeting Aβ, the TIM4 efferocytic receptor backbone, and the effective fragment of downstream ELMO1. SER-MG were demonstrated to mimic the TIM4 signaling pathway, facilitating Aβ clearance via cytoskeletal rearrangement, and exhibited more robust elimination effects than those of aducanumab without eliciting local neuroinflammatory responses ([159]Fig. 1). This therapeutic approach provides a strategy for the noninflammatory clearance of abnormal protein deposits, warranting further exploration in other inflammation-associated diseases. To achieve precise reprogramming of MG, we propose a MERLIN mRNA delivery system modified with mannose to target CD206, which is selectively expressed on MG, and observed desirable specific cellular internalization. Additionally, we demonstrated that substituting helper lipids in conventional LNP components with LPC-oleate achieved meningeal and ventricular penetration to deliver RNA drugs effectively after intrathecal injection. This optimized MERLIN delivery system could serve as an effective tool for delivering nucleic acid drugs into MG, providing therapeutic potential for the treatment of AD and other cerebral diseases ([160]Figs. 2 and [161]3). However, there are several limitations to our current study. Although MERLIN-mediated SER-MG in situ editing therapy has demonstrated notable efficacy in AD treatment, the limitation must be noted that its impact on nervous system still needs intuitive and rigorous evaluations. Additionally, further studies should continue to explore the long-term safety and clinical prospects of this system and evaluate its potential application in other brain diseases. In particular, assessing the immunogenic responses that may arise from repeated administrations is essential for future translation into the clinic. In summary, our findings establish that MG in the AD microenvironment can be reengineered to express Aβ-specific SER via MERLINs, thereby enabling them to eliminate Aβ plaques without inducing neuroinflammation to prevent neural and synaptic damage. Therefore, our work may provide a more efficient immunotherapeutic strategy for patients suffering from AD or other inflammation-related diseases. METHODS Animals APP/PS1 mice (female, 9 months old) were purchased from GemPharmatech Co. Ltd. (Nanjing, China). All mouse experiments were performed according to the Research Ethics Committee of Shandong University and the Ethics Committee of Qilu Hospital (Shandong, China), in compliance with all relevant ethical regulations. The mice were housed in a barrier environment under 12-hour light/dark cycle with a humidity between 40% and 60% and a constant temperature of 24°C. Materials All solvents (analytical grade) and reagents were used as supplied by commercial sources unless otherwise indicated. Cell culture flasks were purchased from NEST Biotechnology (Wuxi, China). mRNA encoding luciferase was purchased from APExBIO (Beijing, China). DMG-PEG-Cy7 was obtained from Chongqing Yusi Pharmaceutical Technology Co. Ltd. (Chongqing, China). Cholesterol, 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE), and DMG-PEG were obtained from AVT Pharmaceutical Tech Co. Ltd. (Shanghai, China). Aβ was purchased from Scierbio (Hefei, China). Information of antibodies used was listed as follows: Mfsd2a (Cell Signaling Technology, clone name: E8U2O), IBA1 (Abcam, clone name: [162]EPR16589), CD206-APC (BioLegend, clone name: C068C2), G4S Linker-phycoerythrin (PE) (Cell Signaling Technology, clone name: E7O2V), DOCK180 (Cell Signaling Technology, clone name: C4C12), β-amyloid (Cell Signaling Technology), and LAMP1-FITC (BioLegend, clone name: 1D4B). Phalloidin-iFluor 488 (Abcam), 4′,6-diamidino-2-phenylindole (DAPI), and Alexa Fluor 647–labeled goat anti-rabbit immunoglobulin G (IgG) were purchased from Beyotime Biotech Inc. (Shanghai, China). DyLight 549–labeled goat anti-rabbit IgG was purchased from Abbkine Scientific Co. Ltd. (Wuhan, China). Thioflavin T was purchased from Sigma (Shanghai, China), and wheat germ agglutinin (WGA)–Alexa Fluor 488, LysoTracker, fetal bovine serum (FBS), Dulbecco’s modified Eagle’s medium (DMEM), penicillin, and trypsin-EDTA were obtained from Thermo Fisher Scientific Inc. (Shanghai, China). Serum-free cell freezing medium was purchased from New Cell & Molecular Biotech (Suzhou, China). ELISA kits for TNF-α, IL-6, IL-1β, IL-10, and TGF-β were purchased from PeproTech Inc. (Cranbury, NJ, USA) and Beijing Solarbio Science & Technology Co., Ltd. (Beijing, China). Extraction of primary cells Meningeal tissues are dissected from anesthetized mice by carefully removing the meninges from the brain. The tissues are then minced into small pieces and enzymatically digested using trypsin at 37°C. After digestion, the cell suspension is filtered through a cell strainer (70 μm) to remove debris, followed by centrifugation to collect the cells. The isolated cells are resuspended in a culture medium (DMEM supplemented with 10% FBS and 1% penicillin-streptomycin solutions) and then plated for cultivation at 37°C in a 5% CO[2] incubator. Initially, the culture medium was replaced every other day. Subculturing was performed every 2 to 3 days when the cell density exceeded 90%. Cells were purified following subculturing by using the differential adhesion method after trypsinization. For the extraction of primary MG, culture surfaces were aseptically coated with poly-d-lysine solution (0.1 mg/ml) for 30 min. The brains were dissected, placed in a dish containing cold DMEM medium, and dissociated by repeated pipetting followed by filtration through a 70-μm cell strainer and centrifuged at 400g for 5 min at room temperature. The supernatant was discarded, and cells were resuspended in culture system (DMEM supplemented with 10% FBS and 1% penicillin-streptomycin solutions) and then plated for cultivation at 37°C in a 5% CO[2] incubator. The culture medium was completely replaced the following day, and thereafter, half of the medium was replaced every 3 days. After 2 weeks, MG were harvested from the medium by shaking the flasks at 180 rpm for 2 hours at 37°C. The MG were then plated at a density of 5 × 10^4 for 24 hours before further experimentation. Preparation of ionizable lipid N-tert-butoxycarbonyl-1,2-ethylenediamine (10 mmol), 1-bromododecane (22 mmol), potassium carbonate (20 mmol), and acetonitrile (60 ml) were added to a 250-ml round-bottom flask and refluxed at 80°C for 72 hours. The solids were removed by filtration, and the residue was purified through column chromatography, yielding tert-butyl (2-(didodecylamino)ethyl)carbamate. The intermediate (5 mmol) was then dissolved in 1,4-dioxane (10 ml), followed by the addition of 4 M hydrochloric acid 1,4-dioxane (12.5 ml). The mixture was stirred at room temperature for 2 hours to yield N,N-didodecylethane-1,2-diamine, which was subsequently reacted with thymine-1-acetic acid (5 mmol) at room temperature for 6 hours in the presence of 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDCI; 5 mmol) and N-hydroxysuccinimide (NHS; 5 mmol). The resulting product was then purified by column chromatography using a dichloromethane/methanol (50:1) eluent, yielding the final compound, T12. Preparation of LPC-oleate Choline glycerophosphate and dibutyltin oxide were dissolved in isopropanol and stirred at 100°C for 6 hours. Triethylamine was then added at 0°C, followed by the successive addition of fatty acid chlorides, including decanoyl chloride, lauryl chloride, palmitoyl chloride, and stearoyl chloride. The reaction mixture was stirred at room temperature for 16 hours. The solvents were subsequently removed under reduced pressure, and the residues were purified by column chromatography to yield the intermediates LPC-C10, LPC-C12, LPC-C14, LPC-C16, and LPC-C18. Next, oleic acid, 1-methylimidazole (NMI), and 2-methyl-6-nitrobenzoic anhydride (MNBA) were added to the dichloromethane solution. After stirring at room temperature for 20 min, the LPC intermediates were added, and the mixtures were stirred at room temperature for 48 hours. The products were concentrated and purified by silica gel column chromatography to obtain LPC-oleate with different carbon chain lengths. Preparation and characterization of the LNP To synthesize LNP, a microfluidic device was used to mix an aqueous phase containing mRNA with an ethanol phase containing lipids and cholesterol. Specifically, the aqueous phase was prepared using 50 mM citric acid buffer (pH 4.5) and mRNA, while the ethanol phase consisted of T12, helper lipid, cholesterol, and DMG-PEG at molar ratios described in table S1, respectively. The two phases were combined in a 3:1 ratio within the microfluidic device. Following this mixing process, the LNP was dialyzed against PBS for 0.5 hours. Additionally, MERLIN was prepared replacing DMG-PEG with DMG-PEG-Mannose according to optimal formula. The average particle size, polydispersity index (PDI), and zeta potential of the mRNA-LNP were determined with Zetasizer Nano ZS90 (Malvern Instruments, Malvern, UK). The data represent the average of three measurements from different samples. The morphology of MERLIN was examined with a transmission electron microscope (HT7800, Hitachi, Japan). Samples were dropped onto a carbon-formvar copper grid and negatively stained with a 2% phosphotungstic acid solution for this analysis. A gel electrophoresis retardation assay was performed to evaluate the encapsulating ability of MERLIN. Agarose gels (2%) were prepared using 1× Tris Acetate-EDTA buffer (TAE) buffer and heated in a microwave oven until the agarose was completely dissolved. The samples were then loaded onto the gels and subjected to electrophoresis at 120 V for 25 min. The resulting bands were visualized under an ultraviolet (UV) lamp. To study MERLIN storage stability at 4°C, the average particle size and zeta potential were monitored for a week. mRNA delivery property characterization of MERLIN To evaluate the MG-targeting property of MERLIN, 1 × 10^5 of BV2, C8-N1A, and bEnd3 cells were embedded on six-well plates. Then, cells were cultured with MERLIN for transfection. Particle phagocytosis was analyzed by confocal microscope imaging at 1 hour and the intracellular behavior of internalized MERLINs was recorded by confocal microscope imaging at 2, 4, and 6 hours. In vitro transwell model An in vitro transwell model was established using a six-well transwell cell culture system. BV2 cells were seeded in the lower chamber at a density of 1 × 10^5 cells per well. Similarly, the upper chamber was seeded with mouse primary meningeal cells at the same density. Cellular integrity was assessed by measuring the transepithelial/transendothelial electrical resistance (TEER) values using a Millicell-ERS voltmeter (Millipore). A TEER value greater than 200 Ω/cm^2 indicated the successful establishment of the transwell model. LNP-Cy7 encapsulating GFP-mRNA with different LPC-oleate compositions was added to the upper chamber, and supernatants of both chambers were collected to evaluate LNP concentration at 1 hour. After 24 hours of incubation, fluorescence microscopy was used to visualize the BV2 cells in the lower chamber and quantify the fluorescence intensity of GFP. In vivo distribution study Five groups of mice (three mice per group) were treated with luciferase mRNA-laden LNPs containing different LPC-oleate by intrathecal injection. After 12 hours of injection, mice were anesthetized with isoflurane and biofluorescence imaging (BLI) was performed using an in vivo imaging system (IVIS) (Spectrum, PerkinElmer, MA, USA) to monitor the distribution of expressed luciferase. SER expression kinetics To determine the duration of SER expression, the cells were fixed at 0, 12, 24, 48, 72, and 96 hours after transfection. The fixed cell samples were stained with DAPI and recorded by Leica TCS Sp8 (Wetzlar, Germany). The fluorescence intensity of GFP fused to the C terminus of SER was quantified using ImageJ. Apoptotic cell phagocytosis of SER-MG The Jurkat cells were stained with 2',7'-bis-(2-carboxyethyl)-5-(and-6)-carboxyfluorescein,acetoxymethyl ester (BCECF-AM) and irradiated under 254-nm UV light for 15 min to induce apoptosis and then added to BV2 cells pretreated with PBS or SER mRNA-laden LNP for 2 hours of coincubation. Subsequently, free Jurkat cells were removed, and the BV2 cells were fixed and stained with DAPI for detection with Leica TCS Sp8 (Wetzlar, Germany). Aβ oligomerization For the oligomerization of Aβ, 1 mg of Aβ was dissolved in 400 μl of HFIP (1,1,1,3,3,3-hexafluoro-2-propanol; Sigma, 105228) at room temperature for 2 hours. The Aβ solution was then lyophilized using a freeze dryer to completely remove HFIP and subsequently resuspended in 5% dimethyl sulfoxide (DMSO) in PBS to achieve a final concentration of 500 μM, followed by 30 min of sonication. The dissolved Aβ was incubated at 4°C for 24 hours to induce oligomerization and stored at −20°C for further analysis. Flow cytometry analysis After 48 hours of transfection by LNP with different LPC-oleate or MERLIN encapsulation GFP or SER-mRNA, cells were collected and suspended in PBS, filtered through a 70-μm nylon cell filter, and counted for flow cytometry analysis. Cytokine measurement Cytokine secretion was detected using ELISA kits using a double antibody sandwich ELISA technique. The wells of the ELISA plates were coated with capture antibodies and incubated overnight at room temperature. This was followed by a 1-hour incubation with a blocking buffer. To observe inflammatory responses under our culture conditions, as to in vitro experiments, BV2 cells were pretreated with lipopolysaccharide (LPS) at a concentration of 200 ng/ml and then incubated with Aβ (5 μM) in the presence of PBS, aducanumab, or MERLIN. Standards and supernatants were detected under the instruction. As to in vivo evaluation, the harvested tissue samples were ground into a homogenate on ice, followed by centrifugation to obtain supernatant for detection. Color development was measured using a microplate reader (Bio-Rad, CA, USA) at 450 nm, with a wavelength correction set at 620 nm. Measurements of mRNA levels of cytokines in cells were performed by quantitative polymerase chain reaction (qPCR). BV2 cells were stimulated with LPS (200 ng/ml) before the drug treatment, and then they were incubated with Aβ and treated with PBS, aducanumab, and MERLIN. mRNA was isolated after drug treatment. mRNA (100 to 500 ng) was reverse-transcribed to generate complementary DNA. Phagocytosis assay For the phagocytosis of Aβ in vitro, BV2 cells were incubated with Aβ (5 μM) in the presence of PBS and aducanumab (5 μg/ml). The experiment group was treated with MERLIN for 24 hours previously. Then, samples were fixed with 4% paraformaldehyde for 15 min and their membranes were penetrated with 0.3% Triton. In vivo, the mice were divided into three groups (six mice per group) and treated via intrathecal injection 10 times a month with 5 μl of PBS, aducanumab (250 μg/ml), or MERLIN (containing mRNA at 250 μg/ml). After the treatment, behavioral tests were conducted on them blindly. Subsequently, brain tissues were collected and prepared into frozen sections for immunofluorescence analysis. Hippocampal tissues were then harvested for scRNA-seq experiments. Additionally, MERLIN-treated mice (three mice per time point) were sacrificed at 1, 2, 3, and 4 weeks following treatment, and the brain tissues were collected for immunofluorescence analysis. Immunofluorescence staining In immunofluorescence analysis, all frozen sections were incubated with corresponding primary antibodies for 12 hours at 4°C according to different experiments. They were washed with PBST (0.1% Tween 20 in 1× PBS) three times before secondary antibodies were added for 1 hour at room temperature. DAPI or WGA (Invitrogen, 1:1000 dilution) was used for the staining of cell membranes or nuclei on demand. For thioflavin T staining, sections were sequentially washed in 70% and 80% ethanol for 1 min each. They were then stained with a 1% thioflavin T solution in 80% ethanol for 15 min at room temperature. Following staining, sections underwent washes in 80% and 70% ethanol for 1 min each and two washes in distilled water. After a 10-min dehydration process, they were mounted using a mounting medium and imaged with a confocal microscope. All confocal images of brain sections were captured using a Leica TCS Sp8 (Wetzlar, Germany). NOR and NOL tests Both novel object recognition (NOR) and novel object location (NOL) tests were conducted in a custom-made acrylic square box (30 cm by 30 cm by 28 cm, width by depth by height), with one side marked by a stripe for location information. Each task comprised three phases: habituation, training, and testing. For the NOL test, mice were first habituated to the testing environment by allowing free exploration of the open-field arena for 10 min. Twenty-four hours later, mice were introduced to the box containing two identical objects for a 10-min session. During the testing phase, one of the objects was relocated within the box, and the mice were observed for another 10 min. In the NOR test, the process was identical to the NOL test except that relocation was replaced with novel object recognition. Morris water maze test Morris water maze test was conducted in a large circular pool filled with opaque water, where a liftable escape platform was in a fixed location. The experiment included a training phase over five consecutive days and a test phase on the sixth day. For the training period, mice were placed in the water facing the wall of the water maze, and the time and path to reach the escape platform beneath were recorded. In the testing phase, the platform was removed, and the time and path reaching the platform’s location were recorded. Bioinformatic analysis and SER structure prediction For scRNA-seq experiments of drug-treated mice, hippocampal tissues were isolated into single cells, counted, processed, and sequenced following the manufacturer’s instructions. Downstream analyses were performed in R using the Seurat package (v.4.2.0). Clustering and Uniform manifold approximation and projection (UMAP) analysis were performed based on the statistically significant principal components. Data visualizations were performed using Seurat functions DimPlot, DotPlot, and FeaturePlot. Other data visualizations were performed using ggplot2. KEGG, GO, and GSEA analysis were conducted using clusterProfiler and AnnotationHub packages. SER structure was predicted under the construction of AlphaFold2 ([163]24). Statistics and reproducibility All data were analyzed using GraphPad Prism 10 software, with results presented as the mean ± SD. The statistical significance of differences between groups was analyzed by Student’s t test, one-way analysis of variance (ANOVA), or two-way ANOVA. A value of P < 0.05 was considered to indicate statistical significance. Acknowledgments