Next Article in Journal
The Use of Non-Invasive Brain Stimulation for the Management of Chronic Musculoskeletal Pain: Fad or Future?
Previous Article in Journal
Symptom-Triggered Alcohol Detoxification Compared to Fixed-Dose Regimen of Benzodiazepines: A Retrospective Case–Control Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Targeting CX3CR1 Signaling Dynamics: A Critical Determinant in the Temporal Regulation of Post-Stroke Neurorepair

1
Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
2
Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Brain Sci. 2025, 15(7), 759; https://doi.org/10.3390/brainsci15070759
Submission received: 22 June 2025 / Revised: 13 July 2025 / Accepted: 16 July 2025 / Published: 17 July 2025

Abstract

Ischemic stroke ranks among the top global causes of disability and mortality, with a highly dynamic pathological process. Post-stroke neuroinflammation, mediated by microglia, demonstrates a dual role in both injury and repair. The CX3CR1/CX3CL1 signaling axis, highly expressed in microglia, acts as a key regulator. This review examines the spatiotemporal dynamics of the axis across the stroke process and its involvement in neural repair. Crucially, this signaling pathway demonstrates stage-dependent functional duality: its cellular sources, receptor expression profiles, and functional consequences undergo temporally orchestrated shifts, manifesting coexisting or interconverting protective and damaging properties. Ignoring this dynamism compromises the therapeutic efficacy of targeted interventions. Thus, we propose a triple precision strategy of “stroke phase—biomarker—targeted intervention”. It uses specific biomarkers for precise staging and designs interventions based on each phase’s signaling characteristics. Despite challenges like biomarker validation, mechanistic exploration, and cross-species differences, integrating cutting-edge technologies such as spatial metabolomics and AI-driven dynamic modeling promises to shift stroke therapy toward personalized spatiotemporal programming. Temporally targeting CX3CR1 signaling may offer a key basis for developing next-generation precision neural repair strategies for stroke.

1. Introduction

Ischemic stroke represents a predominant global cause of mortality and long-term disability [1], with its core pathophysiology marked by the ischemic cascade—a dynamic sequence of excitotoxicity, oxidative stress, and neuroinflammation following cerebral hypoperfusion [2,3]. Microglia-mediated neuroinflammation exerts dual effects on post-stroke injury and repair [4]. CX3CR1, a fractalkine receptor highly expressed on microglia, plays a key role in modulating microglial activation, migration, and synaptic interactions [5]. By regulating microglial polarization and neuron–glia communication, CX3CR1 signaling significantly impacts neuroinflammation, neuronal survival, and regeneration [6], making it a promising therapeutic target. However, current research often uses static or single-timepoint analyses, failing to capture the pathway’s temporal dynamics [7]. The CX3CR1/CX3CL1 axis exhibits phase-dependent functional duality, with its cellular sources, signaling intensity, and biological consequences evolving across stroke progression, exhibiting both neuroprotective and neurotoxic manifestations at different stages [8]. Interventions ignoring this spatiotemporal dimension risk suboptimal outcomes. This knowledge gap in spatiotemporal dynamics impedes therapeutic advancement.
This narrative review rigorously adheres to the SANRA (Scale for the Assessment of Narrative Review Articles) guidelines. Through a comprehensive literature search in PubMed databases through June 2025, we systematically elucidate the spatiotemporal regulatory mechanisms of the CX3CR1/CX3CL1 signaling axis across the full continuum of ischemic stroke and its central role in neural repair processes. This review analyzes the temporal reprogramming of this signaling pathway across critical pathological phases and proposes a phase-stratified therapeutic framework. We explore innovative approaches using biomarker-guided staging to implement targeted interventions while addressing current research challenges and future translational opportunities. By integrating advances across neuroimmunology and systems biology, this work establishes a temporally sequenced targeting strategy for CX3CR1 modulation, providing a foundational framework for next-generation precision stroke therapeutics.

2. Spatiotemporal Dynamics and Dual Regulatory Roles of the CX3CR1/CX3CL1 Signaling Axis in Post-Stroke Pathology

The pathological progression of ischemic stroke exhibits highly dynamic temporal evolution. Although standardized time boundaries remain undefined in the current literature, it is conventionally categorized as follows [9,10,11,12]: 1. Hyperacute phase: <24 h post-onset, with critical therapeutic windows at ≤4.5 and ≤6 h [13]. 2. Acute phase: 24 h to 7 days [14,15] (some definitions emphasize the early subphase: days 1–3 [16]). 3. Subacute phase: 1 week to 3 months, subdivided into early (1–2 weeks) and late (2 weeks–3 months) stages [17,18]. 4. Chronic phase: >3 months [15,16]. Within this cascade, the CX3CR1/CX3CL1 signaling axis serves as a core molecular driver of pathological phase transitions due to its spatiotemporal-specific expression and functional variations [8,19]. The precise dissection of CX3CR1/CX3CL1 spatiotemporal dynamics requires advanced technologies: single-cell RNA sequencing (scRNA-seq) reveals molecular heterogeneity and temporal evolution of CX3CR1-expressing myeloid subsets post-stroke [20,21,22,23], while spatial transcriptomics captures astrocyte-derived CX3CL1 enrichment gradients within critical repair regions [24,25]. These spatial patterns drive localized activation of CX3CR1+ myeloid cells toward a reparative phenotype [26,27]. Although systematic quantitative data remain limited, convergent qualitative findings from multiple studies deploying these technologies establish the molecular and cellular foundations for elucidating how the CX3CL1/CX3CR1 axis drives its stage-dependent dual functions through spatiotemporally precise regulation.

2.1. Hyperacute Phase: Injury Perception and Emergency Response

Within hours of stroke onset, neurons in the ischemic core undergo rapid disintegration, releasing abundant soluble CX3CL1. This triggers a steep local CX3CL1 concentration gradient that functions as a critical “Find-Me” chemotactic signal to recruit phagocytic cells [6,7,28,29]. Concurrently, resident microglia in the peri-infarct zone undergo rapid but transient upregulation of CX3CR1 expression—a spatiotemporally constrained response essential for damage surveillance [5,8].
This spatiotemporal coordination—where CX3CL1 release in the ischemic core coincides with CX3CR1 upregulation in peri-infarct zones—enables the CX3CR1 pathway to mediate injury surveillance [30,31,32]. Activation of the signaling axis induces the transition of peri-infarct microglia from quiescence to activation. Microglial migratory capacity is significantly enhanced through CX3CR1-AKT signaling [33,34], guiding rapid cellular accumulation toward the injury core. At the lesion site, activated microglia initiate essential neuroprotective functions: cellular debris is phagocytosed [5,8], and hazardous substances are cleared. This process represents the beneficial onset of neuroinflammation, serving to contain damage progression.
However, excessive or sustained activation of this signaling axis at the hyperacute stage underlies its dangerous functional duality [8,35]. Current evidence suggests that functional transition of this axis may be driven by multifactorial mechanisms. We postulate that high concentrations of soluble CX3CL1 could potentially enhance receptor-binding efficiency through signaling properties distinct from membrane-bound ligands, potentially inducing receptor hyperactivation [36]. Concurrently, alterations in receptor conformational states—such as subtle helix VI shifts post-CX3CL1 binding—might modulate signal transduction intensity [37]. Crucially, divergent activation patterns of downstream effectors appear to underlie functional dichotomies: CX3CR1-mediated AKT phosphorylation likely promotes neuroprotective migration during initial signaling [38], while under chronic overactivation, identical phospho-effectors may excessively amplify migratory capacity and potentially trigger pro-inflammatory cascades [39]. This inflammatory shift could recruit monocytes and release mediators [40], presumably elevating local oxidative stress. Collectively, these mechanisms might drive microenvironmental evolution from transient debris-clearing repair toward acute neurotoxic inflammation [41,42], potentially exacerbating secondary brain injury.

2.2. Acute Phase: Dual-Phase Regulation of Neuroinflammation

As stroke pathology progresses into the acute phase (24 h–7 d), the CX3CR1/CX3CL1 axis undergoes critical spatiotemporal reprogramming [43,44,45,46]. Its regulatory function manifests dual-phase characteristics: acting as both an amplifier of inflammatory cascades and an intrinsic brake mechanism on neurotoxic overactivation [47,48,49,50,51].
The ischemic core and penumbral microvascular beds constitute the spatial epicenter of acute-phase pathology. Within these regions, hypoperfusion-induced low shear stress synergizes with early inflammatory mediators (e.g., TNF-α) to trigger robust CX3CL1 upregulation on vascular endothelia [43,44,45]. This endothelial-derived chemokine serves as a potent recruitment signal for CX3CR1-expressing peripheral immune cells—particularly pro-inflammatory Ly6C+ monocytes—directing their infiltration into ischemic parenchyma [46].
Thus, CX3CR1 signaling acts as a direct amplifier of inflammatory storms: Endothelially upregulated CX3CL1 specifically drives peripheral CX3CR1+Ly6C+ pro-inflammatory monocytes to initiate the classical adhesion cascade—including rolling, firm adhesion, and transendothelial migration—efficiently mediating their infiltration into ischemic brain parenchyma [52,53]. These newly recruited Ly6C+ monocytes rapidly expand within the ischemic core and penumbra, differentiating into pro-inflammatory macrophages (monocyte-derived macrophages, MDMs) [54]. Upon cerebral entry, this macrophage-differentiated population becomes the primary instigator of neuroinflammatory storms, dominantly releasing potent pro-inflammatory cytokines such as IL-1β and TNF-α [47,48]. These newly differentiated macrophages significantly amplify neuroinflammatory cascades, exacerbating neuronal injury, blood–brain barrier disruption, and secondary brain tissue damage.
Simultaneously, the CX3CR1 signaling pathway in resident microglia exhibits a unique “braking function”: It specifically activates microglia, driving their polarization toward an M2-like phenotype characterized by anti-inflammatory and neuroprotective properties. This CX3CR1-dependent rapid phenotypic reprogramming induces microglial secretion of key anti-inflammatory factors (e.g., IL-10), significantly suppressing local inflammatory storms and thereby modulating neuroinflammatory progression [49,50,51]. This reprogrammed state is recognized as the initiation point for inflammation self-limitation, actively suppressing inflammatory intensity while promoting tissue repair, thereby constraining early-stage neuroinflammatory exacerbation.
The coexistence of these seemingly opposing effects—pro-inflammatory amplification versus anti-inflammatory braking—epitomizes the bimodal regulatory nature of CX3CR1 signaling during the acute phase. The homeostatic equilibrium of this signaling axis critically depends on spatiotemporal cellular contexts and crosstalk with key pathways—particularly the complex co-regulatory interplay between CX3CR1 and TREM2 (triggering receptor expressed on myeloid cells 2). This synergy manifests through: 1. Co-expression modulation (e.g., synchronized downregulation in metabolic/drug models [55,56]). 2. Signaling pathway intersection (LRRK2-NFATc1 axis [57]). 3. Functional complementarity (TREM2-mediated phagocytosis promotion vs. CX3CR1-driven suppression of inflammatory hyperactivation [58,59]). These mechanisms co-regulate microglial phagocytic activity and jointly modulate disease-associated phenotypes—including DAM (disease-associated microglia) transition [60,61]—and the inflammation–phagocytosis equilibrium. The dynamic equilibrium is ultimately determinative for acute neuroinflammation magnitude/duration, directly influencing neuronal survival in the ischemic penumbra and overall tissue damage extent [62,63].

2.3. Subacute Phase: Neural Repair Activation

During the subacute phase (1 week to 3 months post-stroke), neuroinflammation gradually subsides while tissue repair mechanisms initiate. The CX3CR1/CX3CL1 axis undergoes functional repurposing: its spatiotemporal reprogramming primarily promotes neural repair [64,65,66], yet insufficient signaling intensity may constitute a novel therapeutic challenge [67,68,69,70].
As acute inflammation resolves, activated astrocytes—particularly in peri-infarct zones—supersede vascular endothelia as the dominant CX3CL1 source [5,71]. This astrocyte-dominated signaling marks a spatial shift from inflammatory to reparative programs. Within lesioned areas (ischemic core and periphery), microglia/macrophages exhibiting sustained CX3CR1 expression emerge as primary responders to astrocyte-derived CX3CL1, executing neural repair functions [72]. Here, microglia function as dominant regulators—leveraging their CNS-resident immunity to directly drive tissue repair through anti-inflammatory polarization, neuroprotection, and angiogenesis [73,74]. In parallel, macrophages—particularly MDMs—operate as auxiliary units that secrete anti-inflammatory mediators (e.g., IL-13) to dampen inflammation and bolster neuroregeneration, thereby reinforcing microglia-led reparative cascades [75,76].
During this phase, the CX3CR1/CX3CL1 axis centrally coordinates multifaceted repair processes: 1. Debris clearance: CX3CR1 signaling drives microglia/macrophages toward a pro-phagocytic phenotype that efficiently clears injury residues (e.g., myelin debris), establishing a permissive microenvironment for remodeling [6,66]. 2. Angiogenesis induction: CX3CR1-high macrophages secrete VEGF, directly stimulating vascular remodeling and perfusion recovery in peri-infarct zones [77]. 3. Neurogenic support: Neuron-/glial-derived CX3CL1 specifically activates neural progenitor cells (NPCs) via CX3CR1 [78,79,80], enhancing survival and promoting endogenous neurogenesis for circuit reconstruction [63,64,65,66,81]. At the molecular level, extracellular regulated protein kinases (ERK) pathway activation downstream of CX3CR1 constitutes the core mechanism mediating these repair functions [80]. ERK upregulates neurotrophic factor release [64,81], supporting neuronal survival, axonal regeneration, and synaptic plasticity.
However, dysfunction or disruption of the CX3CR1/CX3CL1 axis during this repair-oriented shift carries significant risks. Impaired signaling (e.g., CX3CR1 inhibition or insufficient CX3CL1) may trigger maladaptive astrocyte activation [67], culminating in dense gliotic scar formation [68]. These scars severely impede axonal regeneration, synaptic remodeling, and tissue reintegration [69,70], ultimately compromising long-term functional recovery potential.

2.4. Chronic Phase: Perpetuating Inflammation and Secondary Degeneration

During the chronic phase (>3 months post-stroke), persistent tissue remodeling occurs and neurological sequelae are manifested [63]. The CX3CR1/CX3CL1 axis exhibits spatiotemporally sustained high-level activation—prolonging subacute-phase neurorestorative effects during early stages [4], yet its chronic hyperactivation triggers a paradoxical functional shift: transitioning from pro-repair potential to a driver of persistent neurological deterioration [82].
During this phase, surviving neurons in lesioned cores and adjacent regions, alongside activated astrocytes, persistently produce CX3CL1 [24,83,84] with diffuse spatial distribution. Microglial CX3CR1 expression is stably maintained at elevated levels [85,86], establishing these cells as the primary chronic responders to sustained CX3CL1 signaling.
Sustained CX3CR1 hyperactivity at this stage transitions from reparative to pathogenic, becoming a core driver of chronic neuroinflammation, circuit damage, and secondary neurodegeneration. Its multifaceted destructive mechanisms include the following: 1. Pathological synaptic pruning: Elevated CX3CR1 signaling abnormally activates microglial phagocytosis and complement cascades [55,87], inducing excessive elimination of synaptic components through uncontrolled pruning. This results in widespread synaptic loss [88,89,90], disrupting neural circuit integrity. 2. Chronic pro-inflammatory microenvironment: CX3CR1 drives persistent microglial pro-inflammatory polarization [51,91,92], establishing self-perpetuating neurotoxic microenvironments that amplify neural damage. 3. Accelerated proteinopathy: CX3CR1 hyperactivity accelerates pathological protein aggregation (e.g., amyloid β protein [Aβ] deposition) [93]. Conversely, CX3CR1 deficiency delays Aβ pathology in experimental models [94], indicating its role in promoting post-stroke Alzheimer’s-like neurodegeneration.
Chronic-phase CX3CR1 signaling dysfunction correlates directly with long-term neurocognitive outcomes in stroke survivors. Human genetic studies confirm that the CX3CR1 T280M loss-of-function polymorphism significantly elevates risk for post-stroke cognitive impairment and dementia [63,95,96,97]. This evidence achieves the following: 1. Validates CX3CR1’s central role in maintaining neural circuit integrity and resisting neurodegeneration. 2. Highlights the necessity for precise chronic-phase regulation to prevent detrimental neurocognitive outcomes. These clinical data establish CX3CR1 signaling as a critical molecular determinant of long-term neurological prognosis after stroke.
The spatiotemporal reprogramming and functional duality of the CX3CR1/CX3CL1 signaling axis across stroke progression are systematically characterized in Table 1:
It is now well established that the CX3CR1 signaling axis plays a stage-dependent dual role following stroke [8,36]. However, current research predominantly addresses phenomenological observations, leaving critical mechanistic knowledge gaps: 1. Ligand concentration threshold: How does soluble CX3CL1 (sCX3CL1) switch functional states via concentration gradients during the hyperacute phase? 2. Receptor conformational reprogramming: Do post-stroke CX3CR1 post-translational modifications (e.g., phosphorylation/glycosylation) influence signaling bias? 3. Co-receptor interaction networks: How does spatiotemporal co-localization with receptors like TREM2 and CCR2 modulate CX3CR1 signal output? 4. Downstream signal sorting: Why does the same receptor activate pro-inflammatory versus neuroreparative pathways across distinct pathological stages? Addressing these mechanistic voids represents an urgent research priority. Notably, partial mechanisms have been elucidated. The CX3CL1/CX3CR1 axis confers neuroprotection by inhibiting NLRP3 (NOD-like receptor thermal protein domain associated protein 3) inflammasome activation [6] through two complementary pathways: 1. PPM1A (magnesium-dependent phosphatase 1 A) phosphatase-mediated suppression: CX3CR1 signaling activates PPM1A, which binds NLRP3 to disrupt its complex formation with ASC (apoptosis-associated speck-like protein containing a CARD) and pro-caspase-1, directly blocking inflammasome assembly [98]. 2. NF-κB (nuclear factor kappa-B) pathway inhibition: CX3CR1 activation suppresses NF-κB signaling, reducing pro-inflammatory cytokine release and indirectly impeding NLRP3 oligomerization [99]. Conversely, NLRP3 inflammasome activation may exacerbate neuroinflammation, counteracting CX3CR1-mediated neuroprotection [6]. This reveals a bidirectional regulatory circuit—the PPM1A/NF-κB/CX3CR1-NLRP3 axis—that orchestrates stage-specific transitions between pro-inflammatory and anti-inflammatory/repair responses post-stroke. Future studies must comprehensively dissect CX3CR1’s core regulatory mechanisms to fully decode its stage-dependent duality.

3. Phase-Adapted Therapeutic Strategies Targeting CX3CR1

Given the stage-dependent functional duality of CX3CR1 signaling and its dynamic evolution across stroke pathology, therapeutic interventions must adhere to strict temporal precision. This section synthesizes advances in time-window-specific targeting strategies, examining key technological innovations and clinical translation pathways for phase-adapted modulation of the CX3CR1 axis.

3.1. Timeliness Challenges of Existing Intervention Strategies

Current evidence indicates that systemic CX3CR1 antagonism during the hyperacute phase compromises microglial neuroprotection by inhibiting migration and polarization toward acute surveillance phenotypes [100,101,102]. Critically, such intervention exacerbates early blood–brain barrier (BBB) disruption [103,104,105]. These findings collectively demonstrate that CX3CR1-targeted interventions during this phase may not only impair microglia-mediated neuroprotective mechanisms but also accelerate neural injury cascades and pathological progression.
During the acute phase, CX3CR1 antagonism significantly suppresses microglial phagocytic function [51,106], resulting in the pathological accumulation of necrotic neuronal debris within injured areas [107,108]. This debris accumulation amplifies neuroinflammatory cascades and exacerbates secondary tissue damage [38,51]. Consequently, CX3CR1 inhibition during this phase not only fails to modulate inflammation effectively but also intensifies secondary neural injury by impairing dead cell clearance.
The administration of exogenous CX3CL1 agonists (e.g., recombinant CX3CL1 [rCX3CL1] protein or fractalkine [FKN]-Fc fusion protein) during the subacute phase effectively induces microglial polarization toward a reparative phenotype [29,109]. This strategy enhances microglial clearance efficiency for hematomas and cellular debris while promoting vascular remodeling and improving functional recovery [6,110]. These findings establish the subacute phase as a critical therapeutic window for CX3CR1 pathway activation to maximize microglia-mediated neuroprotection.
While sustained, long-term CX3CR1 inhibition during the chronic phase reduces neuroinflammatory responses [111], concurrent suppression of essential synaptic remodeling and neural regeneration processes occurs [82,112]. Murine model studies confirm that cognitive recovery is significantly delayed by prolonged CX3CR1 pathway antagonism [46]. Thus, chronic, long-term monotherapy targeting CX3CR1 may alleviate inflammation while inducing inhibition of neural plasticity, ultimately resulting in hindered functional recovery.
Collectively, this evidence confirms that CX3CR1-targeted stroke therapies must incorporate strict temporal specificity. Interventions at non-optimal timepoints—such as pathway inhibition during hyperacute/acute phases or simplistic chronic-phase suppression—risk inducing counterproductive outcomes. Conversely, pathway activation during the subacute phase maximizes reparative potential. These findings underscore the imperative for temporally programmed therapeutics.

3.2. Biomarkers Drive Precision Staging

In stroke management, precise pathological staging is fundamental for targeted CX3CR1/CX3CL1 modulation and essential for elucidating disease mechanisms, optimizing therapies, and predicting outcomes. Conventional staging—primarily based on symptom onset and imaging findings [113]—is limited by inadequate sensitivity [114] and temporal lag [115]. Emerging stage-specific biomarkers, which directly mirror molecular pathology, are now driving a paradigm shift toward precision staging characterized by enhanced accuracy, objectivity, and personalization.
During stroke’s hyperacute phase, blood–brain barrier (BBB) disruption constitutes the core pathological event [116,117]. Serum matrix metalloproteinase-9 (MMP-9) levels exhibit a sharp increase during this phase [118,119]. MMP-9 specifically degrades endothelial tight junction proteins (e.g., occludin and zonula occludens-1) and type IV collagen in the basement membrane while activating other protease systems [120,121,122], functioning as a key effector of BBB damage [118,121]. Concurrently, CX3CL1 mediates microvascular endothelial barrier dysfunction through the Src/P115-RhoGEF/ROCK pathway [123]. Therefore, dynamic changes in serum MMP-9 and CX3CL1 levels provide sensitive humoral markers for detecting hyperacute-phase BBB injury.
As previously stated, the CX3CR1 and TREM2 pathways exhibit precise synergistic regulation that jointly controls acute neuroinflammatory dynamics after stroke [124]. Stroke model studies demonstrate that TREM2 functional defects directly inactivate microglial phagocytic capacity [125,126]. This dysfunction consistently coincides with significant downregulation of the scavenger receptor CD36 [127,128], thereby impairing CD36-mediated phagocytic clearance—confirming TREM2’s essential role in sustaining this critical process. Critically, Kota Kurisu et al. revealed that TREM2 deficiency extends beyond microglia to impair phagocytic function in myeloid-derived immune cells, including post-stroke infiltrating macrophages [129], establishing TREM2’s central regulatory role in myeloid responses. Given TREM2’s pivotal regulation of microglial activity and neuroinflammatory cascades, its expression dynamics offer a critical window into neuroinflammatory states. Current cerebrospinal fluid (CSF) TREM2 measurements have generated substantial biomarker data [122,130], supporting its potential use as a bodily fluid biomarker for assessing post-stroke inflammatory responses and pathological burden.
Non-invasive imaging techniques now enable in vivo dynamic monitoring of microglial responses. Lucio D’Anna et al. utilized the PET tracer [11C]PBR28 targeting the 18 kDa translocator protein (TSPO)—highly expressed on microglia/macrophage mitochondria—demonstrating significantly elevated tracer uptake in subacute-phase ischemic stroke lesions [131]. This directly visualizes inflammatory cell aggregation. Complementarily, Alessandro Villa et al. synthesized a novel carbon-11-labeled P2RY12-targeted PET tracer [132], leveraging P2RY12 upregulation during anti-inflammatory responses. This tracer provides a potential tool for precisely assessing microglial subpopulations with reparative phenotypes in the subacute phase. Elevated P2RY12 expression serves as a key molecular signature of microglial “homeostatic” or “reparative” states, correlating with phagocytic clearance and neuroprotective functions [133,134]. Notably, CX3CR1 signaling acts as a core molecular switch driving microglial transition toward this reparative phenotype during this phase [135]. Activation of pathways such as PI3K-Akt likely constitutes a critical mechanism underlying its repair-promoting functions—including pro-angiogenic effects and neurotrophic factor release [136]. Collectively, these advances reinforce the consensus that finely regulated microglial responses are indispensable for neural tissue repair and remodeling during stroke’s subacute phase [137,138].
Neurofilament light chain (NfL) represents the gold standard biomarker for neuronal axonal injury. Its levels in CSF or serum exhibit significant elevation across neurological disorders including stroke, with established utility as a key indicator of axonal damage [139,140,141]. Notably, Magnus Gisslén et al. demonstrated that in neurological diseases (e.g., HIV-associated neurocognitive disorders), CSF soluble TREM2 (sTREM2) levels increase with disease severity while independently correlating with NfL levels reflecting neuronal injury [142]. This indicates that TREM2 signaling and microglial function critically modulate neuronal damage and repair mechanisms. Further evidence reveals that TREM2-mediated microglial activity participates in synaptic loss regulation during brain injury and disease [143], providing key insights into the molecular basis of post-stroke neurological deficits and recovery. Substantial evidence indicates that sustained CX3CR1 overactivation during the chronic phase constitutes a core pathological mechanism: It facilitates microglia-mediated pathological synaptic pruning (through phagocytosis of synaptic components) [144,145], maintains pro-inflammatory microenvironments [51,146], and may accelerate amyloid pathology [93], ultimately causing neuronal damage manifested by elevated NfL. Dynamic NfL monitoring combined with complementary biomarkers (e.g., sTREM2) will enable precision staging of neurological damage severity and prediction of long-term recovery potential.
In summary, biomarkers across stroke progression provide multidimensional molecular evidence for precise pathological staging: 1. Hyperacute phase: MMP-9 and CX3CL1 as early-warning markers of BBB disruption. 2. Acute phase: TREM2 reflecting pro-/anti-inflammatory dynamics. 3. Subacute phase: TSPO/PET imaging capturing microglial/macrophage activation/repair status, alongside emerging P2RY12-targeted PET tracers. 4. Chronic phase: NfL quantifying axonal damage and neural recovery. These biomarkers collectively unveil core stage-specific pathophysiological mechanisms while driving personalized therapeutic strategies, thereby establishing a foundation for biomarker-guided precision staging and targeted interventions in stroke management.

3.3. Cutting-Edge Technologies Enable Precise Interventions

Emerging technologies have now overcome the core spatiotemporal constraints of traditional stroke drug delivery by enabling lesion-specific targeting and chronotherapeutic delivery control, thus making precision interventions feasible.
pH-responsive nanoparticles enable targeted delivery of CX3CR1 modulators (agonists/antagonists) to the acidic microenvironment of ischemic brain regions [147,148,149,150,151]. This system triggers localized drug release under acidic pH conditions, significantly enhancing drug concentration at lesion sites while reducing systemic exposure and toxicity risks [152,153]. These findings demonstrate that mimicking pathological microenvironment characteristics achieves spatiotemporal-specific drug delivery, thereby overcoming limitations of traditional systemic administration in dynamically regulating CX3CR1 function.
Adeno-associated virus (AAV)-based gene regulation systems enable conditional knockout of the CX3CR1 gene in microglia within defined time windows (e.g., post-acute phase) through light-controlled or chemically inducible promoters [101,154,155]. This strategy effectively avoids impairing CX3CR1’s protective functions during hyperacute/acute phases while precisely inhibiting chronic-phase pathogenic signaling [63,86,156], achieving spatiotemporal gene expression control. Collectively, gene editing coupled with inducible systems creates a tunable molecular switch for dynamically regulating CX3CR1’s dual neuroprotective/neurotoxic effects.
Moreover, novel CX3CR1-targeted monoclonal antibodies [157] and aptamers [158]—characterized by small molecular weight, high stability, and low immunogenicity [159,160]—exhibit highly specific CX3CR1-modulating capabilities with promising translational potential. These molecules have demonstrated utility in cancer immunotherapy [157] and inflammatory regulation [158], positioning them as promising candidates for future stroke therapeutics targeting CX3CR1.

3.4. Clinical Translation: Triple Precision Strategy

Integrating stroke pathology staging, biomarker profiling, and targeted interventions, we propose a triple precision strategy for clinical translation:

3.4.1. Hyperacute Phase: sCX3CL1/MMP-9 + Vascular Recanalization/Immune Regulation

Biomarkers-guided: Elevated serum levels of sCX3CL1 and MMP-9 serve as critical biomarkers for predicting blood–brain barrier disruption risk [118,121,123]. Dynamic monitoring of these biomarkers enables identification of the optimal therapeutic window for CX3CR1-targeted interventions, thereby preventing exacerbation of secondary neural damage while supporting vascular recanalization and immune homeostasis restoration.
Precision intervention strategies: The core therapeutic challenge in the hyperacute phase centers on balancing vascular recanalization with inflammation control, rather than direct CX3CR1 targeting. Current evidence consistently establishes vascular recanalization—including intravenous thrombolysis within 4.5 h or endovascular thrombectomy within 6 h [161,162]—as the primary intervention [163]. Complementarily, immunomodulatory therapies targeting specific pro-inflammatory mediators (e.g., TNF-α, IL-1β) are confirmed as essential adjunctive strategies during this phase [164,165,166]. Thus, the optimal approach prioritizes vascular recanalization as the cornerstone treatment, augmented by short-term, non-CX3CR1-targeted immunomodulation to suppress excessive early inflammatory cascades.

3.4.2. Acute Phase: sTREM2/CD36 + rCX3CL1 Nanocarrier Delivery

Biomarkers-guided: Based on our earlier conclusions, significant reductions in CSF levels of sTREM2 and scavenger receptor CD36 serve as validated biomarkers for microglial phagocytic dysfunction [125,126,127,128]. The downregulation of sTREM2/CD36 not only reflects TREM2 pathway dysfunction but also indirectly suggests potential failure of CX3CR1 signaling to effectively drive microglial transition toward a high-efficiency phagocytic phenotype—or the predominance of its pro-inflammatory functions [167,168]. Consequently, monitoring sTREM2/CD36 dynamics offers a crucial window for assessing whether the CX3CR1 pathway resides in a repair-favorable “braking state” that promotes anti-inflammatory/pro-phagocytic phenotypes.
Precision intervention strategies: At precisely this crucial window of CX3CR1 reparative insufficiency, we propose exogenous augmentation of CX3CR1 signaling to directly counteract its dysfunction. This intervention drives microglia/macrophage polarization toward a pro-phagocytic, anti-inflammatory phenotype, thereby removing necrotic debris and curbing inflammatory cascade amplification. To achieve spatial precision, pH-responsive nanocarriers [169,170,171] enable targeted delivery of rCX3CL1. These systems not only efficiently traverse the compromised blood–brain barrier [172] but also trigger localized drug release within the acidic microenvironment of the ischemic penumbra [173,174]. This microenvironment-responsive mechanism maximizes local drug concentration, potentiates reparative signaling pathway activation, and minimizes systemic adverse effects.

3.4.3. Subacute Phase: PET-MRI PBR28/P2RY12 + FKN-Fc/MSCs

Biomarkers-guided: When PET-MRI dual-modality imaging reveals significantly elevated [11C]PBR28 uptake in lesioned areas alongside increased microglial P2RY12 expression, this biomarker profile signifies reparative microglial activation [131,132] and marks the transition to the subacute disease phase [137,138]. Concurrently, the CX3CR1 pathway resides within a therapeutically actionable “time window” during which it can be effectively activated to promote neural repair [135]. Dynamic monitoring of these parameters enables precise identification of the endogenous repair initiation window, providing the basis for combined therapeutic strategies that maximize synergistic effects between endogenous mechanisms and exogenous interventions.
Precision intervention strategy: Mesenchymal stem cells (MSCs) demonstrate significant pro-angiogenic capacity [175,176], with confirmed therapeutic efficacy via intravenous administration during the stroke’s subacute phase [177,178]. Building on this evidence and prior conclusions, a combined intervention strategy can be implemented during this phase to maximize synergistic effects: intravenous co-administration of FKN-Fc fusion protein with MSCs [179]. This approach aims to exogenously enhance CX3CR1 signaling, synergizing with endogenous repair signals to consolidate and amplify microglial reparative phenotypes. Concurrently, leveraging MSC paracrine actions, it coordinately promotes angiogenesis, neurogenesis, and tissue remodeling [180,181,182], ultimately accelerating functional recovery in the subacute phase.

3.4.4. Chronic Phase: sTREM2/NfL + AAV Delivery of CRISPR-Cas9

Biomarkers-guided: CSF levels of sTREM2, as a cleavage product of TREM2, serve as a potential biomarker for microglia-mediated synaptic damage induced by signaling pathways such as neuroinflammation [142,143]. Concurrently, elevated NfL levels directly correlate with neuronal damage pathology [139,140,141]. Therefore, persistently elevated sTREM2 levels in CSF coupled with aberrant NfL elevation collectively signal ongoing synaptic injury [183] and chronic neuroinflammation [184]. This biomarker profile indicates that CX3CR1 signaling has transitioned from its early protective/reparative role into a chronic pathogenic driver [93,144,185]. Dynamic monitoring of CSF sTREM2 and NfL levels provides a critical temporal window reference for targeted chronic-phase therapy. Within this window, reversible gene regulation strategies can be applied using AAV vector technology to achieve precise reprogramming of microglial functional homeostasis.
Precision intervention strategies: Chronic-phase interventions must overcome prior limitations of purely inhibitory approaches. Upon detecting this detrimental biomarker shift (sTREM2↑ + NfL↑), precisely and reversibly inhibiting CX3CR1 signaling emerges as a theoretically viable strategy. This intervention aims to block its pathogenic drivers—pathological synaptic pruning, chronic neuroinflammation, and neurodegeneration—thereby preserving neural circuits and improving long-term prognosis. Critical research demonstrates that the HEXB gene’s natural/minimal promoter drives specific and stable expression in microglia under physiological and pathological conditions, making it ideal for CNS-targeted AAV therapy [186]. The team led by Minmin Luo pioneered the AAV-MG1.2 vector, which effectively delivers CRISPR-Cas9 for microglial gene knockout [187]. Concurrently, studies confirm the HEXB locus enables tamoxifen-induced Cre recombinase-dependent gene manipulation [188]. Building on these findings, we propose a chronic-phase strategy: use AAV-MG1.2 vectors carrying the HEXB promoter-driven CRISPR-Cas9 system to target CX3CR1. Tamoxifen induction then achieves spatiotemporal-specific, reversible CX3CR1 knockout—suppressing pathological overactivation while preserving basal physiological functions. While AAV-CRISPR targeting of CX3CR1 holds significant therapeutic promise, its broad expression in peripheral monocytes [189] and specific neuronal populations [190] necessitates vigilance against potential systemic off-target effects (e.g., immune dysregulation) [191,192] and perturbation of compensatory chemokine pathways (e.g., CCR2 upregulation) [193]. Consequently, optimizing delivery systems and dosage parameters represents a critical research priority.
The proposed clinical translation framework is systematically summarized in Table 2:

4. Challenges and Future Directions

4.1. Bottlenecks in Clinical Translation

Implementing disease-stage-based precision interventions faces a core challenge: translating candidate biomarkers from mechanistic research to clinical application. The feasibility of this review’s advocated “Triple Precision Strategy” critically depends on the real-time accuracy of biomarker monitoring and clearly established pathological-stage thresholds within the framework. However, the stroke field currently lacks authoritative standardized definitions and detection thresholds for key biomarkers, as validated through large-scale clinical cohorts. Moreover, stage-specific therapeutic challenges manifest with distinct priorities: Hyperacute phase interventions carry significant immunosuppression risks [194,195], where imprecise systemic cytokine modulation may compromise host defense mechanisms [196,197]; chronic phase management faces predominant delivery hurdles—illustrated by fibrotic scarring in infarct cores that severely impairs vector penetration [198,199] despite AAV-MG1.2 vector’s microglia-targeting capability; ultimately, gene-editing technologies employed in precision interventions inevitably raise complex ethical considerations. Although the “Triple Precision Strategy” theoretically enables accurate staging across stroke phases, its clinical implementation faces significant practical constraints—including reliance on specialized instrumentation, time-intensive procedures, and substantial costs. Consequently, translating this strategy requires critical balancing of diagnostic precision against operational efficiency and economic feasibility. Future research should develop cost-effective implementation protocols that maintain staging accuracy while enhancing real-world deployability.
Beyond the critical knowledge gaps in core CX3CR1 signaling mechanisms previously noted, significant uncertainties persist in upstream regulatory pathways governing this axis [8]. Four fundamental knowledge gaps require urgent resolution: 1. Cell-type-specific transcriptional control identifying core transcription factors and regulatory networks driving CX3CR1 expression across distinct cellular contexts (monocyte subsets, microglia, NK cells, neurons). 2. Transcriptional network plasticity elucidating how regulatory components dynamically adapt to cellular state transitions (resting → activated) and microenvironmental cues (inflammatory/metabolic signals). 3. Chemokine receptor crosstalk defining molecular mechanisms (heterodimerization, co-stimulation, or mutual inhibition) enabling functional integration with receptors like CCR2/CCR5. 4. Ligand-induced signaling diversification determining signal characteristics (intensity thresholds, duration) and decoding mechanisms underlying functional bias in CX3CL1/CX3CR1-mediated multifunctionality (chemotaxis, stable adhesion, cell survival/anti-apoptosis) [200,201]. These mechanistic gaps constitute critical barriers to therapeutic translation.
Meanwhile, current CX3CR1 research faces significant interspecies temporal scale differences in clinical translation [202]. CITE-seq (Cellular Indexing of Transcriptomes and Epitopes by Sequencing) analysis reveals distinct expression patterns of CX3CR1 across T-cell differentiation gradients in humans versus mice. In humans, CX3CR1 expression strongly correlates with CD8+ T-cell differentiation status and migratory/functional properties, serving as a key discriminator between CD8+ and CD4+ T-cell states. However, murine CX3CR1 primarily mediates microglial functions including neuroinflammation modulation and phagocytic activity [203]. Further investigation of human immune disorders uncovers the dynamic complexity of the CX3CL1/CX3CR1 axis: In systemic lupus erythematosus (SLE), CX3CR1 expression in B cells/plasmablasts is negatively regulated by Notch signaling [204]—a mechanism absent in murine models, indicating broader immune regulatory hierarchies in humans. Additionally, the CX3CR1-dependent neural repair window identified in rodent models [205] may not directly translate to human stroke patients due to longer disease courses with greater pathophysiological complexity. Cross-disease evidence—including systemic inflammatory disorders [206], low-grade gliomas (LGGs) [207], subarachnoid hemorrhage (SAH) [208], and aortic aneurysms (AAs) [40])—has elucidated the clinical translational potential of CX3CR1 through single-cell transcriptomics, genome-wide association studies (GWAS), or postmortem tissue analyses; however, direct clinical evidence defining optimal therapeutic time windows for CX3CR1-targeted interventions in ischemic stroke remains limited. To precisely define the human brain repair critical window, advanced models must be developed that authentically recapitulate human neuropathological features. Therefore, a research system integrating human brain organoids with high-resolution in vivo imaging technology [209] will provide a powerful tool for simulating the unique spatiotemporal dynamics of neural repair in the human brain.
The core scientific challenges surrounding CX3CR1 constitute critical bottlenecks impeding the clinical translation of stroke precision therapeutics. Overcoming these barriers demands a multidimensional integrated strategy: 1. Conduct rigorous large-scale clinical studies to validate and clinically implement key biomarkers. 2. Integrate dynamic CD4+/CD8+ ratio monitoring [210] into clinical protocols to control immunosuppression risks. 3. Explore novel blood–brain barrier-opening technologies (e.g., ultrasound-mediated approaches [211,212]) to overcome vector delivery hurdles. 4. Establish reversible editing frameworks (e.g., degron-tagged Cas9 [213,214]) with strict prohibition of germline editing [215,216] to conform with ethical norms. 5. Employ advanced molecular/cellular techniques for mechanistic dissection of regulatory networks—particularly receptor signaling crosstalk and functional signal preference. 6. Develop cross-validated humanized models that accurately simulate human pathophysiology to bridge interspecies gaps and define precise brain-repair therapeutic windows. These systematic endeavors will establish the scientific foundation for implementing the CX3CR1-based triple precision strategy in stroke patients.

4.2. Breakthroughs in Cutting-Edge Technologies

Spatial metabolomics technology enables high-resolution dynamic mapping of key metabolites (e.g., lactate) within post-stroke infarct marginal zones [217,218]. Integrating in situ mass spectrometry imaging with metabolic flux analysis, this approach constructs three-dimensional (3D) dynamic atlases of metabolic microenvironments essential for deciphering functional coupling mechanisms. These atlases reveal how lactate fluctuations regulate local HIF-1α signaling and CX3CR1 activity [219], thereby driving the temporal transition of microglia from pro-inflammatory to reparative phenotypes. Consequently, spatial metabolomics holds significant promise for elucidating the spatiotemporal logic governing metabolic reprogramming and neuroimmune signal crosstalk, thereby identifying novel intervention targets for future CX3CR1 dynamic function regulation through energy metabolic pathway modulation.
Breakthroughs in deep learning algorithms—particularly graph neural networks and Transformer architectures—provide powerful computational tools with nonlinear modeling capabilities for integrating massive heterogeneous multi-omics data (including single-cell transcriptomics, spatial proteomics, metabolomics, and multimodal imaging) [220,221]. Dynamic predictive models constructed from such data can learn patient-specific temporal data streams to accurately predict individualized post-stroke CX3CR1 signaling trajectories and identify optimal intervention time windows. This has catalyzed the innovative “artificial (AI)-enabled neuroimmune clock” framework, whose core lies in identifying critical dynamic signatures of neuroimmunological interactions, such as key transition nodes in microglial state transitions [222,223]. This framework establishes both theoretical foundations and precise temporal coordinates for personalized neural repair strategies. Deep learning is driving a fundamental paradigm shift from static omics mapping to dynamic mechanism modeling, ultimately enabling high-dimensional spatiotemporal navigation of neuroimmunoregulation.
In summary, technological innovations now achieve breakthroughs through dual-path empowerment integrating spatial dimensions (metabolic microenvironment profiling) and temporal dimensions (dynamic modeling/prediction). By synergizing these technologies—where spatial metabolomic maps provide high-precision physiological substrates for AI-enabled neuroimmune clocks, while critical kinetic features derived from clock analyses feedback into spatiotemporal optimization of metabolic targets—we will establish a closed-loop system spanning metabolic remodeling to precise cellular state navigation. This will orchestrate a paradigm transition from “stage-dependent” to “individualized spatiotemporal programming” for stroke neural repair.
The above translation challenges and technical breakthroughs can be summarized in Table 3:

5. Conclusions

This review delineates the core pathophysiological signature of stroke-induced CX3CR1/CX3CL1 signaling: its spatiotemporally evolving dual neurotoxic/neuroreparative effects. The stage-specific spatiotemporal reprogramming of signaling sources and cellular targets critically steers pathological progression and neurological outcomes. Consequently, effective therapeutic interventions must implement spatiotemporally precise modulation to suppress detrimental pathways while amplifying reparative functions, thereby optimizing neurological recovery and preventing chronic neurodegeneration. This review accordingly proposes a “Triple Precision Strategy” clinical translation framework integrating stroke phase stratification, biomarker profiling, and targeted interventions. However, CX3CR1-targeted therapy’s clinical translation faces persistent bottlenecks: inadequate validation of biomarkers, incomplete elucidation of upstream regulatory mechanisms, and clinically significant interspecies differences. Technological innovations are now breaking through these barriers: spatial profiling technologies decode neuroimmune spatiotemporal logic, while AI-driven dynamic modeling predicts critical intervention points for personalized therapy. Consequently, the deep integration of these technological directions will establish a closed-loop regulatory system, driving the convergence of intervention timing, biomarker-guided staging, and advanced delivery/gene regulation technologies in CX3CR1-targeted therapy. This synergy will ultimately establish a paradigm shift—from stage-dependent protocols toward individualized spatiotemporal precision programming in stroke treatment. In summary, the CX3CR1/CX3CL1 signaling axis serves as the central regulatory mechanism for post-stroke brain repair. Precision modulation of this axis holds transformative potential as a fundamental therapeutic strategy for stroke recovery. Consequently, sustained technological refinement targeting this pathway represents a critical research imperative moving forward.
As this review adopted a narrative review methodology without strict adherence to PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) protocols, it may not exhaustively encompass all of the relevant literature. Consequently, readers should interpret the conclusions with critical discernment. Nonetheless, through synthesizing cross-disciplinary evidence and contentious viewpoints, this work constructs an innovative framework for CX3CR1-targeted stroke therapeutics. Our analysis uncovers pivotal knowledge gaps and paradoxical findings, establishing a conceptual foundation for future mechanistic investigations and clinical translation of CX3CR1 pathway modulation.

Author Contributions

Conceptualization, Q.H. (Quan He) and T.Z.; methodology, Q.H. (Quanwei He); validation, Q.H. (Quan He), T.Z., and Q.H. (Quanwei He); formal analysis, Q.H. (Quan He) and T.Z.; writing—original draft preparation, Q.H. (Quan He); writing—review and editing, Q.H. (Quan He) and T.Z.; supervision, Q.H. (Quanwei He). All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

This work was supported by grants 82071335 from the Program of National Natural Science Foundation of China to Quanwei He.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Yu, C.C.; Liu, L.B.; Chen, S.Y.; Wang, X.F.; Wang, L.; Du, Y.J. Ancient Chinese Herbal Recipe Huanglian Jie Du Decoction for Ischemic Stroke: An Overview of Current Evidence. Aging Dis. 2022, 13, 1733–1744. [Google Scholar] [CrossRef]
  2. Majumder, D. Ischemic Stroke: Pathophysiology and Evolving Treatment Approaches. Neurosci. Insights 2024, 19, 26331055241292600. [Google Scholar] [CrossRef] [PubMed]
  3. Qin, C.; Yang, S.; Chu, Y.H.; Zhang, H.; Pang, X.W.; Chen, L.; Zhou, L.Q.; Chen, M.; Tian, D.S.; Wang, W. Signaling pathways involved in ischemic stroke: Molecular mechanisms and therapeutic interventions. Signal Transduct. Target. Ther. 2022, 7, 215. [Google Scholar] [CrossRef] [PubMed]
  4. Lian, L.; Zhang, Y.; Liu, L.; Yang, L.; Cai, Y.; Zhang, J.; Xu, S. Neuroinflammation in Ischemic Stroke: Focus on MicroRNA-mediated Polarization of Microglia. Front. Mol. Neurosci. 2020, 13, 612439. [Google Scholar] [CrossRef] [PubMed]
  5. Zhang, Q.; He, L.; Chen, M.; Yang, H.; Cao, X.; Liu, X.; Hao, Q.; Chen, Z.; Liu, T.; Wei, X.E.; et al. PSD-93 mediates the crosstalk between neuron and microglia and facilitates acute ischemic stroke injury by binding to CX3CL1. J. Neurochem. 2021, 157, 2145–2157. [Google Scholar] [CrossRef]
  6. Ge, Y.; Wang, L.; Wang, C.; Chen, J.; Dai, M.; Yao, S.; Lin, Y. CX3CL1 inhibits NLRP3 inflammasome-induced microglial pyroptosis and improves neuronal function in mice with experimentally-induced ischemic stroke. Life Sci. 2022, 300, 120564. [Google Scholar] [CrossRef]
  7. Cao, X.W.; Yang, H.; Liu, X.M.; Lou, S.Y.; Kong, L.P.; Rong, L.Q.; Shan, J.J.; Xu, Y.; Zhang, Q.X. Blocking postsynaptic density-93 binding to C-X3-C motif chemokine ligand 1 promotes microglial phenotypic transformation during acute ischemic stroke. Neural Regen. Res. 2023, 18, 1033–1039. [Google Scholar] [CrossRef]
  8. Pawelec, P.; Ziemka-Nalecz, M.; Sypecka, J.; Zalewska, T. The Impact of the CX3CL1/CX3CR1 Axis in Neurological Disorders. Cells 2020, 9, 2277. [Google Scholar] [CrossRef]
  9. Psychogios, M.; Brehm, A.; López-Cancio, E.; Marco De Marchis, G.; Meseguer, E.; Katsanos, A.H.; Kremer, C.; Sporns, P.; Zedde, M.; Kobayashi, A.; et al. European Stroke Organisation guidelines on treatment of patients with intracranial atherosclerotic disease. Eur. Stroke J. 2022, 7, XLII–LXXX. [Google Scholar] [CrossRef]
  10. Strbian, D.; Tsivgoulis, G.; Ospel, J.; Räty, S.; Cimflova, P.; Georgiopoulos, G.; Ullberg, T.; Arquizan, C.; Gralla, J.; Zeleňák, K.; et al. European Stroke Organisation and European Society for Minimally Invasive Neurological Therapy guideline on acute management of basilar artery occlusion. Eur. Stroke J. 2024, 9, 835–884. [Google Scholar] [CrossRef]
  11. Mead, G.E.; Sposato, L.A.; Sampaio Silva, G.; Yperzeele, L.; Wu, S.; Kutlubaev, M.; Cheyne, J.; Wahab, K.; Urrutia, V.C.; Sharma, V.K.; et al. A systematic review and synthesis of global stroke guidelines on behalf of the World Stroke Organization. Int. J. Stroke 2023, 18, 499–531. [Google Scholar] [CrossRef]
  12. Bernardo-Castro, S.; Sousa, J.A.; Martins, E.; Donato, H.; Nunes, C.; d’Almeida, O.C.; Castelo-Branco, M.; Abrunhosa, A.; Ferreira, L.; Sargento-Freitas, J. The evolution of blood-brain barrier permeability changes after stroke and its implications on clinical outcome: A systematic review and meta-analysis. Int. J. Stroke 2023, 18, 783–794. [Google Scholar] [CrossRef]
  13. Wen, X.; Shu, Z.; Li, Y.; Hu, X.; Gong, X. Developing a model for estimating infarction onset time based on computed tomography radiomics in patients with acute middle cerebral artery occlusion. BMC Med. Imaging 2021, 21, 147. [Google Scholar] [CrossRef] [PubMed]
  14. Xu, J.; Witchalls, J.; Preston, E.; Pan, L.; Zhang, G.; Waddington, G.; Adams, R.; Han, J. Proprioception After Unilateral Stroke: Changes in the Affected and Unaffected Lower Limbs over Time. Physiother. Res. Int. 2025, 30, e70027. [Google Scholar] [CrossRef] [PubMed]
  15. Zimmermann, S.; Weissenfels, M.; Krummer, N.; Michalski, D.; Weise, G.; Branzan, D.; Pelz, J.O. Long-Term Course of Circulating Elastin, Collagen Type I, and Collagen Type III in Patients with Spontaneous Cervical Artery Dissection: A Prospective Multicenter Study. Transl. Stroke Res. 2025, 16, 238–247. [Google Scholar] [CrossRef] [PubMed]
  16. Robba, C.; Aspide, R.; Pegoli, M.; Kondratyeva, E.; Gritti, P.; Fustini, M.F.; Battaglini, D.; Pelosi, P.; Hutchinson, P.J.; Helmy, A.; et al. Pituitary Dysfunction After Aneurysmal Subarachnoid Hemorrhage: A Prospective Cohort Study. J. Neurosurg. Anesthesiol. 2022, 34, 44–50. [Google Scholar] [CrossRef]
  17. Dai, S.; Lemaire, C.; Piscicelli, C.; Perennou, D. Lateropulsion Prevalence After Stroke: A Systematic Review and Meta-analysis. Neurology 2022, 98, e1574–e1584. [Google Scholar] [CrossRef]
  18. Zhou, C.; Yang, Y.; Lin, X.; Fang, N.; Chen, L.; Jiang, J.; Deng, H.; Deng, Y.; Wan, M.; Qiu, G.; et al. Proposed clinical phases for the improvement of personalized treatment of checkpoint inhibitor-related pneumonitis. Front. Immunol. 2022, 13, 935779. [Google Scholar] [CrossRef]
  19. Inoue, K. Potential significance of CX3CR1 dynamics in stress resilience against neuronal disorders. Neural Regen. Res. 2022, 17, 2153–2156. [Google Scholar] [CrossRef]
  20. Kim, S.; Lee, W.; Jo, H.; Sonn, S.K.; Jeong, S.J.; Seo, S.; Suh, J.; Jin, J.; Kweon, H.Y.; Kim, T.K.; et al. The antioxidant enzyme Peroxiredoxin-1 controls stroke-associated microglia against acute ischemic stroke. Redox Biol. 2022, 54, 102347. [Google Scholar] [CrossRef]
  21. Luczak-Sobotkowska, Z.M.; Rosa, P.; Lopez, M.B.; Ochocka, N.; Kiryk, A.; Lenkiewicz, A.M.; Furhmann, M.; Jankowski, A.; Kaminska, B. Tracking changes in functionality and morphology of repopulated microglia in young and old mice. J. Neuroinflamm. 2024, 21, 248. [Google Scholar] [CrossRef]
  22. Yang, L.; Geng, Y.; Qian, Y.; Zong, N.; Xia, S.; Yang, H.; Bao, X.; Chen, J.; Xu, Y. Loss of Plxdc2 exacerbates microglia-mediated neuroinflammation and ischemic brain injury. Exp. Neurol. 2025, 391, 115302. [Google Scholar] [CrossRef] [PubMed]
  23. Healy, L.M.; Zia, S.; Plemel, J.R. Towards a definition of microglia heterogeneity. Commun. Biol. 2022, 5, 1114. [Google Scholar] [CrossRef] [PubMed]
  24. Kubickova, L.; Dubovy, P. Dynamics of Cellular Regulation of Fractalkine/CX3CL1 and Its Receptor CX3CR1 in the Rat Trigeminal Subnucleus Caudalis after Unilateral Infraorbital Nerve Lesion-Extended Cellular Signaling of the CX3CL1/CX3CR1 Axis in the Development of Trigeminal Neuropathic Pain. Int. J. Mol. Sci. 2024, 25, 6069. [Google Scholar] [CrossRef]
  25. Brett, C.A.; Carroll, J.B.; Gabriele, M.L. Compromised fractalkine signaling delays microglial occupancy of emerging modules in the multisensory midbrain. Glia 2022, 70, 697–711. [Google Scholar] [CrossRef] [PubMed]
  26. Bolos, M.; Perea, J.R.; Terreros-Roncal, J.; Pallas-Bazarra, N.; Jurado-Arjona, J.; Avila, J.; Llorens-Martin, M. Absence of microglial CX3CR1 impairs the synaptic integration of adult-born hippocampal granule neurons. Brain Behav. Immun. 2018, 68, 76–89. [Google Scholar] [CrossRef] [PubMed]
  27. Zhang, T.; Zhang, M.; Cui, S.; Liang, W.; Jia, Z.; Guo, F.; Ou, W.; Wu, Y.; Zhang, S. The core of maintaining neuropathic pain: Crosstalk between glial cells and neurons (neural cell crosstalk at spinal cord). Brain Behav. 2023, 13, e2868. [Google Scholar] [CrossRef] [PubMed]
  28. Urban, J.; Suchankova, M.; Ganovska, M.; Leksa, V.; Sandor, F.; Tedlova, E.; Konig, B.; Bucova, M. The Role of CX3CL1 and ADAM17 in Pathogenesis of Diffuse Parenchymal Lung Diseases. Diagnostics 2021, 11, 1074. [Google Scholar] [CrossRef]
  29. Ye, J.; Shan, F.; Xu, X.; Liang, C.; Zhang, N.; Hu, H.; Li, J.; Ouyang, F.; Wang, J.; Zhao, Y.; et al. Centripetal migration and prolonged retention of microglia promotes spinal cord injury repair. J. Neuroinflamm. 2025, 22, 77. [Google Scholar] [CrossRef]
  30. Wang, X.; Xie, Y.; Niu, Y.; Wan, B.; Lu, Y.; Luo, Q.; Zhu, L. CX3CL1/CX3CR1 signal mediates M1-type microglia and accelerates high-altitude-induced forgetting. Front. Cell. Neurosci. 2023, 17, 1189348. [Google Scholar] [CrossRef]
  31. Murai, N.; Mitalipova, M.; Jaenisch, R. Functional analysis of CX3CR1 in human induced pluripotent stem (iPS) cell-derived microglia-like cells. Eur. J. Neurosci. 2020, 52, 3667–3678. [Google Scholar] [CrossRef]
  32. Olveda, G.E.; Barasa, M.N.; Hill, R.A. Microglial phagocytosis of single dying oligodendrocytes is mediated by CX3CR1 but not MERTK. Cell Rep. 2024, 43, 114385. [Google Scholar] [CrossRef]
  33. Lu, J.; Wang, C.; Cheng, X.; Wang, R.; Yan, X.; He, P.; Chen, H.; Yu, Z. A breakdown in microglial metabolic reprogramming causes internalization dysfunction of alpha-synuclein in a mouse model of Parkinson’s disease. J. Neuroinflamm. 2022, 19, 113. [Google Scholar] [CrossRef]
  34. Mukai, T.; Di Martino, E.; Tsuji, S.; Blomgren, K.; Nagamura-Inoue, T.; Aden, U. Umbilical cord-derived mesenchymal stromal cells immunomodulate and restore actin dynamics and phagocytosis of LPS-activated microglia via PI3K/Akt/Rho GTPase pathway. Cell Death Discov. 2021, 7, 46. [Google Scholar] [CrossRef] [PubMed]
  35. Lombardozzi, G.; Castelli, V.; Giorgi, C.; Cimini, A.; d’Angelo, M. Neuroinflammation strokes the brain: A double-edged sword in ischemic stroke. Neural Regen. Res. 2025, 24, 1456. [Google Scholar] [CrossRef] [PubMed]
  36. Subbarayan, M.S.; Joly-Amado, A.; Bickford, P.C.; Nash, K.R. CX3CL1/CX3CR1 signaling targets for the treatment of neurodegenerative diseases. Pharmacol. Ther. 2022, 231, 107989. [Google Scholar] [CrossRef] [PubMed]
  37. Lu, M.; Zhao, W.; Han, S.; Lin, X.; Xu, T.; Tan, Q.; Wang, M.; Yi, C.; Chu, X.; Yang, W.; et al. Activation of the human chemokine receptor CX3CR1 regulated by cholesterol. Sci. Adv. 2022, 8, eabn8048. [Google Scholar] [CrossRef]
  38. Zhan, L.; Qiu, M.; Zheng, J.; Lai, M.; Lin, K.; Dai, J.; Sun, W.; Xu, E. Fractalkine/CX3CR1 axis is critical for neuroprotection induced by hypoxic postconditioning against cerebral ischemic injury. Cell Commun. Signal. 2024, 22, 457. [Google Scholar] [CrossRef]
  39. von Beek, C.; Fahlgren, A.; Geiser, P.; Di Martino, M.L.; Lindahl, O.; Prensa, G.I.; Mendez-Enriquez, E.; Eriksson, J.; Hallgren, J.; Fallman, M.; et al. A two-step activation mechanism enables mast cells to differentiate their response between extracellular and invasive enterobacterial infection. Nat. Commun. 2024, 15, 904. [Google Scholar] [CrossRef]
  40. Qian, X.; Zheng, Y.; Xu, L.; Liu, Z.; Chen, M.; Tong, F.; Fan, P.; Chen, Z.; Dong, N.; Zhang, C.; et al. Deciphering the role of CX3CL1-CX3CR1 in aortic aneurysm pathogenesis: Insights from Mendelian randomization and transcriptomic analyses. Front. Immunol. 2024, 15, 1383607. [Google Scholar] [CrossRef]
  41. Park, S.H.; Lee, Y.S.; Yang, H.J.; Song, G.J. Fluoxetine Potentiates Phagocytosis and Autophagy in Microglia. Front. Pharmacol. 2021, 12, 770610. [Google Scholar] [CrossRef]
  42. Zhu, L.; Sun, S.; Wu, W.; Zhang, Y.; Lin, C.; Ji, L. Xanthotoxol alleviates secondary brain injury after intracerebral hemorrhage by inhibiting microglia-mediated neuroinflammation and oxidative stress. Neurochirurgie 2023, 69, 101426. [Google Scholar] [CrossRef]
  43. Babendreyer, A.; Molls, L.; Dreymueller, D.; Uhlig, S.; Ludwig, A. Shear Stress Counteracts Endothelial CX3CL1 Induction and Monocytic Cell Adhesion. Mediators Inflamm. 2017, 2017, 1515389. [Google Scholar] [CrossRef]
  44. Jia, M.; Li, Q.; Guo, J.; Shi, W.; Zhu, L.; Huang, Y.; Li, Y.; Wang, L.; Ma, S.; Zhuang, T.; et al. Deletion of BACH1 Attenuates Atherosclerosis by Reducing Endothelial Inflammation. Circ. Res. 2022, 130, 1038–1055. [Google Scholar] [CrossRef]
  45. Buscemi, L.; Price, M.; Bezzi, P.; Hirt, L. Spatio-temporal overview of neuroinflammation in an experimental mouse stroke model. Sci. Rep. 2019, 9, 507. [Google Scholar] [CrossRef] [PubMed]
  46. Lecordier, S.; Menet, R.; Allain, A.S.; ElAli, A. Non-classical monocytes promote neurovascular repair in cerebral small vessel disease associated with microinfarctions via CX3CR1. J. Cereb. Blood Flow Metab. 2023, 43, 1873–1890. [Google Scholar] [CrossRef] [PubMed]
  47. Brandi, E.; Torres-Garcia, L.; Svanbergsson, A.; Haikal, C.; Liu, D.; Li, W.; Li, J.Y. Brain region-specific microglial and astrocytic activation in response to systemic lipopolysaccharides exposure. Front. Aging Neurosci. 2022, 14, 910988. [Google Scholar] [CrossRef] [PubMed]
  48. Chamera, K.; Kotarska, K.; Szuster-Gluszczak, M.; Trojan, E.; Skorkowska, A.; Pomierny, B.; Krzyzanowska, W.; Bryniarska, N.; Basta-Kaim, A. The prenatal challenge with lipopolysaccharide and polyinosinic:polycytidylic acid disrupts CX3CL1-CX3CR1 and CD200-CD200R signalling in the brains of male rat offspring: A link to schizophrenia-like behaviours. J. Neuroinflamm. 2020, 17, 247. [Google Scholar] [CrossRef]
  49. Mou, K.J.; Shen, K.F.; Li, Y.L.; Wu, Z.F.; Duan, W. Adenosine A(2A) Receptor in Bone Marrow-Derived Cells Mediated Macrophages M2 Polarization via PPARgamma-P65 Pathway in Chronic Hypoperfusion Situation. Front. Aging Neurosci. 2021, 13, 792733. [Google Scholar] [CrossRef]
  50. Zhang, Z.; Chen, T.; Liu, W.; Xiong, J.; Jiang, L.; Liu, M. Paeonol accelerates skin wound healing by regulating macrophage polarization and inflammation in diabetic rats. Korean J. Physiol. Pharmacol. 2023, 27, 437–448. [Google Scholar] [CrossRef]
  51. Tutrow, K.; Harkin, J.; Hernandez, M.; Huang, K.-C.S.; Bissel, S.J.; Puntambekar, S.S.; Lamb, B.T.; Meyer, J.S. A human induced pluripotent stem cell model of Alzheimer’s Disease-associated fractalkine receptor polymorphism to assess AD-related microglial dysfunction. Alzheimer Dement. 2024, 20, e089429. [Google Scholar] [CrossRef]
  52. Park, J.; Kim, J.Y.; Kim, Y.R.; Huang, M.; Chang, J.Y.; Sim, A.Y.; Jung, H.; Lee, W.T.; Hyun, Y.M.; Lee, J.E. Reparative System Arising from CCR2(+) Monocyte Conversion Attenuates Neuroinflammation Following Ischemic Stroke. Transl. Stroke Res. 2021, 12, 879–893. [Google Scholar] [CrossRef]
  53. Zhang, C.; Li, K.; Zhu, H.; Cheng, M.; Chen, S.; Ling, R.; Wang, C.; Chen, D. ITGB6 modulates resistance to anti-CD276 therapy in head and neck cancer by promoting PF4(+) macrophage infiltration. Nat. Commun. 2024, 15, 7077. [Google Scholar] [CrossRef]
  54. Zhong, W.; Kokhaei, P.; Mulder, T.A.; Ghaderi, A.; Moshfegh, A.; Lundin, J.; Palma, M.; Schultz, J.; Olin, T.; Osterborg, A.; et al. A Small Molecule Antagonist of CX3CR1 (KAND567) Inhibited the Tumor Growth-Promoting Effect of Monocytes in Chronic Lymphocytic Leukemia (CLL). Cancers 2024, 16, 3821. [Google Scholar] [CrossRef] [PubMed]
  55. Church, K.A.; Rodriguez, D.; Vanegas, D.; Gutierrez, I.L.; Cardona, S.M.; Madrigal, J.L.M.; Kaur, T.; Cardona, A.E. Models of microglia depletion and replenishment elicit protective effects to alleviate vascular and neuronal damage in the diabetic murine retina. J. Neuroinflamm. 2022, 19, 300. [Google Scholar] [CrossRef]
  56. Bordeleau, M.; Lacabanne, C.; Fernandez de Cossio, L.; Vernoux, N.; Savage, J.C.; Gonzalez-Ibanez, F.; Tremblay, M.E. Microglial and peripheral immune priming is partially sexually dimorphic in adolescent mouse offspring exposed to maternal high-fat diet. J. Neuroinflamm. 2020, 17, 264. [Google Scholar] [CrossRef] [PubMed]
  57. Feng, L.; Lo, H.; Hong, Z.; Zheng, J.; Yan, Y.; Ye, Z.; Chen, X.; Pan, X. Microglial LRRK2-mediated NFATc1 attenuates alpha-synuclein immunotoxicity in association with CX3CR1-induced migration and the lysosome-initiated degradation. Glia 2023, 71, 2266–2284. [Google Scholar] [CrossRef]
  58. Popescu, A.S.; Butler, C.A.; Allendorf, D.H.; Piers, T.M.; Mallach, A.; Roewe, J.; Reinhardt, P.; Cinti, A.; Redaelli, L.; Boudesco, C.; et al. Alzheimer’s disease-associated R47H TREM2 increases, but wild-type TREM2 decreases, microglial phagocytosis of synaptosomes and neuronal loss. Glia 2023, 71, 974–990. [Google Scholar] [CrossRef] [PubMed]
  59. Gonzalez-Prieto, M.; Gutierrez, I.L.; Garcia-Bueno, B.; Caso, J.R.; Leza, J.C.; Ortega-Hernandez, A.; Gomez-Garre, D.; Madrigal, J.L.M. Microglial CX3CR1 production increases in Alzheimer’s disease and is regulated by noradrenaline. Glia 2021, 69, 73–90. [Google Scholar] [CrossRef]
  60. Greve, H.J.; Mumaw, C.L.; Messenger, E.J.; Kodavanti, P.R.S.; Royland, J.L.; Kodavanti, U.P.; Block, M.L. Diesel exhaust impairs TREM2 to dysregulate neuroinflammation. J. Neuroinflamm. 2020, 17, 351. [Google Scholar] [CrossRef]
  61. Delizannis, A.T.; Nonneman, A.; Tsering, W.; De Bondt, A.; Van den Wyngaert, I.; Zhang, B.; Meymand, E.; Olufemi, M.F.; Koivula, P.; Maimaiti, S.; et al. Effects of microglial depletion and TREM2 deficiency on Abeta plaque burden and neuritic plaque tau pathology in 5XFAD mice. Acta Neuropathol. Commun. 2021, 9, 150. [Google Scholar] [CrossRef] [PubMed]
  62. Zhang, Q.; Dai, J.; Lin, Y.; Li, M. Isobavachalcone alleviates ischemic stroke by suppressing HDAC1 expression and improving M2 polarization. Brain Res. Bull. 2024, 211, 110944. [Google Scholar] [CrossRef]
  63. Ge, Y.; Yang, J.; Chen, J.; Dai, M.; Dou, X.; Yao, S.; Yao, C.; Lin, Y. Absence in CX3CR1 receptor signaling promotes post-ischemic stroke cognitive function recovery through suppressed microglial pyroptosis in mice. CNS Neurosci. Ther. 2024, 30, e14551. [Google Scholar] [CrossRef]
  64. Bivona, G.; Iemmolo, M.; Ghersi, G. CX3CL1 Pathway as a Molecular Target for Treatment Strategies in Alzheimer’s Disease. Int. J. Mol. Sci. 2023, 24, 8230. [Google Scholar] [CrossRef]
  65. Fritze, J.; Muralidharan, C.; Stamp, E.; Ahlenius, H. Microglia undergo disease-associated transcriptional activation and CX3C motif chemokine receptor 1 expression regulates neurogenesis in the aged brain. Dev. Neurobiol. 2024, 84, 128–141. [Google Scholar] [CrossRef]
  66. Ge, Y.; Dou, X.; Chen, P.; Chen, J.; Dai, M.; Yao, S.; Lin, Y. Treadmill Exercise Enhances Post-Stroke Functional Recovery in Mice via the CX3CL1/CX3CR1 Signaling Pathway. Mol. Neurobiol. 2025, 62, 591–603. [Google Scholar] [CrossRef]
  67. Chen, W.; Su, G.; Chai, M.; An, Y.; Song, J.; Zhang, Z. Astrogliosis and glial scar in ischemic stroke—Focused on mechanism and treatment. Exp. Neurol. 2025, 385, 115131. [Google Scholar] [CrossRef]
  68. Ren, Z.L.; Zhang, C.X.; Zheng, Y.X.; Chen, C.A.; Dan, C.; Lan, X.; Yan, X.; Liu, Y.; He, Y.H.; Cheng, J.L.; et al. Refined qingkailing attenuates reactive astrocytes and glial scar formation after ischemia stroke via the EGFR/PLCgamma pathway. Phytomedicine 2025, 142, 156696. [Google Scholar] [CrossRef]
  69. Williamson, M.R.; Fuertes, C.J.A.; Dunn, A.K.; Drew, M.R.; Jones, T.A. Reactive astrocytes facilitate vascular repair and remodeling after stroke. Cell Rep. 2021, 35, 109048. [Google Scholar] [CrossRef]
  70. Kijima, C.; Inaba, T.; Hira, K.; Miyamoto, N.; Yamashiro, K.; Urabe, T.; Hattori, N.; Ueno, Y. Astrocytic Extracellular Vesicles Regulated by Microglial Inflammatory Responses Improve Stroke Recovery. Mol. Neurobiol. 2024, 61, 1002–1021. [Google Scholar] [CrossRef]
  71. Zhu, Y.; Li, L.; Jin, X.; Li, Z.; Wang, C.; Teng, L.; Li, Y.; Zhang, Y.; Wang, D. Structure characterisation of polysaccharides purified from Boletus aereus Bull. and its improvement on AD-like behaviours via reliving neuroinflammation in APP/PS1 mice. Int. J. Biol. Macromol. 2024, 258, 128819. [Google Scholar] [CrossRef] [PubMed]
  72. Hata, M.; Hata, M.; Dejda, A.; Pilon, F.; Diaz-Marin, R.; Fournier, F.; Joyal, J.S.; Cagnone, G.; Ochi, Y.; Crespo-Garcia, S.; et al. Corticosteroids reduce pathological angiogenesis yet compromise reparative vascular remodeling in a model of retinopathy. Proc. Natl. Acad. Sci. USA 2024, 121, e2411640121. [Google Scholar] [CrossRef] [PubMed]
  73. Choi, B.R.; Johnson, K.R.; Maric, D.; McGavern, D.B. Monocyte-derived IL-6 programs microglia to rebuild damaged brain vasculature. Nat. Immunol. 2023, 24, 1110–1123. [Google Scholar] [CrossRef] [PubMed]
  74. Raffaele, S.; Thougaard, E.; Laursen, C.C.H.; Gao, H.; Andersen, K.M.; Nielsen, P.V.; Orti-Casan, N.; Blichfeldt-Eckhardt, M.; Koch, S.; Deb-Chatterji, M.; et al. Microglial TNFR2 signaling regulates the inflammatory response after CNS injury in a sex-specific fashion. Brain Behav. Immun. 2024, 116, 269–285. [Google Scholar] [CrossRef]
  75. Zhang, X.; Li, H.; Gu, Y.; Ping, A.; Chen, J.; Zhang, Q.; Xu, Z.; Wang, J.; Tang, S.; Wang, R.; et al. Repair-associated macrophages increase after early-phase microglia attenuation to promote ischemic stroke recovery. Nat. Commun. 2025, 16, 3089. [Google Scholar] [CrossRef]
  76. Park, K.W.; Ju, H.; Kim, I.D.; Cave, J.W.; Guo, Y.; Wang, W.; Wu, Z.; Cho, S. Delayed Infiltration of Peripheral Monocyte Contributes to Phagocytosis and Transneuronal Degeneration in Chronic Stroke. Stroke 2022, 53, 2377–2388. [Google Scholar] [CrossRef]
  77. Gu, H.J.; Zuo, S.; Liu, H.Y.; Gu, L.L.; Yang, X.W.; Liao, J.; Wang, Q.Q.; Zhao, R.; Feng, X.S.; Li, H.Y. CX3CR1 participates in pulmonary angiogenesis in experimental hepatopulmonary syndrome mice through inhibiting AKT/ERK signaling pathway and regulating NO/NOS release. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 6645–6656. [Google Scholar] [CrossRef]
  78. Chen, P.; Zhao, W.; Guo, Y.; Xu, J.; Yin, M. CX3CL1/CX3CR1 in Alzheimer’s Disease: A Target for Neuroprotection. BioMed Res. Int. 2016, 2016, 8090918. [Google Scholar] [CrossRef]
  79. Madrigal, J.L.; Caso, J.R.; Garcia-Bueno, B.; Gutierrez, I.L.; Leza, J.C. Noradrenaline induces CX3CL1 production and release by neurons. Neuropharmacology 2017, 114, 146–155. [Google Scholar] [CrossRef]
  80. Collar, A.L.; Swamydas, M.; O’Hayre, M.; Sajib, M.S.; Hoffman, K.W.; Singh, S.P.; Mourad, A.; Johnson, M.D.; Ferre, E.M.; Farber, J.M.; et al. The homozygous CX3CR1-M280 mutation impairs human monocyte survival. JCI Insight 2018, 3, e95417. [Google Scholar] [CrossRef]
  81. Fan, Q.; He, W.; Gayen, M.; Benoit, M.R.; Luo, X.; Hu, X.; Yan, R. Activated CX3CL1/Smad2 Signals Prevent Neuronal Loss and Alzheimer’s Tau Pathology-Mediated Cognitive Dysfunction. J. Neurosci. 2020, 40, 1133–1144. [Google Scholar] [CrossRef]
  82. Watroba, M.; Grabowska, A.D.; Szukiewicz, D. Chemokine CX3CL1 (Fractalkine) Signaling and Diabetic Encephalopathy. Int. J. Mol. Sci. 2024, 25, 7527. [Google Scholar] [CrossRef]
  83. He, H.Y.; Ren, L.; Guo, T.; Deng, Y.H. Neuronal autophagy aggravates microglial inflammatory injury by downregulating CX3CL1/fractalkine after ischemic stroke. Neural Regen. Res. 2019, 14, 280–288. [Google Scholar] [CrossRef]
  84. O’Sullivan, S.A.; Gasparini, F.; Mir, A.K.; Dev, K.K. Fractalkine shedding is mediated by p38 and the ADAM10 protease under pro-inflammatory conditions in human astrocytes. J. Neuroinflamm. 2016, 13, 189. [Google Scholar] [CrossRef]
  85. Zhang, J.; Liu, Y.; Liu, X.; Li, S.; Cheng, C.; Chen, S.; Le, W. Dynamic changes of CX3CL1/CX3CR1 axis during microglial activation and motor neuron loss in the spinal cord of ALS mouse model. Transl. Neurodegener. 2018, 7, 35. [Google Scholar] [CrossRef] [PubMed]
  86. van der Maten, G.; Henck, V.; Wieloch, T.; Ruscher, K. CX(3)C chemokine receptor 1 deficiency modulates microglia morphology but does not affect lesion size and short-term deficits after experimental stroke. BMC Neurosci. 2017, 18, 11. [Google Scholar] [CrossRef] [PubMed]
  87. Ikezu, T. Microglia and extracellular vesicle-mediated spread of Tau pathology. Alzheimer’s Dement. 2024, 20, e087779. [Google Scholar] [CrossRef]
  88. Wang, L.; Ling, H.; He, H.; Hu, N.; Xiao, L.; Zhang, Y.; Xie, L.; You, Z. Dysfunctional synaptic pruning by microglia correlates with cognitive impairment in sleep-deprived mice: Involvement of CX3CR1 signaling. Neurobiol. Stress 2023, 25, 100553. [Google Scholar] [CrossRef]
  89. Cornell, J.; Salinas, S.; Huang, H.Y.; Zhou, M. Microglia regulation of synaptic plasticity and learning and memory. Neural Regen. Res. 2022, 17, 705–716. [Google Scholar] [CrossRef]
  90. Liu, H.; Jiang, M.; Chen, Z.; Li, C.; Yin, X.; Zhang, X.; Wu, M. The Role of the Complement System in Synaptic Pruning after Stroke. Aging Dis. 2024, 16, 1452–1470. [Google Scholar] [CrossRef]
  91. Wang, J.; Gan, Y.; Han, P.; Yin, J.; Liu, Q.; Ghanian, S.; Gao, F.; Gong, G.; Tang, Z. Ischemia-induced Neuronal Cell Death Is Mediated by Chemokine Receptor CX3CR1. Sci. Rep. 2018, 8, 556. [Google Scholar] [CrossRef]
  92. Faustino, J.; Chip, S.; Derugin, N.; Jullienne, A.; Hamer, M.; Haddad, E.; Butovsky, O.; Obenaus, A.; Vexler, Z.S. CX3CR1-CCR2-dependent monocyte-microglial signaling modulates neurovascular leakage and acute injury in a mouse model of childhood stroke. J. Cereb. Blood Flow Metab. 2019, 39, 1919–1935. [Google Scholar] [CrossRef]
  93. Puntambekar, S.S.; Moutinho, M.; Lin, P.B.; Jadhav, V.; Tumbleson-Brink, D.; Balaji, A.; Benito, M.A.; Xu, G.; Oblak, A.; Lasagna-Reeves, C.A.; et al. CX3CR1 deficiency aggravates amyloid driven neuronal pathology and cognitive decline in Alzheimer’s disease. Mol. Neurodegener. 2022, 17, 47. [Google Scholar] [CrossRef]
  94. Hickman, S.E.; Allison, E.K.; Coleman, U.; Kingery-Gallagher, N.D.; El Khoury, J. Heterozygous CX3CR1 Deficiency in Microglia Restores Neuronal beta-Amyloid Clearance Pathways and Slows Progression of Alzheimer’s Like-Disease in PS1-APP Mice. Front. Immunol. 2019, 10, 2780. [Google Scholar] [CrossRef]
  95. Sakai, M.; Takeuchi, H.; Yu, Z.; Kikuchi, Y.; Ono, C.; Takahashi, Y.; Ito, F.; Matsuoka, H.; Tanabe, O.; Yasuda, J.; et al. Polymorphisms in the microglial marker molecule CX3CR1 affect the blood volume of the human brain. Psychiatry Clin. Neurosci. 2018, 72, 409–422. [Google Scholar] [CrossRef] [PubMed]
  96. Lopez-Lopez, A.; Gelpi, E.; Lopategui, D.M.; Vidal-Taboada, J.M. Association of the CX3CR1-V249I Variant with Neurofibrillary Pathology Progression in Late-Onset Alzheimer’s Disease. Mol. Neurobiol. 2018, 55, 2340–2349. [Google Scholar] [CrossRef] [PubMed]
  97. Du, B.; Liang, M.; Zheng, H.; Fan, C.; Zhang, H.; Lu, X.; Du, Z.; Lian, Y.; Zhang, Y.; Bi, X. Anti-mouse CX3CR1 Antibody Alleviates Cognitive Impairment, Neuronal Loss and Myelin Deficits in an Animal Model of Brain Ischemia. Neuroscience 2020, 438, 169–181. [Google Scholar] [CrossRef] [PubMed]
  98. Lv, J.; Shen, X.; Shen, X.; Zhao, S.; Xu, R.; Yan, Q.; Lu, J.; Zhu, D.; Zhao, Y.; Dong, J.; et al. NPLC0393 from Gynostemma pentaphyllum ameliorates Alzheimer’s disease-like pathology in mice by targeting protein phosphatase magnesium-dependent 1A phosphatase. Phytother. Res. 2023, 37, 4771–4790. [Google Scholar] [CrossRef]
  99. Jiang, L.; Zhang, X.; Wang, S.; Zhang, J.; Chen, J.; Lu, J.; Yao, L.; Jin, W.; Li, N.; Li, Q. Functional Monomers Equipped Microgel System for Managing Parkinson’s Disease by Intervening Chemokine Axis-mediated Nerve Cell Communications. Adv. Sci. 2025, 12, e2410070. [Google Scholar] [CrossRef]
  100. Huang, M.; Wan, Y.; Mao, L.; He, Q.W.; Xia, Y.P.; Li, M.; Li, Y.N.; Jin, H.J.; Hu, B. Inhibiting the Migration of M1 Microglia at Hyperacute Period Could Improve Outcome of tMCAO Rats. CNS Neurosci. Ther. 2017, 23, 222–232. [Google Scholar] [CrossRef]
  101. Ye, S.Y.; Apple, J.E.; Ren, X.; Tang, F.L.; Yao, L.L.; Wang, Y.G.; Mei, L.; Zhou, Y.G.; Xiong, W.C. Microglial VPS35 deficiency regulates microglial polarization and decreases ischemic stroke-induced damage in the cortex. J. Neuroinflamm. 2019, 16, 235. [Google Scholar] [CrossRef]
  102. Rayasam, A.; Faustino, J.; Lecuyer, M.; Vexler, Z.S. Neonatal Stroke and TLR1/2 Ligand Recruit Myeloid Cells through the Choroid Plexus in a CX3CR1-CCR2- and Context-Specific Manner. J. Neurosci. 2020, 40, 3849–3861. [Google Scholar] [CrossRef]
  103. Guo, X.; Liu, R.; Jia, M.; Wang, Q.; Wu, J. Ischemia Reperfusion Injury Induced Blood Brain Barrier Dysfunction and the Involved Molecular Mechanism. Neurochem. Res. 2023, 48, 2320–2334. [Google Scholar] [CrossRef] [PubMed]
  104. Zhou, Y.F.; Li, Y.N.; Jin, H.J.; Wu, J.H.; He, Q.W.; Wang, X.X.; Lei, H.; Hu, B. Sema4D/PlexinB1 inhibition ameliorates blood-brain barrier damage and improves outcome after stroke in rats. FASEB J. 2018, 32, 2181–2196. [Google Scholar] [CrossRef] [PubMed]
  105. Zhao, J.; Mu, H.; Liu, L.; Jiang, X.; Wu, D.; Shi, Y.; Leak, R.K.; Ji, X. Transient selective brain cooling confers neurovascular and functional protection from acute to chronic stages of ischemia/reperfusion brain injury. J. Cereb. Blood Flow Metab. 2019, 39, 1215–1231. [Google Scholar] [CrossRef] [PubMed]
  106. Liu, Y.; Yao, F.; Li, Z.; Jiang, Y.; Li, J.; Yu, S.; Hu, X.; Ouyang, F.; Zheng, M.; Cheng, L.; et al. Dynamic phosphorylation of Fascin-1 orchestrates microglial phagocytosis and neurological recovery after spinal cord injury. J. Neuroinflamm. 2025, 22, 121. [Google Scholar] [CrossRef]
  107. Brown, G.C. Neuronal Loss after Stroke Due to Microglial Phagocytosis of Stressed Neurons. Int. J. Mol. Sci. 2021, 22, 13442. [Google Scholar] [CrossRef]
  108. Doust, Y.V.; Bindoff, A.; Holloway, O.G.; Wilson, R.; King, A.E.; Ziebell, J.M. Temporal changes in the microglial proteome of male and female mice after a diffuse brain injury using label-free quantitative proteomics. Glia 2023, 71, 880–903. [Google Scholar] [CrossRef]
  109. Zhao, A.; Ding, Y.; Zhong, M.; Niu, M.; Wang, L.; Jiao, Y.; Liu, J.; Li, Y. Neuronal DJ-1 regulates microglial activation in Parkinson’s disease. Neural Regen. Res. 2025, 24, 1047. [Google Scholar] [CrossRef]
  110. Chen, X.; He, X.; Xu, F.; Xu, N.; Sharifi, N.H.; Zhang, P.; Flores, J.J.; Wu, L.; He, Q.; Kanamaru, H.; et al. Fractalkine Enhances Hematoma Resolution and Improves Neurological Function via CX3CR1/AMPK/PPARgamma Pathway after GMH. Stroke 2023, 54, 2420–2433. [Google Scholar] [CrossRef]
  111. Yang, H.; Yang, J.; Park, N.; Hwang, D.S.; Park, S.Y.; Kim, S.; Bae, H. Adoptive Transfer of CX3CR1-Transduced Tregs Homing to the Forebrain in Lipopolysaccharide-Induced Neuroinflammation and 3xTg Alzheimer’s Disease Models. Int. J. Mol. Sci. 2024, 25, 13682. [Google Scholar] [CrossRef]
  112. Camacho-Hernández, N.P.; Peña-Ortega, F. Fractalkine/CX3CR1-Dependent Modulation of Synaptic and Network Plasticity in Health and Disease. Neural Plast. 2023, 2023, 4637073. [Google Scholar] [CrossRef]
  113. Li, Z.; Qiang, Y.; Chen, D.; Hu, D.; Gao, D.; Xu, X.; Sun, L.; Li, Y.; Qiu, W.; Sheng, Z. Dual-modal super-resolution ultrasound and NIR-II fluorescence imaging of ischemic stroke with ICG-doped porous PLGA microspheres. Mater. Today Bio 2025, 31, 101513. [Google Scholar] [CrossRef] [PubMed]
  114. Alkhiri, A.; Alturki, F.; Alansari, N.M.; Almaghrabi, A.A.; Alghamdi, B.A.; Alamri, A.F.; Alghamdi, S.; Makkawi, S. Prognosis and distribution of ischemic stroke with negative diffusion-weighted imaging: A systematic review and meta-analysis. Front. Neurol. 2024, 15, 1376439. [Google Scholar] [CrossRef] [PubMed]
  115. Chavva, I.R.; Crawford, A.L.; Mazurek, M.H.; Yuen, M.M.; Prabhat, A.M.; Payabvash, S.; Sze, G.; Falcone, G.J.; Matouk, C.C.; de Havenon, A.; et al. Deep Learning Applications for Acute Stroke Management. Ann. Neurol. 2022, 92, 574–587. [Google Scholar] [CrossRef]
  116. Protzmann, J.; Jung, F.; Jakobsson, L.; Fredriksson, L. Analysis of ischemic stroke-mediated effects on blood-brain barrier properties along the arteriovenous axis assessed by intravital two-photon imaging. Fluids Barriers CNS 2024, 21, 35. [Google Scholar] [CrossRef]
  117. Zhao, Y.H.; Liang, Y.; Wang, K.J.; Jin, S.N.; Yu, X.M.; Zhang, Q.; Wei, J.Y.; Liu, H.; Fang, W.G.; Zhao, W.D.; et al. Endothelial lincRNA-p21 alleviates cerebral ischemia/reperfusion injury by maintaining blood-brain barrier integrity. J. Cereb. Blood Flow Metab. 2024, 44, 1532–1550. [Google Scholar] [CrossRef]
  118. Du, L.D.; Fang, C.; Wang, Y.Q.; Feng, Z.Y.; Abiola, O.F.; Gao, Z.L.; Huang, J.Y.; Ma, Y.Z. MMP-9 inhibitor SB-3CT improves neurological outcomes in ischemic stroke mice by modulation of astrocytic lipid metabolism. Acta Pharmacol. Sin. 2025, 25, 1505. [Google Scholar] [CrossRef]
  119. Ji, Y.; Gao, Q.; Ma, Y.; Wang, F.; Tan, X.; Song, D.; Hoo, R.L.C.; Wang, Z.; Ge, X.; Han, H.; et al. An MMP-9 exclusive neutralizing antibody attenuates blood-brain barrier breakdown in mice with stroke and reduces stroke patient-derived MMP-9 activity. Pharmacol. Res. 2023, 190, 106720. [Google Scholar] [CrossRef]
  120. Foley, K.E.; Weekman, E.M.; Wilcock, D.M. Understanding the cellular responses to anti-Abeta antibodies to gain insights into mechanisms of ARIA. Alzheimer’s Dement. 2024, 20, e085573. [Google Scholar] [CrossRef]
  121. Guo, P.; Li, H.; Zhang, X.; Liu, Y.; Xue, S.; Yong, V.W.; Xue, M. Matrix metalloproteinase-9 in hemorrhagic transformation after acute ischemic stroke (Review). Mol. Med. Rep. 2025, 32, 13590. [Google Scholar] [CrossRef]
  122. Schauer, S.P.; Cho, C.H.; Novikova, G.; Roth, G.A.; Lee, J.; Sharma, A.D.; Foley, A.R.; Ng, C.; Shen, P.; Choi, M.; et al. Primate cerebrospinal fluid CHI3L1 reflects brain TREM2 agonism. Alzheimer’s Dement. 2024, 20, 5861–5888. [Google Scholar] [CrossRef]
  123. Yi, L.; Liang, Y.; Zhao, Q.; Wang, H.; Dong, J. CX3CL1 Induces Vertebral Microvascular Barrier Dysfunction via the Src/P115-RhoGEF/ROCK Signaling Pathway. Front. Cell. Neurosci. 2020, 14, 96. [Google Scholar] [CrossRef] [PubMed]
  124. Shi, J.; Wang, X.; Kang, C.; Liu, J.; Ma, C.; Yang, L.; Hu, J.; Zhao, N. TREM2 regulates BV2 microglia activation and influences corticosterone-induced neuroinflammation in depressive disorders. Brain Res. 2024, 1822, 148664. [Google Scholar] [CrossRef] [PubMed]
  125. Xie, M.; Liu, Y.U.; Zhao, S.; Zhang, L.; Bosco, D.B.; Pang, Y.P.; Zhong, J.; Sheth, U.; Martens, Y.A.; Zhao, N.; et al. TREM2 interacts with TDP-43 and mediates microglial neuroprotection against TDP-43-related neurodegeneration. Nat. Neurosci. 2022, 25, 26–38. [Google Scholar] [CrossRef] [PubMed]
  126. Ulland, T.K.; Song, W.M.; Huang, S.C.; Ulrich, J.D.; Sergushichev, A.; Beatty, W.L.; Loboda, A.A.; Zhou, Y.; Cairns, N.J.; Kambal, A.; et al. TREM2 Maintains Microglial Metabolic Fitness in Alzheimer’s Disease. Cell 2017, 170, 649–663.e13. [Google Scholar] [CrossRef]
  127. Hwang, M.; Savarin, C.; Kim, J.; Powers, J.; Towne, N.; Oh, H.; Bergmann, C.C. Trem2 deficiency impairs recovery and phagocytosis and dysregulates myeloid gene expression during virus-induced demyelination. J. Neuroinflamm. 2022, 19, 267. [Google Scholar] [CrossRef]
  128. Zhou, W.; He, J.; Shen, G.; Liu, Y.; Zhao, P.; Li, J. TREM2-dependent activation of microglial cell protects photoreceptor cell during retinal degeneration via PPARgamma and CD36. Cell Death Dis. 2024, 15, 623. [Google Scholar] [CrossRef]
  129. Kurisu, K.; Zheng, Z.; Kim, J.Y.; Shi, J.; Kanoke, A.; Liu, J.; Hsieh, C.L.; Yenari, M.A. Triggering receptor expressed on myeloid cells-2 expression in the brain is required for maximal phagocytic activity and improved neurological outcomes following experimental stroke. J. Cereb. Blood Flow Metab. 2019, 39, 1906–1918. [Google Scholar] [CrossRef]
  130. Zhang, X.; Tang, L.; Yang, J.; Meng, L.; Chen, J.; Zhou, L.; Wang, J.; Xiong, M.; Zhang, Z. Soluble TREM2 ameliorates tau phosphorylation and cognitive deficits through activating transgelin-2 in Alzheimer’s disease. Nat. Commun. 2023, 14, 6670. [Google Scholar] [CrossRef]
  131. D’Anna, L.; Searle, G.; Harvey, K.; Matthews, P.M.; Veltkamp, R. Time course of neuroinflammation after human stroke—A pilot study using co-registered PET and MRI. BMC Neurol. 2023, 23, 193. [Google Scholar] [CrossRef]
  132. Villa, A.; Klein, B.; Janssen, B.; Pedragosa, J.; Pepe, G.; Zinnhardt, B.; Vugts, D.J.; Gelosa, P.; Sironi, L.; Beaino, W.; et al. Identification of new molecular targets for PET imaging of the microglial anti-inflammatory activation state. Theranostics 2018, 8, 5400–5418. [Google Scholar] [CrossRef] [PubMed]
  133. Cadiz, M.P.; Jensen, T.D.; Sens, J.P.; Zhu, K.; Song, W.M.; Zhang, B.; Ebbert, M.; Chang, R.; Fryer, J.D. Culture shock: Microglial heterogeneity, activation, and disrupted single-cell microglial networks in vitro. Mol. Neurodegener. 2022, 17, 26. [Google Scholar] [CrossRef] [PubMed]
  134. Wan, Y.; Feng, B.; You, Y.; Yu, J.; Xu, C.; Dai, H.; Trapp, B.D.; Shi, P.; Chen, Z.; Hu, W. Microglial Displacement of GABAergic Synapses Is a Protective Event during Complex Febrile Seizures. Cell Rep. 2020, 33, 108346. [Google Scholar] [CrossRef] [PubMed]
  135. Zhou, K.; Han, J.; Lund, H.; Boggavarapu, N.R.; Lauschke, V.M.; Goto, S.; Cheng, H.; Wang, Y.; Tachi, A.; Xie, C.; et al. An overlooked subset of Cx3cr1(wt/wt) microglia in the Cx3cr1(CreER-Eyfp/wt) mouse has a repopulation advantage over Cx3cr1(CreER-Eyfp/wt) microglia following microglial depletion. J. Neuroinflamm. 2022, 19, 20. [Google Scholar] [CrossRef]
  136. Zhang, Y.; Liu, K.; Sun, Q.; Qi, Y.; Li, F.; Su, X.; Song, M.; Lv, R.; Sui, H.; Shi, Y.; et al. Collagenase Degradable Biomimetic Nanocages Attenuate Porphyromonas gingivalis Mediated Neurocognitive Dysfunction via Targeted Intracerebral Antimicrobial Photothermal and Gas Therapy. ACS Nano 2025, 19, 16448–16468. [Google Scholar] [CrossRef]
  137. Li, N.; Wang, H.; Hu, C.; Qie, S.; Liu, Z. Regulatory T Cells for Stroke Recovery: A Promising Immune Therapeutic Strategy. CNS Neurosci. Ther. 2025, 31, e70248. [Google Scholar] [CrossRef]
  138. Wicks, E.E.; Ran, K.R.; Kim, J.E.; Xu, R.; Lee, R.P.; Jackson, C.M. The Translational Potential of Microglia and Monocyte-Derived Macrophages in Ischemic Stroke. Front. Immunol. 2022, 13, 897022. [Google Scholar] [CrossRef]
  139. Tauil, C.B.; Rocha-Lima, A.D.; Ferrari, B.B.; Silva, F.M.D.; Machado, L.A.; Ramari, C.; Brandao, C.O.; Santos, L.; Santos-Neto, L.L.D. Depression and anxiety disorders in patients with multiple sclerosis: Association with neurodegeneration and neurofilaments. Braz. J. Med. Biol. Res. 2021, 54, e10428. [Google Scholar] [CrossRef]
  140. Rauchmann, B.S.; Sadlon, A.; Perneczky, R. Soluble TREM2 and Inflammatory Proteins in Alzheimer’s Disease Cerebrospinal Fluid. J. Alzheimer’s Dis. 2020, 73, 1615–1626. [Google Scholar] [CrossRef]
  141. Li, Y.; Wang, K.; Shi, G.; Qi, A.; Li, Y.; Jiao, Y.; Hu, R.; Hu, G.; Li, Y.; Shen, Z.; et al. Metal exposure is associated with cognitive function and plasma sTrem2 levels in coal miners: A cross-sectional study. Environ. Res. 2025, 279, 121726. [Google Scholar] [CrossRef]
  142. Gisslen, M.; Heslegrave, A.; Veleva, E.; Yilmaz, A.; Andersson, L.M.; Hagberg, L.; Spudich, S.; Fuchs, D.; Price, R.W.; Zetterberg, H. CSF concentrations of soluble TREM2 as a marker of microglial activation in HIV-1 infection. Neurol. Neuroimmunol. Neuroinflamm. 2019, 6, e512. [Google Scholar] [CrossRef] [PubMed]
  143. Jay, T.R.; von Saucken, V.E.; Munoz, B.; Codocedo, J.F.; Atwood, B.K.; Lamb, B.T.; Landreth, G.E. TREM2 is required for microglial instruction of astrocytic synaptic engulfment in neurodevelopment. Glia 2019, 67, 1873–1892. [Google Scholar] [CrossRef] [PubMed]
  144. Sun, H.; Wang, H.; Wu, C.; Liu, G.; He, M.; Zhang, H.; Hou, F.; Liao, H. Enhancing Neuron Activity Promotes Functional Recovery by Inhibiting Microglia-Mediated Synapse Elimination After Stroke. Stroke 2025, 56, 505–516. [Google Scholar] [CrossRef] [PubMed]
  145. Teter, O.M.; McQuade, A.; Hagan, V.; Liang, W.; Drager, N.M.; Sattler, S.M.; Holmes, B.B.; Castillo, V.C.; Papakis, V.; Leng, K.; et al. CRISPRi-based screen of autism spectrum disorder risk genes in microglia uncovers roles of ADNP in microglia endocytosis and synaptic pruning. Mol. Psychiatry 2025, 25, 2997. [Google Scholar] [CrossRef]
  146. Zhang, M.M.; Guo, M.X.; Zhang, Q.P.; Chen, X.Q.; Li, N.Z.; Liu, Q.; Cheng, J.; Wang, S.L.; Xu, G.H.; Li, C.F.; et al. IL-1R/C3aR signaling regulates synaptic pruning in the prefrontal cortex of depression. Cell Biosci. 2022, 12, 90. [Google Scholar] [CrossRef]
  147. Pichla, M.; Bartosz, G.; Stefaniuk, I.; Sadowska-Bartosz, I. pH-Responsive Redox Nanoparticles Protect SH-SY5Y Cells at Lowered pH in a Cellular Model of Parkinson’s Disease. Molecules 2021, 26, 543. [Google Scholar] [CrossRef]
  148. Cheng, Y.; Cheng, A.; Jia, Y.; Yang, L.; Ning, Y.; Xu, L.; Zhong, Y.; Zhuang, Z.; Guan, J.; Zhang, X.; et al. pH-Responsive Multifunctional Theranostic Rapamycin-Loaded Nanoparticles for Imaging and Treatment of Acute Ischemic Stroke. ACS Appl. Mater. Interfaces 2021, 13, 56909–56922. [Google Scholar] [CrossRef]
  149. Li, T.; Ashrafizadeh, M.; Shang, Y.; Nuri Ertas, Y.; Orive, G. Chitosan-functionalized bioplatforms and hydrogels in breast cancer: Immunotherapy, phototherapy and clinical perspectives. Drug Discov. Today 2024, 29, 103851. [Google Scholar] [CrossRef]
  150. Xu, M.; Qin, H.; Zheng, Y.; Chen, J.; Liang, X.; Huang, J.; Luo, W.; Yang, R.; Guan, Y.Q. Construction of a double-responsive modified guar gum nanoparticles and its application in oral insulin administration. Colloids Surf. B Biointerfaces 2022, 220, 112858. [Google Scholar] [CrossRef]
  151. Sun, S.; Lv, W.; Li, S.; Zhang, Q.; He, W.; Min, Z.; Teng, C.; Chen, Y.; Liu, L.; Yin, J.; et al. Smart Liposomal Nanocarrier Enhanced the Treatment of Ischemic Stroke through Neutrophil Extracellular Traps and Cyclic Guanosine Monophosphate-Adenosine Monophosphate Synthase-Stimulator of Interferon Genes (cGAS-STING) Pathway Inhibition of Ischemic Penumbra. ACS Nano 2023, 17, 17845–17857. [Google Scholar] [CrossRef] [PubMed]
  152. Pourradi, N.M.A.; Babaei, H.; Hamishehkar, H.; Baradaran, B.; Shokouhi-Gogani, B.; Shanehbandi, D.; Ghorbani, M.; Azarmi, Y. Targeted delivery of doxorubicin by Thermo/pH-responsive magnetic nanoparticles in a rat model of breast cancer. Toxicol. Appl. Pharmacol. 2022, 446, 116036. [Google Scholar] [CrossRef] [PubMed]
  153. Jin, L.; Zhu, Z.; Hong, L.; Qian, Z.; Wang, F.; Mao, Z. ROS-responsive 18beta-glycyrrhetic acid-conjugated polymeric nanoparticles mediate neuroprotection in ischemic stroke through HMGB1 inhibition and microglia polarization regulation. Bioact. Mater. 2023, 19, 38–49. [Google Scholar] [CrossRef] [PubMed]
  154. Wang, J.; Lin, J.; Chen, Y.; Liu, J.; Zheng, Q.; Deng, M.; Wang, R.; Zhang, Y.; Feng, S.; Xu, Z.; et al. An ultra-compact promoter drives widespread neuronal expression in mouse and monkey brains. Cell Rep. 2023, 42, 113348. [Google Scholar] [CrossRef]
  155. Zhang, B.; Zou, J.; Han, L.; Beeler, B.; Friedman, J.L.; Griffin, E.; Piao, Y.S.; Rensing, N.R.; Wong, M. The specificity and role of microglia in epileptogenesis in mouse models of tuberous sclerosis complex. Epilepsia 2018, 59, 1796–1806. [Google Scholar] [CrossRef]
  156. Cisbani, G.; Le Behot, A.; Plante, M.M.; Prefontaine, P.; Lecordier, M.; Rivest, S. Role of the chemokine receptors CCR2 and CX3CR1 in an experimental model of thrombotic stroke. Brain Behav. Immun. 2018, 70, 280–292. [Google Scholar] [CrossRef]
  157. Chaudhri, A.; Bu, X.; Wang, Y.; Gomez, M.; Torchia, J.A.; Hua, P.; Hung, S.H.; Davies, M.A.; Lizee, G.A.; von Andrian, U.; et al. The CX3CL1-CX3CR1 chemokine axis can contribute to tumor immune evasion and blockade with a novel CX3CR1 monoclonal antibody enhances response to anti-PD-1 immunotherapy. Front. Immunol. 2023, 14, 1237715. [Google Scholar] [CrossRef]
  158. Chen, H.; Ye, Z.; Korpershoek, J.V.; Creemers, L.B.; Weinans, H.; Rios, J.L. Antibody and aptamer-based therapies for osteoarthritis: Application of antibodies and promise of aptamers. Mol. Ther. Nucleic Acids 2025, 36, 102552. [Google Scholar] [CrossRef]
  159. Shoemaker, R.L.; Larsen, R.J.; Larsen, P.A. Single-domain antibodies and aptamers drive new opportunities for neurodegenerative disease research. Front. Immunol. 2024, 15, 1426656. [Google Scholar] [CrossRef]
  160. Doherty, C.; Wilbanks, B.; Khatua, S.; Maher, L.J., 3rd. Aptamers in neuro-oncology: An emerging therapeutic modality. Neuro-Oncology 2024, 26, 38–54. [Google Scholar] [CrossRef]
  161. Sharma, R.; Lee, K. Advances in treatments for acute ischemic stroke. BMJ 2025, 389, e076161. [Google Scholar] [CrossRef]
  162. Kim, H.J.; Roh, H.G. Imaging in Acute Anterior Circulation Ischemic Stroke: Current and Future. Neurointervention 2022, 17, 2–17. [Google Scholar] [CrossRef]
  163. Broocks, G.; Rajput, F.; Hanning, U.; Faizy, T.D.; Leischner, H.; Schon, G.; Gellissen, S.; Sporns, P.; Deb-Chatterji, M.; Thomalla, G.; et al. Highest Lesion Growth Rates in Patients with Hyperacute Stroke: When Time Is Brain Particularly Matters. Stroke 2019, 50, 189–192. [Google Scholar] [CrossRef]
  164. Lambertsen, K.L.; Finsen, B.; Clausen, B.H. Post-stroke inflammation-target or tool for therapy? Acta Neuropathol. 2019, 137, 693–714. [Google Scholar] [CrossRef] [PubMed]
  165. Malone, K.; Amu, S.; Moore, A.C.; Waeber, C. Immunomodulatory Therapeutic Strategies in Stroke. Front. Pharmacol. 2019, 10, 630. [Google Scholar] [CrossRef] [PubMed]
  166. Veltkamp, R.; Gill, D. Clinical Trials of Immunomodulation in Ischemic Stroke. Neurotherapeutics 2016, 13, 791–800. [Google Scholar] [CrossRef] [PubMed]
  167. Zhou, H.; Li, J.; Hu, L.; Yu, J.; Fu, X.; Liang, F.; Yan, F.; Chen, G. Soluble Trem2 is a negative regulator of erythrophagocytosis after intracerebral hemorrhage in a CD36 receptor recycling manner. J. Adv. Res. 2023, 44, 185–199. [Google Scholar] [CrossRef]
  168. Huang, X.; Guo, M.; Zhang, Y.; Xie, J.; Huang, R.; Zuo, Z.; Saw, P.E.; Cao, M. Microglial IL-1RA ameliorates brain injury after ischemic stroke by inhibiting astrocytic CXCL1-mediated neutrophil recruitment and microvessel occlusion. Glia 2023, 71, 1607–1625. [Google Scholar] [CrossRef]
  169. Yang, L.; Zhang, S.; Luo, Q.; Shu, H.; Xu, W.; Zhu, X.; Hu, P.; Wu, Y.; Shu, L.; Liu, J.; et al. A pH-responsive guanidino-based covalent organic framework nanodrugs for enhanced neuroprotection against subarachnoid hemorrhage by targeting NLRP3 inflammasome. Biomaterials 2025, 324, 123467. [Google Scholar] [CrossRef]
  170. Zhang, L.; Li, W.; Xu, Z.; Mao, Z.; Yang, M.; Wang, C.; Liu, Z. Promoting transcellular traversal of the blood-brain barrier by simultaneously improving cellular uptake and accelerating lysosomal escape. Nanoscale 2025, 17, 6780–6792. [Google Scholar] [CrossRef] [PubMed]
  171. Chen, Z.; Wang, X.; Zhao, N.; Chen, H.; Guo, G. Advancements in pH-responsive nanocarriers: Enhancing drug delivery for tumor therapy. Expert Opin. Drug Deliv. 2023, 20, 1623–1642. [Google Scholar] [CrossRef]
  172. Ji, P.; Xu, Q.; Li, J.; Wang, Z.; Mao, W.; Yan, P. Advances in nanoparticle-based therapeutics for ischemic stroke: Enhancing drug delivery and efficacy. Biomed. Pharmacother. 2024, 180, 117564. [Google Scholar] [CrossRef] [PubMed]
  173. Choudhary, M.; Chaudhari, S.; Gupta, T.; Kalyane, D.; Sirsat, B.; Kathar, U.; Sengupta, P.; Tekade, R.K. Stimuli-Responsive Nanotherapeutics for Treatment and Diagnosis of Stroke. Pharmaceutics 2023, 15, 1036. [Google Scholar] [CrossRef] [PubMed]
  174. Yang, Q.; Pu, W.; Hu, K.; Hu, Y.; Feng, Z.; Cai, J.; Li, C.; Li, L.; Zhou, Z.; Zhang, J. Reactive Oxygen Species-Responsive Transformable and Triple-Targeting Butylphthalide Nanotherapy for Precision Treatment of Ischemic Stroke by Normalizing the Pathological Microenvironment. ACS Nano 2023, 17, 4813–4833. [Google Scholar] [CrossRef]
  175. Li, J.; Zhang, Q.; Wang, W.; Lin, F.; Wang, S.; Zhao, J. Mesenchymal stem cell therapy for ischemic stroke: A look into treatment mechanism and therapeutic potential. J. Neurol. 2021, 268, 4095–4107. [Google Scholar] [CrossRef]
  176. Zhao, Y.H. Essential Role of Chinese Medicines in Mesenchymal Stem Cells Transplantation for Treatment of Ischemic Stroke. Chin. J. Integr. Med. 2019, 25, 723–727. [Google Scholar] [CrossRef]
  177. Jaillard, A.; Hommel, M.; Moisan, A.; Zeffiro, T.A.; Favre-Wiki, I.M.; Barbieux-Guillot, M.; Vadot, W.; Marcel, S.; Lamalle, L.; Grand, S.; et al. Autologous Mesenchymal Stem Cells Improve Motor Recovery in Subacute Ischemic Stroke: A Randomized Clinical Trial. Transl. Stroke Res. 2020, 11, 910–923. [Google Scholar] [CrossRef]
  178. Wang, K.; Rong, L.; Wei, X.; Zhang, Q.; Xiao, L. The effectiveness of various cytotherapeutic strategies for the treatment of ischemic stroke: A Bayesian network meta-analysis of randomized controlled trials. Neurol. Sci. 2020, 41, 1705–1717. [Google Scholar] [CrossRef]
  179. Riopel, M.; Seo, J.B.; Bandyopadhyay, G.K.; Li, P.; Wollam, J.; Chung, H.; Jung, S.R.; Murphy, A.; Wilson, M.; de Jong, R.; et al. Chronic fractalkine administration improves glucose tolerance and pancreatic endocrine function. J. Clin. Investig. 2018, 128, 1458–1470. [Google Scholar] [CrossRef]
  180. Yang, Y.; Ding, T.; Xiao, G.; Huang, J.; Luo, D.; Yue, M.; Su, Y.; Jiang, S.; Zeng, J.; Liu, Y. Anti-Inflammatory Effects of Allocryptopine via the Target on the CX3CL1-CX3CR1 axis/GNB5/AKT/NF-kappaB/Apoptosis in Dextran Sulfate-Induced Mice. Biomedicines 2023, 11, 464. [Google Scholar] [CrossRef]
  181. Lehner, C.; Spitzer, G.; Gehwolf, R.; Wagner, A.; Weissenbacher, N.; Deininger, C.; Emmanuel, K.; Wichlas, F.; Tempfer, H.; Traweger, A. Tenophages: A novel macrophage-like tendon cell population expressing CX3CL1 and CX3CR1. Dis. Model. Mech. 2019, 12, dmm041384. [Google Scholar] [CrossRef]
  182. Li, Y.; Cheng, Q.; Hu, G.; Deng, T.; Wang, Q.; Zhou, J.; Su, X. Extracellular vesicles in mesenchymal stromal cells: A novel therapeutic strategy for stroke. Exp. Ther. Med. 2018, 15, 4067–4079. [Google Scholar] [CrossRef] [PubMed]
  183. Meilandt, W.J.; Ngu, H.; Gogineni, A.; Lalehzadeh, G.; Lee, S.H.; Srinivasan, K.; Imperio, J.; Wu, T.; Weber, M.; Kruse, A.J.; et al. Trem2 Deletion Reduces Late-Stage Amyloid Plaque Accumulation, Elevates the Abeta42:Abeta40 Ratio, and Exacerbates Axonal Dystrophy and Dendritic Spine Loss in the PS2APP Alzheimer’s Mouse Model. J. Neurosci. 2020, 40, 1956–1974. [Google Scholar] [CrossRef] [PubMed]
  184. Pawlitzki, M.; Schreiber, S.; Bittner, D.; Kreipe, J.; Leypoldt, F.; Rupprecht, K.; Carare, R.O.; Meuth, S.G.; Vielhaber, S.; Kortvelyessy, P. CSF Neurofilament Light Chain Levels in Primary Progressive MS: Signs of Axonal Neurodegeneration. Front. Neurol. 2018, 9, 1037. [Google Scholar] [CrossRef] [PubMed]
  185. Mendez-Salcido, F.A.; Torres-Flores, M.I.; Ordaz, B.; Pena-Ortega, F. Abnormal innate and learned behavior induced by neuron-microglia miscommunication is related to CA3 reconfiguration. Glia 2022, 70, 1630–1651. [Google Scholar] [CrossRef]
  186. Shah, S.; Wong, L.M.; Ellis, K.; Bodnar, B.; Saribas, S.; Ting, J.; Wei, Z.; Tang, Y.; Wang, X.; Wang, H.; et al. Microglia-Specific Promoter Activities of HEXB Gene. Front. Cell. Neurosci. 2022, 16, 808598. [Google Scholar] [CrossRef]
  187. Lin, R.; Zhou, Y.; Yan, T.; Wang, R.; Li, H.; Wu, Z.; Zhang, X.; Zhou, X.; Zhao, F.; Zhang, L.; et al. Directed evolution of adeno-associated virus for efficient gene delivery to microglia. Nat. Methods 2022, 19, 976–985. [Google Scholar] [CrossRef]
  188. Masuda, T.; Amann, L.; Sankowski, R.; Staszewski, O.; Lenz, M.; Paolo, D.E.; Snaidero, N.; Costa Jordao, M.J.; Bottcher, C.; Kierdorf, K.; et al. Novel Hexb-based tools for studying microglia in the CNS. Nat. Immunol. 2020, 21, 802–815. [Google Scholar] [CrossRef]
  189. Tanaka, Y.; Hoshino-Negishi, K.; Kuboi, Y.; Tago, F.; Yasuda, N.; Imai, T. Emerging Role of Fractalkine in the Treatment of Rheumatic Diseases. Immunotargets Ther. 2020, 9, 241–253. [Google Scholar] [CrossRef]
  190. Chidambaram, H.; Das, R.; Chinnathambi, S. Interaction of Tau with the chemokine receptor, CX3CR1 and its effect on microglial activation, migration and proliferation. Cell Biosci. 2020, 10, 109. [Google Scholar] [CrossRef]
  191. Bedolla, A.M.; McKinsey, G.L.; Ware, K.; Santander, N.; Arnold, T.D.; Luo, Y. A comparative evaluation of the strengths and potential caveats of the microglial inducible CreER mouse models. Cell Rep. 2024, 43, 113660. [Google Scholar] [CrossRef] [PubMed]
  192. Zhang, W.; Karschnia, P.; von Mucke-Heim, I.A.; Mulazzani, M.; Zhou, X.; Blobner, J.; Mueller, N.; Teske, N.; Dede, S.; Xu, T.; et al. In vivo two-photon characterization of tumor-associated macrophages and microglia (TAM/M) and CX3CR1 during different steps of brain metastasis formation from lung cancer. Neoplasia 2021, 23, 1089–1100. [Google Scholar] [CrossRef] [PubMed]
  193. Maduka, C.V.; Schmitter-Sanchez, A.D.; Makela, A.V.; Ural, E.; Stivers, K.B.; Pope, H.; Kuhnert, M.M.; Habeeb, O.M.; Tundo, A.; Alhaj, M.; et al. Immunometabolic cues recompose and reprogram the microenvironment around implanted biomaterials. Nat. Biomed. Eng. 2024, 8, 1308–1321. [Google Scholar] [CrossRef] [PubMed]
  194. Qiao, H.; Xu, Q.; Xu, Y.; Zhao, Y.; He, N.; Tang, J.; Zhao, J.; Liu, Y. Molecular chaperones in stroke-induced immunosuppression. Neural Regen. Res. 2023, 18, 2638–2644. [Google Scholar] [CrossRef]
  195. Xie, W.; Simats, A.; Guo, Y.; Huang, T.; Sun, X.; Chen, W.; Lin, Y.; Wang, X.; Lai, Z.; Yu, W.; et al. Perspective Review of Myeloid Immune Cell Responses and Poststroke Immunosuppression. Stroke 2023, 54, 1920–1929. [Google Scholar] [CrossRef]
  196. Faura, J.; Bustamante, A.; Miro-Mur, F.; Montaner, J. Stroke-induced immunosuppression: Implications for the prevention and prediction of post-stroke infections. J. Neuroinflamm. 2021, 18, 127. [Google Scholar] [CrossRef]
  197. Wang, J.; Zhang, J.; Ye, Y.; Xu, Q.; Li, Y.; Feng, S.; Xiong, X.; Jian, Z.; Gu, L. Peripheral Organ Injury After Stroke. Front. Immunol. 2022, 13, 901209. [Google Scholar] [CrossRef]
  198. Holl, D.; Hau, W.F.; Julien, A.; Banitalebi, S.; Kalkitsas, J.; Savant, S.; Llorens-Bobadilla, E.; Herault, Y.; Pavlovic, G.; Amiry-Moghaddam, M.; et al. Distinct origin and region-dependent contribution of stromal fibroblasts to fibrosis following traumatic injury in mice. Nat. Neurosci. 2024, 27, 1285–1298. [Google Scholar] [CrossRef]
  199. Kwon, H.R.; Olson, L.E. Imatinib on target in stroke recovery. J. Clin. Investig. 2025, 135, e190024. [Google Scholar] [CrossRef]
  200. Rivas-Fuentes, S.; Salgado-Aguayo, A.; Arratia-Quijada, J.; Gorocica-Rosete, P. Regulation and biological functions of the CX3CL1-CX3CR1 axis and its relevance in solid cancer: A mini-review. J. Cancer 2021, 12, 571–583. [Google Scholar] [CrossRef] [PubMed]
  201. Iwahashi, Y.; Ishida, Y.; Mukaida, N.; Kondo, T. Pathophysiological Roles of the CX3CL1-CX3CR1 Axis in Renal Disease, Cardiovascular Disease, and Cancer. Int. J. Mol. Sci. 2025, 26, 5352. [Google Scholar] [CrossRef] [PubMed]
  202. Kumar, A.; Kitago, T. Pharmacological Enhancement of Stroke Recovery. Curr. Neurol. Neurosci. Rep. 2019, 19, 43. [Google Scholar] [CrossRef] [PubMed]
  203. Zwijnenburg, A.J.; Pokharel, J.; Varnaite, R.; Zheng, W.; Hoffer, E.; Shryki, I.; Comet, N.R.; Ehrstrom, M.; Gredmark-Russ, S.; Eidsmo, L.; et al. Graded expression of the chemokine receptor CX3CR1 marks differentiation states of human and murine T cells and enables cross-species interpretation. Immunity 2023, 56, 1955–1974.e1910. [Google Scholar] [CrossRef]
  204. Zhang, Z.; Yuan, Z.; Wang, Y.; Zhang, Y.H.; Li, Q.; Zeng, X.; Guan, Z.; Bahabayi, A.; Wang, P.; Liu, C. Upregulation of granzyme B and C-X3-C motif receptor 1 in circulating plasmablasts was negatively regulated by Notch signal in patients with systemic lupus erythematosus. J. Leukoc. Biol. 2024, 116, 1061–1071. [Google Scholar] [CrossRef]
  205. Yu, H.; Shen, B.; Han, R.; Zhang, Y.; Xu, S.; Zhang, Y.; Guo, Y.; Huang, P.; Huang, S.; Zhong, Y. CX3CL1-CX3CR1 axis protects retinal ganglion cells by inhibiting microglia activation in a distal optic nerve trauma model. Inflamm. Regen. 2024, 44, 30. [Google Scholar] [CrossRef]
  206. Hiddingh, S.; Pandit, A.; Verhagen, F.; Rijken, R.; Servaas, N.H.; Wichers, R.; Loon, N.; Imhof, S.M.; Radstake, T.; de Boer, J.H.; et al. Transcriptome network analysis implicates CX3CR1-positive type 3 dendritic cells in non-infectious uveitis. eLife 2023, 12, e74913. [Google Scholar] [CrossRef]
  207. Lee, S.; Latha, K.; Manyam, G.; Yang, Y.; Rao, A.; Rao, G. Role of CX3CR1 signaling in malignant transformation of gliomas. Neuro-Oncology 2020, 22, 1463–1473. [Google Scholar] [CrossRef]
  208. Chen, X.; Jiang, M.; Li, H.; Wang, Y.; Shen, H.; Li, X.; Zhang, Y.; Wu, J.; Yu, Z.; Chen, G. CX3CL1/CX3CR1 axis attenuates early brain injury via promoting the delivery of exosomal microRNA-124 from neuron to microglia after subarachnoid hemorrhage. J. Neuroinflamm. 2020, 17, 209. [Google Scholar] [CrossRef]
  209. Kong, J.; Wen, S.; Cao, W.; Yue, P.; Xu, X.; Zhang, Y.; Luo, L.; Chen, T.; Li, L.; Wang, F.; et al. Lung organoids, useful tools for investigating epithelial repair after lung injury. Stem Cell Res. Ther. 2021, 12, 95. [Google Scholar] [CrossRef]
  210. Frydrychowicz, M.; Telec, M.; Aniola, J.; Kazmierski, R.; Chowaniec, H.; Dworacki, G.; Wojtasz, I.; Kozubski, W.; Lukasik, M. The Alteration of Circulating Invariant Natural Killer T, gammadeltaT, and Natural Killer Cells after Ischemic Stroke in Relation to Clinical Outcomes: A Prospective Case-Control Study. Cells 2024, 13, 1401. [Google Scholar] [CrossRef] [PubMed]
  211. Durham, P.G.; Butnariu, A.; Alghorazi, R.; Pinton, G.; Krishna, V.; Dayton, P.A. Current clinical investigations of focused ultrasound blood-brain barrier disruption: A review. Neurotherapeutics 2024, 21, e00352. [Google Scholar] [CrossRef] [PubMed]
  212. Ji, R.; Karakatsani, M.E.; Burgess, M.; Smith, M.; Murillo, M.F.; Konofagou, E.E. Cavitation-modulated inflammatory response following focused ultrasound blood-brain barrier opening. J. Control. Release 2021, 337, 458–471. [Google Scholar] [CrossRef] [PubMed]
  213. Zhao, J.; Hu, H.; Zhou, H.; Zhang, J.; Wang, L.; Wang, R. Reactive oxygen signaling molecule inducible regulation of CRISPR-Cas9 gene editing. Cell Biol. Toxicol. 2023, 39, 2421–2429. [Google Scholar] [CrossRef]
  214. Zhang, Y.; Ling, X.; Su, X.; Zhang, S.; Wang, J.; Zhang, P.; Feng, W.; Zhu, Y.Y.; Liu, T.; Tang, X. Optical Control of a CRISPR/Cas9 System for Gene Editing by Using Photolabile crRNA. Angew. Chem. Int. Ed. Engl. 2020, 59, 20895–20899. [Google Scholar] [CrossRef]
  215. Bonowicz, K.; Jerka, D.; Piekarska, K.; Olagbaju, J.; Stapleton, L.; Shobowale, M.; Bartosinski, A.; Lapot, M.; Bai, Y.; Gagat, M. CRISPR-Cas9 in Cardiovascular Medicine: Unlocking New Potential for Treatment. Cells 2025, 14, 131. [Google Scholar] [CrossRef]
  216. Salomonsson, S.E.; Clelland, C.D. Building CRISPR Gene Therapies for the Central Nervous System: A Review. JAMA Neurol. 2024, 81, 283–290. [Google Scholar] [CrossRef]
  217. Mavroudakis, L.; Golubova, A.; Lanekoff, I. Spatial metabolomics platform combining mass spectrometry imaging and in-depth chemical characterization with capillary electrophoresis. Talanta 2025, 286, 127460. [Google Scholar] [CrossRef]
  218. Zhang, Y.; Zhu, D.; Li, T.; Wang, X.; Zhao, L.; Yang, X.; Dang, M.; Li, Y.; Wu, Y.; Lu, Z.; et al. Detection of acute ischemic stroke and backtracking stroke onset time via machine learning analysis of metabolomics. Biomed. Pharmacother. 2022, 155, 113641. [Google Scholar] [CrossRef]
  219. Zhang, B.; Li, F.; Shi, Y.; Ji, C.; Kong, Q.; Sun, K.; Sun, X. Single-cell RNA sequencing integrated with bulk RNA sequencing analysis reveals the protective effects of lactate-mediated lactylation of microglia-related proteins on spinal cord injury. CNS Neurosci. Ther. 2024, 30, e70028. [Google Scholar] [CrossRef]
  220. Xu, W.; Lin, J.; Gao, M.; Chen, Y.; Cao, J.; Pu, J.; Huang, L.; Zhao, J.; Qian, K. Rapid Computer-Aided Diagnosis of Stroke by Serum Metabolic Fingerprint Based Multi-Modal Recognition. Adv. Sci. 2020, 7, 2002021. [Google Scholar] [CrossRef]
  221. Choi, Y.A.; Park, S.J.; Jun, J.A.; Pyo, C.S.; Cho, K.H.; Lee, H.S.; Yu, J.H. Deep Learning-Based Stroke Disease Prediction System Using Real-Time Bio Signals. Sensors 2021, 21, 4269. [Google Scholar] [CrossRef]
  222. Gellner, A.K.; Reis, J.; Fiebich, B.L.; Fritsch, B. Cx3cr1 deficiency interferes with learning- and direct current stimulation-mediated neuroplasticity of the motor cortex. Eur. J. Neurosci. 2024, 59, 177–191. [Google Scholar] [CrossRef]
  223. Lechtenberg, K.J.; Meyer, S.T.; Doyle, J.B.; Peterson, T.C.; Buckwalter, M.S. Augmented beta2-adrenergic signaling dampens the neuroinflammatory response following ischemic stroke and increases stroke size. J. Neuroinflamm. 2019, 16, 112. [Google Scholar] [CrossRef]
Table 1. Phase-specific dynamics and functional duality of the CX3CR1/CX3CL1 axis in post-stroke pathology.
Table 1. Phase-specific dynamics and functional duality of the CX3CR1/CX3CL1 axis in post-stroke pathology.
PhaseCX3CL1 SourceCX3CR1 DynamicsBeneficial FunctionsDetrimental Functions
HyperacuteDamaged core neurons [7,28]Rapid upregulation in peri-infarct microglia [5,8]Mediates microglial migration [33,34] and debris clearance [5,8]Exacerbates oxidative stress and neuroinflammation [41,42]
AcuteVascular endothelium [43,44,45]Overexpression in Ly6C+ monocytes [52,53]/microglia [49,50,51]Induces IL-10+ anti-inflammatory polarization [49,50,51]Drives monocytes infiltration [52,53] → cytokine storm [47,48]
SubacutePeri-infarct astrocytes [5,71]Sustained overexpression in microglia/MDMs [6,66]Phagocytic clearance [6,66], angiogenesis [77], neurogenesis [64,65,81]Insufficient signaling → glial scarring [67,68,69,70]
ChronicNeurons and astrocytes [24,83,84]Stable overexpression in microglia [85,86]Early phase:
Continues repair [4]
Long-term:
Pathological synaptic pruning [88,89,90], chronic neuroinflammation [51,91,92], Aβ acceleration [93,94]
Table 2. “Stroke phase-biomarkers-targeted interventions” triple precision strategy.
Table 2. “Stroke phase-biomarkers-targeted interventions” triple precision strategy.
PhaseBiomarkersTargeted Interventions
HyperacuteSerum: sCX3CL1↑ + MMP-9↑ [7,118,119]1. Avoid CX3CR1-targeted therapy [100,101,102,103,104,105].
2. Core therapy: Intravenous thrombolysis (≤4.5 h)/thrombectomy (≤6 h) [161,162,163].
3. Adjunct: Short-term non-CX3CR1 immunomodulation [164,165,166].
AcuteCSF: sTREM2↓ + CD36↓ [127,128]pH-responsive nanocarriers delivering rCX3CL1 to penumbra [169,170,171,172,173,174].
SubacutePET-MRI: [11C]PBR28↑ + microglial P2RY12↑ [131,132]Intravenous FKN-Fc fusion protein + MSCs [175,176,177,178,179].
ChronicCSF: sTREM2↑ + NfL↑ [139,140,141,142]1. AAV-HEXB promoter-driven CRISPR-Cas9 CX3CR1 knockout [186,187,188].
2. Tamoxifen-inducible regulation [188].
Table 3. Clinical translation bottlenecks and tech breakthroughs.
Table 3. Clinical translation bottlenecks and tech breakthroughs.
Key Challenges/Breakthrough AreasCritical Issues/Technical InnovationsFuture Directions
Biomarkers Translation Barriers1. Clinically validated standard definitions and testing thresholds remain unavailable
2. Unresolved core regulatory and upstream mechanisms governing the CX3CR1 signaling axis [8]
1. Large-scale clinical studies to validate biomarker utility
2. Mechanistic dissection of CX3CR1 regulatory networks (transcriptional control, signaling preferences)
Immunosuppression RiskImprecise systemic cytokine modulation compromises host defense [194,195,196,197]Integrate dynamic CD4+/CD8+ ratio monitoring [210]
Delivery HurdlesFibrotic scarring impairs penetration [198,199]Develop novel BBB-opening tech (e.g., ultrasound-mediated) [211,212]
Gene Editing EthicsEthical implications of irreversible editingImplement reversible systems (e.g., degron-tagged Cas9) [213,214] + Ban germline editing [215,216]
Cross-Species Temporal Scale DiscrepanciesRodent neural repair windows mismatch human disease progression complexity [205]Humanized model systems (e.g., brain organoids integrated with in vivo imaging) development for precise human brain repair window definition [209]
Spatial Metabolomics ImplementationIn situ mass spectrometry imaging with metabolic flux analysis enables 3D microenvironmental mapping [217,218]Spatiotemporal logic decoding of lactate/HIF-1α/CX3CR1 signaling axis driving microglial phenotypic conversion [219]
Deep Learning-Driven Multi-Omics IntegrationHeterogeneous data synthesis via graph neural networks/transformers establishes “neuroimmune clock” frameworks [220,221]AI-powered prediction of individualized CX3CR1 activity trajectories and optimal intervention windows, defining critical kinetic nodes [222,223]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

He, Q.; Zhou, T.; He, Q. Targeting CX3CR1 Signaling Dynamics: A Critical Determinant in the Temporal Regulation of Post-Stroke Neurorepair. Brain Sci. 2025, 15, 759. https://doi.org/10.3390/brainsci15070759

AMA Style

He Q, Zhou T, He Q. Targeting CX3CR1 Signaling Dynamics: A Critical Determinant in the Temporal Regulation of Post-Stroke Neurorepair. Brain Sciences. 2025; 15(7):759. https://doi.org/10.3390/brainsci15070759

Chicago/Turabian Style

He, Quan, Tong Zhou, and Quanwei He. 2025. "Targeting CX3CR1 Signaling Dynamics: A Critical Determinant in the Temporal Regulation of Post-Stroke Neurorepair" Brain Sciences 15, no. 7: 759. https://doi.org/10.3390/brainsci15070759

APA Style

He, Q., Zhou, T., & He, Q. (2025). Targeting CX3CR1 Signaling Dynamics: A Critical Determinant in the Temporal Regulation of Post-Stroke Neurorepair. Brain Sciences, 15(7), 759. https://doi.org/10.3390/brainsci15070759

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop