v
Search
Advanced

Publications > Journals > Neurosurgical Subspecialties> Article Full Text

  • OPEN ACCESS

Inflammatory Biomarkers in Ischemic Stroke: Mechanisms, Clinical Applications, and Future Directions

  • Yi Yang1,2,
  • Hong Zhu1,2,
  • Tianqing Xiong1,2,3,4,*  and
  • Shun Li4,*
 Author information 

Abstract

Ischemic stroke is a complex cerebrovascular disorder characterized by highly unpredictable outcomes influenced by patient-specific variables, including age, stroke severity, and preventable stroke-related complications such as infections. Analyses of clinical data have indicated a cumulative post-stroke infection rate of approximately 30%, with reported rates ranging from 5% to 65%. Post-stroke infections pose a significant challenge, as they not only increase the financial burden of stroke care but are also associated with adverse clinical outcomes, prolonged hospital stays, and a higher risk of stroke recurrence. The inflammatory response plays a pivotal role in the pathophysiology of ischemic stroke, encompassing the activation of inflammatory cells, the release of inflammatory mediators, and the engagement of inflammatory signaling pathways. Recent advances in molecular biology have facilitated the identification and investigation of numerous inflammation-related biomarkers. This article reviews the roles and mechanisms of key inflammatory biomarkers, including cytokines, chemokines, adhesion molecules, inflammation-related enzymes and mediators, receptors, signaling pathway molecules, and acute-phase proteins in the context of ischemic stroke, highlighting their significance in stroke pathophysiology and prognostic assessment. Additionally, in conjunction with the latest research advances, the article discusses novel biomarkers such as microRNAs and galectin-3, which are emerging as important tools in multiple domains, including diagnosis and treatment. Drawing on clinical diagnostic and therapeutic practices, this review analyzes the diagnostic and therapeutic roles of both novel and traditional biomarkers in the progression of ischemic stroke, following the temporal sequence from disease onset to prognosis. Finally, the article addresses the limitations of current research and offers perspectives on future directions, providing insights that may contribute to the advancement of precision medicine in the management of ischemic stroke.

Keywords

Ischemic stroke, Inflammation, Biomarkers, Cytokines, Chemokines, Acute phase protein, Adhesive molecules, Matrix metalloproteinase, Endovascular treatment

Introduction

Ischemic stroke is characterized by the interruption of cerebral blood flow, leading to localized ischemia and hypoxia in brain tissue, which results in neurological deficits.1,2 The pathophysiological processes involved are complex, encompassing not only direct damage to local brain tissue but also extensive remodeling of neural networks and functional impairments. Key mechanisms include disturbances in energy metabolism, excitotoxicity from excessive release of excitatory amino acids such as glutamate, oxidative stress, and inflammatory responses.3,4 The over-release of excitatory amino acids exacerbates neuronal toxicity, perpetuating a vicious cycle. Additionally, oxidative stress is a critical factor in ischemic stroke; under hypoxic conditions, the generation of reactive oxygen species (ROS) increases, damaging cellular membranes and DNA, and promoting apoptosis.

Following cerebral ischemia, injured neurons and glial cells release a variety of damage-associated molecular patterns (DAMPs), which activate the innate immune system and trigger both local and systemic inflammatory responses.5 In this context, inflammation plays a dual role: it serves as a protective response to injury while also possessing the potential to cause secondary damage through excessive activation.6 The infiltration of inflammatory cells and the release of cytokines not only exacerbate local damage but may also impair the function of distal brain regions, leading to more widespread neural network dysfunction. Therefore, a comprehensive understanding of the dynamic changes in inflammatory responses during ischemic stroke, as well as their interactions with other pathophysiological processes, is crucial for developing novel intervention strategies and improving patient outcomes.

The utilization of inflammatory biomarkers in the investigation of ischemic stroke holds considerable significance. Firstly, these biomarkers can function as early diagnostic tools, facilitating the identification of high-risk patients and enabling timely intervention. Secondly, they can be employed to assess disease severity and predict prognosis, thereby informing personalized treatment strategies.7 Furthermore, certain biomarkers may serve as therapeutic targets, guiding the development of novel treatment approaches. In the domain of inflammation-related biomarkers, researchers have identified numerous potential candidate molecules, including cytokines, chemokines, and acute-phase proteins. These biomarkers exhibit substantial promise for application in the diagnosis, prognostic evaluation, and treatment of ischemic stroke.

This literature review aims to synthesize and critically evaluate the current evidence on classical and emerging inflammatory biomarkers implicated across all phases of ischemic stroke (acute, subacute, and chronic). Specifically, it seeks to: (1) elucidate the pathophysiological mechanisms linking inflammatory cascades to cerebral ischemia-reperfusion injury; (2) assess the clinical efficacy of these biomarkers in stroke diagnosis, prognosis, and monitoring of surgical thrombectomy treatment; and (3) identify novel biomarker candidates with translational potential. By integrating preclinical and clinical research, this review establishes a comprehensive framework to guide future biomarker validation studies and precision medicine approaches in cerebrovascular disease.

Search strategy

A computer-based search was performed on the PubMed database to retrieve articles published up to July 20, 2025. To maximize the specificity and sensitivity of the search, a combination of the following terms was used: inflammation, biomarkers, cytokines, ischemic stroke, chemokines, endovascular treatment, and adhesion molecules. Further screening was conducted through titles and abstracts, and only studies analyzing the role of inflammatory biomarkers in the treatment and prognosis of ischemic stroke were included. There were no restrictions on language or study type. The focus was on articles published within the past 10 years.

The inflammatory response following ischemic stroke exhibits a dual nature

Initially, cell death within the infarcted core occurs rapidly due to energy depletion, resulting in the release of DAMPs such as high mobility group protein B1, adenosine triphosphate, and heat shock proteins.8 These DAMPs engage pattern recognition receptors, including Toll-like receptors (TLR2 and TLR4) and scavenger receptors, thereby initiating innate immune responses.9 For instance, the interaction of high mobility group protein B1 with TLR4 activates the nuclear factor kappa B (NF-κB) signaling pathway, which subsequently induces the expression of pro-inflammatory cytokines such as interleukin (IL)-1 beta, tumor necrosis factor-alpha (TNF-α), and IL-6.10 This cascade promotes the infiltration of peripheral immune cells, leading to disruption of the blood-brain barrier (BBB), exacerbation of cerebral edema, and neuronal cell death. Concurrently, ROS generated from mitochondrial damage further amplify inflammatory signaling through oxidative stress, establishing a detrimental feedback loop.11 Conversely, a moderate inflammatory response is essential for tissue repair and functional recovery. Anti-inflammatory cytokines, including IL-10 and transforming growth factor-beta, play a pivotal role in mitigating excessive inflammation, thereby facilitating nerve regeneration and angiogenesis (Fig. 1).12

The dual role of the inflammatory response in ischemic stroke.
Fig. 1  The dual role of the inflammatory response in ischemic stroke.

Cellular death within the infarcted core occurs rapidly due to energy depletion, resulting in the release of DAMPs such as HMGB1, ATP, and HSPs. These DAMPs engage pattern recognition receptors, including Toll-like receptors (TLR2 and TLR4) and scavenger receptors, thereby initiating innate immune responses. For instance, the interaction of HMGB1 with TLR4 activates the NF-κB signaling pathway, which subsequently induces the expression of pro-inflammatory cytokines such as IL-1β, TNF-α, and IL-6. This cascade promotes the infiltration of peripheral immune cells, leading to disruption of the blood-brain barrier, exacerbation of cerebral edema, and neuronal cell death. Concurrently, ROS generated from mitochondrial damage further amplify inflammatory signaling through oxidative stress, establishing a detrimental feedback loop. Conversely, a moderate inflammatory response is essential for tissue repair and functional recovery. Anti-inflammatory cytokines, including IL-10 and TGF-β, play a pivotal role in mitigating excessive inflammation, thereby facilitating nerve regeneration and angiogenesis. The figure was generated using Figdraw (https://www.figdraw.com/ ). ATP, adenosine triphosphate; DAMPs, damage-associated molecular patterns; HMGB1, high mobility group protein B1; HSPs, heat shock proteins; IL, interleukin; NF-κB, nuclear factor kappa B; ROS, reactive oxygen species; TGF-β, transforming growth factor-beta; TLR2, Toll-like receptor 2; TNF-α, tumor necrosis factor-alpha.

The significance of inflammatory biomarkers in preoperative evaluation and postoperative treatment of post-stroke mechanical thrombectomy

Inflammatory biomarkers are closely associated with the extent of brain injury, thrombus formation, and mechanisms of secondary injury. Post-stroke sympathetic activation induces neutrophilia and lymphocyte apoptosis,13 leading to an elevated neutrophil-to-lymphocyte ratio (NLR), which reflects an imbalance in immune homeostasis. A high NLR level (>4.5) is positively correlated with the risk of stroke-associated pneumonia (odds ratio = 1.55). In patients undergoing endovascular thrombectomy (EVT), an elevated NLR predicts a significantly increased risk of poor 90-day functional outcomes (hazard ratio ≈ 2.0).14

The platelet-to-lymphocyte ratio serves as an effective indicator of platelet activation and systemic inflammatory response. The systemic immune-inflammation index (calculated as platelet count × neutrophil count / lymphocyte count) integratively reflects thrombogenesis and the inflammatory cascade. Studies confirm that platelet-to-lymphocyte ratio > 145 is an independent predictor of increased hemorrhagic transformation risk after intravenous thrombolysis (area under the curve = 0.81).15 Notably, the systemic immune-inflammation index has been demonstrated to be the strongest independent predictor of poor prognosis in EVT patients (area under the curve = 0.85).

Furthermore, elevated fibrinogen levels promote thrombosis, while decreased albumin levels exacerbate oxidative stress injury. Consequently, an increased fibrinogen-to-albumin ratio indicates disruption of BBB integrity.16 Fibrinogen-to-albumin ratio > 0.09 is significantly associated with a higher risk of hemorrhagic transformation (odds ratio = 3.2).

During EVT, neurofilament light chain levels within the ischemic region directly reflect the degree of acute neuronal axonal injury. High neurofilament light chain levels are associated with a 2.4-fold increase in 90-day mortality. Recent research advances, particularly the elucidation of the C-C chemokine receptor type 7 (CCR7) three-dimensional structure,17 have significantly enhanced understanding of its role in disease and its clinical applications. In the regulation of neuroinflammation post-brain injury, CCR7+ T cells (primarily enriched in the central memory T cell subset) play a complex and critical role. Their mediation of immune cell infiltration into the central nervous system via the C-C motif chemokine ligand 19 (CCL19)/CCR7 ligand-receptor axis directly impacts the risk of secondary injury, timing of surgical intervention, and postoperative neurological recovery. Clinical studies reveal that in cerebral ischemia patients, the frequency of infiltrating CD8+GZMK+CCR7+ T cells (often termed T stroke-associated cell cells) shows a significant positive correlation with the National Institutes of Health Stroke Scale score (r = 0.331).17 T stroke-associated cell (TSA) cells promote the release of pro-inflammatory cytokines such as TNF-α and interferon-gamma, activating microglia/macrophages. This activation subsequently enhances the release of matrix metalloproteinases (MMPs) (e.g., MMP-9), leading to BBB disruption and exacerbated cerebral edema.

Therefore, elevated levels of circulating or lesion-localized CCL19+ endothelial cells and CCR7+ T cells represent critical biological markers indicating an active phase of neuroinflammation. Surgical interventions (such as recanalization therapy) performed during this active inflammatory phase may increase the risk of reperfusion injury or secondary hemorrhage. Dynamic monitoring of CCR7+ T cell frequency in peripheral blood or cerebrospinal fluid (e.g., using single-cell RNA sequencing technology) provides a powerful tool for assessing neuroinflammatory intensity. This facilitates individualized, delayed surgical decision-making to optimize patient outcomes (Table 1).18

Table 1

The guiding role of selected inflammatory markers before and after EVT surgery

BiomarkersTesting timingJudgment valueClinical prediction applicationClinical impact
NLR24 h after surgery>4.5SAP poor prognosisOR = 1.55 (SAP)
SIIPreoperative>900×109EVT has poor prognosisAUC = 0.85
FAR24 h before/after surgery>0.09Hemorrhagic transformationOR = 3.29
Local NfLDuring EVT surgery (ischemic area)>9.4 pg/mL90-day mortality rateHR = 2.343
CCR7+ T cellPeripheral blood single-cell sequencing_Reperfusion injuryTargeted peptides can reduce damage

Key inflammatory biomarkers and their mechanisms of action in ischemic stroke

Cytokines and chemokines serve as principal modulators of the inflammatory response.19 In the early stage (within 24 h), activated microglia release IL-1β and TNF-α, which promote neutrophil infiltration through the activation of the NLRP3 inflammasome.20 During the subacute phase (24 h to seven days), monocytes differentiate into pro-inflammatory M1 macrophages, with MMP-9 contributing to BBB disruption.21,22 In the chronic phase (beyond seven days), regulatory T cells mediate immune suppression and upregulate reparative cytokines such as IL-10 and transforming growth factor-beta.23

Acute phase proteins are synthesized in the liver and are modulated by pro-inflammatory cytokines, serving as key inflammatory biomarkers in ischemic stroke.24 C-reactive protein (CRP) is the most prominent acute phase protein, playing a pivotal role in activating the complement system, facilitating inflammatory cell infiltration, and exacerbating oxidative stress following a stroke.25 During the acute phase of stroke (initial hours to 72 h post-onset), CRP levels begin to rise within six to twelve hours after ischemic or hemorrhagic events, peaking within 24 to 48 h, often exceeding 10 mg/L. Mechanistic studies suggest that this elevation is primarily driven by brain tissue injury, which activates microglia and astrocytes, leading to the release of pro-inflammatory mediators such as IL-6 and subsequently stimulating hepatic CRP synthesis.26

In the subacute phase (three to seven days), persistent brain injury, exemplified by secondary infections or exacerbated cerebral edema, may result in sustained elevated CRP levels; conversely, in the absence of such complications, CRP concentrations typically decrease. In the chronic phase (weeks to months), CRP levels gradually return to the normal range (<3 mg/L), although chronic inflammatory conditions, such as atherosclerosis or ongoing infections, may sustain or slightly elevate CRP levels. Elevated serum CRP concentrations have been correlated with stroke severity, increased infarct size, and adverse prognostic outcomes.27

Differences in CRP levels across stroke subtypes can serve as critical biomarkers for clinical differential diagnosis. For instance, patients with ischemic strokes involving large-area infarctions, such as those resulting from large vessel occlusions, exhibit significantly higher CRP levels than those with lacunar infarctions, indicative of small vessel disease.28 In thrombotic strokes, atherothrombotic patients often present with elevated CRP levels, reflecting systemic inflammation associated with unstable atherosclerotic plaques.29 In hemorrhagic strokes, specifically cerebral hemorrhage, a pronounced inflammatory response occurs surrounding the hematoma, and CRP levels may peak more rapidly (within 24 h) and reach higher levels compared to ischemic strokes. CRP levels have also been positively correlated with hematoma expansion and the degree of surrounding edema.

Clinically, CRP has emerged as a valuable biomarker for diagnosing and monitoring stroke across various temporal windows, aiding both acute management and prognostic evaluation due to its rapid, cost-effective, and widely accessible nature, as well as its suitability for dynamic assessment of inflammatory status. Other acute phase proteins, including serum amyloid A and fibrinogen, also play significant roles in the pathophysiology of ischemic stroke.30 Adhesion molecules such as intercellular adhesion molecule-1 and vascular cell adhesion molecule-1 facilitate leukocyte-endothelial interactions, promoting the migration of inflammatory cells into ischemic cerebral tissue.31

The inflammatory response following a stroke involves multiple cytokines, with their dynamic fluctuations closely linked to underlying pathological processes and prognostic outcomes. IL-6 is predominantly produced by microglia, astrocytes, neurons, and peripheral immune cells. Acting as an intracellular signaling pathway activator, IL-6 exerts anti-inflammatory effects slightly earlier than CRP and regulates CRP synthesis. Clinical observations indicate that IL-6 levels rise more rapidly (within 24 h) and reach higher peaks in hemorrhagic stroke compared to ischemic stroke. Studies demonstrate that concurrent assessment of IL-6 and CRP enhances the accuracy of early ischemic stroke diagnosis.32 Patients with large-area infarctions exhibit significantly elevated IL-6 levels compared to those with smaller infarctions. Elevated IL-6 levels correlate with neurological deficits, as measured by the National Institutes of Health Stroke Scale, and are associated with poorer outcomes, as indicated by a modified Rankin Scale score of ≥3.

TNF-α, another prominent pro-inflammatory cytokine, is primarily secreted by activated microglia, macrophages, neurons, and other cell types. TNF-α levels rise during the acute phase (1–12 h post-stroke), peaking between 12 and 24 h. If inflammation persists, TNF-α concentrations remain elevated. Research indicates that TNF-α levels increase earlier in ischemic stroke than in hemorrhagic stroke.33 Clinically, TNF-α may transiently elevate following thrombolysis, which is associated with reperfusion injury.34 Elevated TNF-α levels have been implicated in BBB disruption, cerebral edema, and neuronal apoptosis.

IL-1β is primarily derived from microglia and infiltrating neutrophils.35 During the acute phase of stroke (1–6 h post-onset), IL-1β levels rise rapidly, peaking at approximately 12 h. In the presence of secondary infections or injuries, IL-1β levels may continue to rise. Laboratory studies have shown that IL-1β concentrations in cerebrospinal fluid are significantly higher than those in serum. Clinical case studies indicate that patients with severe pathologies, such as malignant brain edema, demonstrate higher peak IL-1β levels.

MMPs are a family of zinc-dependent endopeptidases that play a multifaceted and crucial role in the pathological processes of ischemic stroke (acute cerebral ischemia). Their involvement includes disruption of the BBB, regulation of the inflammatory response, and mediation of neuronal damage and repair.36

Activation of MMPs in the early stage of ischemia (Acute Phase, 0–24h): Ischemic and hypoxic conditions result in intracellular calcium overload, which activates calcium-dependent proteases such as calpain. This cascade subsequently leads to the activation of pro-MMPs, notably MMP-2 and MMP-9.37 MMP-9, in particular, has garnered significant attention due to its pronounced post-ischemic activity, which correlates with hemorrhagic transformation and adverse prognostic outcomes. Concurrently, ROS and inflammatory mediators, including TNF-α and IL-1β, further stimulate MMP expression. MMP-9 and MMP-3 facilitate the degradation of extracellular matrix components, including collagen, laminin, and tight junction proteins (such as occludin and claudin), leading to BBB disruption, increased vascular permeability, and the onset of cerebral edema and hemorrhagic transformation (hemorrhagic infarction).38

Subacute Phase (24 h–seven Days) – amplification of the inflammatory cascade: MMPs contribute to the infiltration of inflammatory cells, such as neutrophils and macrophages, into ischemic brain tissue. MMP-9 exacerbates neuroinflammation by activating pro-inflammatory cytokines (e.g., IL-1β, IL-6) and chemokines [e.g., C-X-C motif chemokine ligand 12 (CXCL12)].39 Moreover, neuronal damage and apoptosis are facilitated by MMP-3 and MMP-7, which can directly cleave neuronal membrane proteins (e.g., N-cadherin),40 thereby disrupting neuronal connectivity. MMP-9 also contributes to the activation of pro-apoptotic pathways, including caspase-3.

Tissue remodeling and repair during the recovery period (weeks to months): MMPs, including MMP-2 and MMP-12, promote angiogenesis and synaptic plasticity through the degradation of scar tissue.41 However, excessive MMP activation may hinder repair processes. Notably, MMP-9 activates the NF-κB pathway by cleaving the precursor of IL-1β into its active form, thereby amplifying inflammatory signals (Fig. 2).42–47 Simultaneously, MMP-9 degrades extracellular matrices and may contribute to neuroinflammation by increasing monocyte migration into the CNS or by activating chemokines such as CXCL8, which facilitate leukocyte infiltration.48 Some MMPs, such as MMP-8, can degrade pro-inflammatory cytokines (e.g., TNF-α), although this effect is often overshadowed in ischemic conditions. MMP-9 levels in the bloodstream rise significantly 2–6 h post-ischemia, peaking at 24 h, and are associated with infarct volume and clinical prognosis. In contrast, MMP-9 concentrations in cerebrospinal fluid may provide a more sensitive measure, although clinical sampling remains limited.

Temporal dynamics of matrix metalloproteinases (MMPs) in the pathophysiological mechanisms underlying the acute, subacute, and chronic phases of ischemic stroke.
Fig. 2  Temporal dynamics of matrix metalloproteinases (MMPs) in the pathophysiological mechanisms underlying the acute, subacute, and chronic phases of ischemic stroke.

The figure was generated using Figdraw (https://www.figdraw.com/ ). BBB, blood-brain barrier. With the emergence of high-throughput and sensitive technologies, the molecular interaction patterns of long non-coding RNAs in cells have been increasingly elucidated, and they have been continuously linked to disease progression.44,45 Recent investigations have elucidated the significant roles of novel inflammatory regulatory factors in the pathogenesis and progression of ischemic stroke. MicroRNAs (miRNAs), as small non-coding RNAs, exhibit dual functions in the pathological processes associated with ischemic stroke by targeting genes related to inflammation. Among the miRNA family, miR-126 has been shown to preserve BBB integrity and mitigate brain edema and neuronal injury by inhibiting the expression of vascular cell adhesion molecule-1 and pro-inflammatory cytokines such as TNF-α and IL-6.46 Studies indicate that overexpression of miR-126 can reduce inflammatory cell infiltration in the ischemic penumbra, promote angiogenesis, and enhance neurological recovery.47

Similarly, miR-874-3p attenuates neuronal apoptosis and oxidative stress by inhibiting microglial activation and the subsequent release of pro-inflammatory cytokines, including IL-1β and TNF-α.49 In animal models, overexpression of miR-874-3p significantly reduces the extent of cerebral infarction. Conversely, miR-155, recognized as a pro-inflammatory factor, exacerbates BBB disruption, promotes neutrophil infiltration, and amplifies neuroinflammatory responses by downregulating tight junction proteins such as claudin-1 and zonula occludens-1.50

In the context of diagnosis and therapeutic intervention for ischemic stroke, miR-124 and miR-9 exhibit significant declines in serum levels within 24 h post-onset, demonstrating negative correlations with cerebral infarction volume and high-sensitivity C-reactive protein (hs-CRP), suggesting their potential as early diagnostic biomarkers. Additionally, miR-151a-3p and miR-384-5p show abnormal expression profiles in the serum of ischemic stroke patients, correlating with disease progression and neurological dysfunction, and thus may serve as indicators for evaluating thrombolytic therapy efficacy and recurrence risk. Evidence suggests that inhibition of miR-15a/16-1 exerts beneficial effects on the prognosis of ischemic stroke. The promising results of miR-15a/16-1 antagomir across diverse demographic groups highlight its potential as a versatile and effective treatment option (Table 2).51

Table 2

Pathophysiological regulation mechanism and role of small non-coding RNAs in ischemic stroke

Small molecule non-coding RNARegulation mechanismPathophysiological effects
miR-126Inhibit the expression of VCAM-1 and pro-inflammatory cytokines (such as TNF-α, IL-6)Protecting the integrity of BBB, reducing brain edema, and alleviating neuronal damage
miR-874-3pInhibition of microglial activation and subsequent release of pro-inflammatory cytokines (such as IL-1β, TNF-α)Reduce neuronal apoptosis and oxidative stress damage
miR-155Downregulate the expression of tight junction proteins such as claudin-1 and ZO-1Intensify BBB damage, promote neutrophil infiltration and neuroinflammatory response
miR-124Similar to miR-9, there is a significant negative correlation between infarct volume and hs CRP levelsAs an early diagnostic biomarker, it has the potential to evaluate the degree of brain damage and inflammatory status
miR-151a-3pSimilar to miR-384-5p, it is abnormally expressed in the serum of IS patients and is associated with disease progression and the degree of neurological deficitAs a potential biomarker for evaluating the efficacy of thrombolytic therapy and the risk of recurrence
miR-15a/16-1Inhibition of expression can improve stroke prognosis; Antagonists show therapeutic effects in different populationsTargeting neuroprotection and repair therapy to promote neurovascular repair and functional recovery

Future research may focus on targeted delivery systems, such as specific liposomes or exosomes, to administer miRNA mimetics (e.g., miR-126) or antagonists (e.g., miR-155 inhibitors). This approach aims to precisely regulate target gene expression while minimizing non-specific toxicity, extending potential applications beyond stroke to other cardiovascular and cerebrovascular conditions.

Furthermore, agglutinin family proteins represent another class of novel inflammatory regulatory factors contributing to neuroinflammation and BBB disruption following ischemic stroke. After ischemic stroke, galectin-3 (Gal-3) is released by activated microglia and subsequently activates the NF-κB signaling pathway via interaction with TLR-4, promoting inflammatory cytokines such as IL-1β and TNF-α and exacerbating neuronal injury.52,53 Notably, Gal-3 levels are negatively correlated with high-density lipoprotein cholesterol, and its elevated expression can exacerbate dyslipidemia and oxidative stress, accelerating atherosclerotic plaque formation and increasing the risk of ischemic stroke recurrence.

Prospective cohort studies have demonstrated that elevated serum Gal-3 levels are independently associated with a heightened risk of 90-day mortality or severe disability in ischemic stroke patients. Dynamic changes in Gal-3 levels may provide a basis for stratified management of high-risk individuals. Current investigations indicate that Gal-3 inhibitors (e.g., TD139) can reduce microglial activation and neutrophil infiltration in animal models,54 though clinical trials remain preliminary.

Application of inflammatory biomarkers in the diagnosis of ischemic stroke

Inflammatory biomarkers possess significant potential for the early diagnosis of ischemic stroke. Given the acute onset of ischemic stroke, timely diagnosis is critical for effective treatment and improved prognosis. Certain inflammatory markers, including IL-6, CRP, and MMP-9, have been shown to exhibit elevated levels within hours of stroke onset, thereby serving as auxiliary indicators for early diagnosis. Emerging research has also highlighted the potential of novel biomarkers, such as miRNAs and exosome-associated proteins, in facilitating early diagnostic processes.55

Furthermore, inflammatory biomarkers may play a pivotal role in identifying stroke subtypes. Ischemic strokes arising from different etiologies exhibit distinct inflammatory profiles. Studies indicate that patients with atherosclerotic strokes frequently present with elevated levels of CRP and IL-6, whereas those with cardioembolic strokes may exhibit a unique spectrum of inflammatory markers.56 By analyzing specific combinations of inflammatory markers, it may be possible to differentiate between various stroke subtypes, thereby providing a foundation for personalized treatment strategies.

However, the clinical application of inflammatory biomarkers presents several challenges. First, the sensitivity and specificity of individual biomarkers are often inadequate, necessitating the development of multi-biomarker diagnostic models. Second, the inflammatory response is dynamic, emphasizing the importance of determining optimal sampling time points. Additionally, extrinsic factors that may influence biomarker levels, such as infections and autoimmune diseases, must be considered. Future research should prioritize the development of standardized and reproducible detection methodologies, along with the validation of these biomarkers’ diagnostic utility through large-scale clinical trials.

The role of inflammatory biomarkers in prognostic evaluation of ischemic stroke

Inflammatory biomarkers have demonstrated considerable promise in the prognostic assessment of ischemic stroke. Numerous studies have established a close correlation between the levels of specific inflammatory markers and clinical outcomes in stroke patients.57 For instance, elevated levels of CRP, IL-6, and MMP-9 have been associated with poorer neurological recovery and increased mortality rates.58 These biomarkers may reflect a persistent inflammatory state following stroke, indicative of more severe tissue damage and impaired repair mechanisms. Recent studies have highlighted the integral role of peroxisome proliferator-activated receptors (PPARs) in modulating the complex pathological network underlying cerebrovascular diseases and their associated risk factors.59 The activation of PPARγ can reduce the inflammatory response by regulating the production and activation of inflammatory cytokines closely linked to the pathophysiology of intracranial aneurysms.60 Specifically, PPARγ activation can inhibit mononuclear/macrophage cells from releasing various inflammatory cytokines, including IL-1, IL-6, and monocyte chemoattractant protein-1, thereby reducing the occurrence of inflammatory responses.61

Moreover, inflammatory biomarkers may provide critical insights for predicting stroke recurrence. Atherosclerosis, a major contributor to ischemic stroke, is intricately linked to inflammatory processes.62 Research has shown that sustained elevations in biomarkers such as hs-CRP and IL-6 correlate with an increased risk of stroke recurrence.63 These findings suggest that monitoring dynamic fluctuations of inflammatory markers could aid in identifying high-risk patients and informing secondary prevention strategies.

Nonetheless, despite recognition of the association between hs-CRP and ischemic stroke incidence and prognosis since approximately 2017, challenges remain in the clinical application of inflammatory biomarkers for prognostic assessment. First, standardized detection methods and reference ranges are essential. Second, integrating multiple biomarkers may enhance predictive accuracy. Furthermore, large-scale, long-term prospective studies are necessary to validate the prognostic significance of these biomarkers. Future research should focus on developing prognostic scoring systems based on inflammatory markers and incorporating them into clinical decision-making processes to achieve more accurate prognostic evaluations and personalized management of ischemic stroke patients.

The potential value of inflammatory biomarkers in the treatment of ischemic stroke

The potential value of inflammatory biomarkers in managing ischemic stroke is primarily reflected in two key areas: as therapeutic targets and as tools for monitoring and adjusting treatment regimens.

Regarding therapeutic targets, numerous inflammation-related molecules and pathways have emerged as focal points for drug development. For instance, monoclonal antibodies targeting IL-1β and IL-1 receptor antagonists have demonstrated neuroprotective effects in clinical trials.64 Additionally, MMP inhibitors have been investigated for their capacity to mitigate BBB disruption and reduce the risk of hemorrhagic transformation.65 Strategies aimed at modulating microglial polarization and enhancing the expression of anti-inflammatory mediators have also garnered significant attention.

In terms of treatment monitoring and adjustment, inflammatory biomarkers have the potential to evaluate therapeutic efficacy and facilitate personalized approaches. Dynamic assessment of biomarkers post-treatment may provide insights into treatment response and serve as a basis for optimizing therapeutic strategies. Emerging studies are exploring decision-making models based on inflammatory markers to achieve more precise and individualized treatment outcomes. However, clinical implementation faces challenges: patient resistance to research sampling, differences in medical histories, and heterogeneity in biomarker levels often limit statistical significance in studies. More research studies are needed to validate the effectiveness and feasibility of these methodologies.

Despite the promising prospects of inflammation-targeted therapies in ischemic stroke, several challenges persist. First, the inflammatory response is inherently complex and dynamic; thus, interventions aimed at a single target may not yield optimal results. Future research should incorporate multifaceted approaches, including investigating the regulatory effects of CRP gene polymorphisms on post-stroke inflammatory responses and exploring the therapeutic potential of targeting the CRP pathway using CRP inhibitors. Second, it is essential to balance anti-inflammatory treatment with the inflammatory responses necessary for tissue repair and regeneration. Additionally, individual variability and the identification of optimal treatment windows must be taken into account. Consequently, future investigations should prioritize multi-target combination therapies and individualized treatment plans guided by biomarker profiles to enhance both efficacy and safety.

Challenges and future directions

Despite the promising potential of inflammation-targeted therapies, significant challenges remain. Firstly, the inflammatory response is complex and dynamic; thus, interventions targeting a single molecule may not yield optimal outcomes. Future research should employ diverse and integrative approaches, including investigating the regulatory effects of CRP gene polymorphisms on post-stroke inflammation and exploring the therapeutic potential of CRP inhibitors.

Secondly, it is imperative to achieve a balance between anti-inflammatory treatments and the essential inflammatory processes for tissue repair and regeneration. Over-suppression of inflammation may inadvertently hinder recovery. Furthermore, individual patient variability and the identification of optimal treatment windows must be carefully considered to maximize therapeutic efficacy.

Consequently, future investigations should prioritize the development of multi-target combination therapies and individualized treatment plans guided by biomarker profiles. Such approaches hold promise for improving both the efficacy and safety of ischemic stroke interventions.

Conclusions

The inflammatory response plays a pivotal role in the pathophysiology of ischemic stroke. Investigating inflammatory biomarkers offers novel insights and opportunities for precision medicine approaches. This review analyzed the diagnostic and therapeutic roles of both novel and traditional biomarkers throughout ischemic stroke progression. These biomarkers are expected to play an increasingly important role in clinical practice, ultimately improving patient outcomes.

Declarations

Acknowledgement

The authors thank Ms. Xiaohong Wang (Yangzhou University, Jiangsu, China) for providing technical assistance.

Funding

This work was supported by the China Postdoctoral Science Foundation (No. 2022M712689), the Jiangsu Provincial Science and Technology Talent Project (No. FZ20240964), and the Postgraduate Research & Practice Innovation Program of Jiangsu Province (No. KYCX25_4099).

Conflict of interest

The authors declare no conflicts of interest.

Authors’ contributions

Study concept and design (YY), data acquisition (HZ, SL, TX), data analysis and interpretation (YY, HZ, SL, TX), manuscript drafting (YY), manuscript revision (HZ, SL, TX), guarantor of integrity of entire study (SL). All authors reviewed and approved the final version of the manuscript.

References

  1. Payabvash S, Souza LC, Wang Y, Schaefer PW, Furie KL, Halpern EF, et al. Regional ischemic vulnerability of the brain to hypoperfusion: the need for location specific computed tomography perfusion thresholds in acute stroke patients. Stroke 2011;42(5):1255-1260 View Article PubMed/NCBI
  2. Wang C, Yang Y, Xiong T, Li S. Neurovascular unit in ischemic stroke in older adults: a narrative review. Aging Adv 2025;2(1):29-39 View Article
  3. Zhang Y, Yu L, Yang J, Ding Z, He Y, Wan H. Spectrum effect correlation of yangyin tongnao granules on cerebral ischemia-reperfusion injury rats. Front Pharmacol 2022;13:947978 View Article PubMed/NCBI
  4. Chen Q, Xu X, Li S, Xiong T. LncRNA regulation in ischemic stroke and their application prospects. Neural Regen Res 2026;21(3):1058-1073 View Article PubMed/NCBI
  5. Ma Z, Liu CF, Zhang L, Xiang N, Zhang Y, Chu L. The Construction and Analysis of Immune Infiltration and Competing Endogenous RNA Network in Acute Ischemic Stroke. Front Aging Neurosci 2022;14:806200 View Article PubMed/NCBI
  6. Tang H, Li J, Zhou Q, Li S, Xie C, Niu L, et al. Vagus nerve stimulation alleviated cerebral ischemia and reperfusion injury in rats by inhibiting pyroptosis via α7 nicotinic acetylcholine receptor. Cell Death Discov 2022;8(1):54 View Article PubMed/NCBI
  7. Ramiro L, Simats A, García-Berrocoso T, Montaner J. Inflammatory molecules might become both biomarkers and therapeutic targets for stroke management. Ther Adv Neurol Disord 2018;11:1756286418789340 View Article PubMed/NCBI
  8. Iadecola C, Anrather J. The immunology of stroke: from mechanisms to translation. Nat Med 2011;17(7):796-808 View Article PubMed/NCBI
  9. Yuan C, Shentu Y, Ji Q. Research on the innate immune response in transgenic mice following ischemic stroke. Front Aging Neurosci 2024;16:1476913 View Article PubMed/NCBI
  10. Yan X, Cao Y, Chen W, Yu Q, Chen Y, Yao S, et al. Peptide (Tat(48-60)) YVEEL protects against necrotizing enterocolitis through inhibition of toll-like receptor 4-mediated signaling in a phosphatidylinositol 3-kinase/AKT dependent manner. Front Nutr 2022;9:992145 View Article PubMed/NCBI
  11. Cenini G, Lloret A, Cascella R. Oxidative Stress in Neurodegenerative Diseases: From a Mitochondrial Point of View. Oxid Med Cell Longev 2019;2019:2105607 View Article PubMed/NCBI
  12. Li Z, Li Q, Tong K, Zhu J, Wang H, Chen B, et al. BMSC-derived exosomes promote tendon-bone healing after anterior cruciate ligament reconstruction by regulating M1/M2 macrophage polarization in rats. Stem Cell Res Ther 2022;13(1):295 View Article PubMed/NCBI
  13. Xu Q, Ye Y, Wang Z, Zhu H, Li Y, Wang J, et al. NLRP3 Knockout Protects against Lung Injury Induced by Cerebral Ischemia-Reperfusion. Oxid Med Cell Longev 2022;2022:6260102 View Article PubMed/NCBI
  14. Li S, Hu L, Wang J, Zou F, Han B, Wang Y, et al. Prolonged increased neutrophil-to-lymphocyte ratio is associated with mortality after successful revascularization for treatment of acute ischemic stroke. BMC Neurol 2022;22(1):326 View Article PubMed/NCBI
  15. Sun YY, Wang MQ, Wang Y, Sun X, Qu Y, Zhu HJ, et al. Platelet-to-lymphocyte ratio at 24h after thrombolysis is a prognostic marker in acute ischemic stroke patients. Front Immunol 2022;13:1000626 View Article PubMed/NCBI
  16. Ryu JK, Petersen MA, Murray SG, Baeten KM, Meyer-Franke A, Chan JP, et al. Blood coagulation protein fibrinogen promotes autoimmunity and demyelination via chemokine release and antigen presentation. Nat Commun 2015;6:8164 View Article PubMed/NCBI
  17. Bai Y, Ren H, Leng S, Yuan M, Jiang Y, Zhang S, et al. CD8(+)GZMK(+)CD27(+)CCR7(+) T cells mobilized by splenic sympathetic nerves aggravate brain ischemia–reperfusion injury via CCL19-positive endothelial cells. Cell Mol Immunol 2025;22(9):1061-1076 View Article PubMed/NCBI
  18. Jaeger K, Bruenle S, Weinert T, Guba W, Muehle J, Miyazaki T, et al. Structural Basis for Allosteric Ligand Recognition in the Human CC Chemokine Receptor 7. Cell 2019;178(5):1222-1230.e10 View Article PubMed/NCBI
  19. Luster AD. Chemokines—chemotactic cytokines that mediate inflammation. N Engl J Med 1998;338(7):436-445 View Article PubMed/NCBI
  20. Su SH, Wu YF, Lin Q, Wang DP, Hai J. URB597 protects against NLRP3 inflammasome activation by inhibiting autophagy dysfunction in a rat model of chronic cerebral hypoperfusion. J Neuroinflammation 2019;16(1):260 View Article PubMed/NCBI
  21. Yoshida Y, Takagi T, Kuramoto Y, Tatebayashi K, Shirakawa M, Yamahara K, et al. Intravenous Administration of Human Amniotic Mesenchymal Stem Cells in the Subacute Phase of Cerebral Infarction in a Mouse Model Ameliorates Neurological Disturbance by Suppressing Blood Brain Barrier Disruption and Apoptosis via Immunomodulation. Cell Transplant 2021;30:9636897211024183 View Article PubMed/NCBI
  22. Rosell A, Cuadrado E, Ortega-Aznar A, Hernández-Guillamon M, Lo EH, Montaner J. MMP-9-positive neutrophil infiltration is associated to blood-brain barrier breakdown and basal lamina type IV collagen degradation during hemorrhagic transformation after human ischemic stroke. Stroke 2008;39(4):1121-1126 View Article PubMed/NCBI
  23. Gong Y, Liu YC, Ding XL, Fu Y, Cui LJ, Yan YP. Tanshinone IIA Ameliorates CNS Autoimmunity by Promoting the Differentiation of Regulatory T Cells. Neurotherapeutics 2020;17(2):690-703 View Article PubMed/NCBI
  24. Gabay C, Kushner I. Acute-phase proteins and other systemic responses to inflammation. N Engl J Med 1999;340(6):448-454 View Article PubMed/NCBI
  25. Mouliou DS. C-Reactive Protein: Pathophysiology, Diagnosis, False Test Results and a Novel Diagnostic Algorithm for Clinicians. Diseases 2023;11(4):132 View Article PubMed/NCBI
  26. Bourne JH, Suthya AR, Wanrooy BJ, Wilson JL, Wen SW, Bastow CR, et al. Microglia are prominent producers of inflammatory cytokines during the hyperacute phase of ischemic stroke. Commun Biol 2025;8(1):1193 View Article PubMed/NCBI
  27. Wang D, Wang J, Li Z, Gu H, Yang K, Zhao X, et al. C-Reaction Protein and the Severity of Intracerebral Hemorrhage: A Study from Chinese Stroke Center Alliance. Neurol Res 2022;44(4):285-290 View Article PubMed/NCBI
  28. Sikora M, Lewandowska I, Kupc M, Kubalska J, Graban A, Marczak Ł, et al. Serum Proteome Alterations in Human Cystathionine β-Synthase Deficiency and Ischemic Stroke Subtypes. Int J Mol Sci 2019;20(12):3096 View Article PubMed/NCBI
  29. Kondapalli MP, Shiddapur G, Jagdale N, Reddy VKK, Adapa S. The Role of High-Density Lipoprotein, C-reactive Protein, and Serum Ferritin in Ischemic and Hemorrhagic Stroke: An Observational Cross-Sectional Comparative Study. Cureus 2024;16(8):e66606 View Article PubMed/NCBI
  30. Chang Q, Li Y, Xue M, Yu C, He J, Duan X. Serum amyloid A is a potential predictor of prognosis in acute ischemic stroke patients after intravenous thrombolysis. Front Neurol 2023;14:1219604 View Article PubMed/NCBI
  31. Al-Rubiay HF, Al-Kuraishy HM, Al-Gareeb AI. Intercellular adhesive molecule 1(ICAM-1) and acute ischaemic stroke: Role of statins. J Pak Med Assoc 2021;71(12 Suppl 8):S11-S16 PubMed/NCBI
  32. Li X, Lin S, Chen X, Huang W, Li Q, Zhang H, et al. The Prognostic Value of Serum Cytokines in Patients with Acute Ischemic Stroke. Aging Dis 2019;10(3):544-556 View Article PubMed/NCBI
  33. Jiang C, Kong W, Wang Y, Ziai W, Yang Q, Zuo F, et al. Changes in the cellular immune system and circulating inflammatory markers of stroke patients. Oncotarget 2017;8(2):3553-3567 View Article PubMed/NCBI
  34. Bieber M, Schuhmann MK, Volz J, Kumar GJ, Vaidya JR, Nieswandt B, et al. Description of a Novel Phosphodiesterase (PDE)-3 Inhibitor Protecting Mice From Ischemic Stroke Independent From Platelet Function. Stroke 2019;50(2):478-486 View Article PubMed/NCBI
  35. Wang H, Guo W, Liu H, Zeng R, Lu M, Chen Z, et al. Inhibition of inflammatory mediator release from microglia can treat ischemic/hypoxic brain injury. Neural Regen Res 2013;8(13):1157-1168 View Article PubMed/NCBI
  36. Yang Y, Estrada EY, Thompson JF, Liu W, Rosenberg GA. Matrix metalloproteinase-mediated disruption of tight junction proteins in cerebral vessels is reversed by synthetic matrix metalloproteinase inhibitor in focal ischemia in rat. J Cereb Blood Flow Metab 2007;27(4):697-709 View Article PubMed/NCBI
  37. Li YW, Liu Y, Luo SZ, Huang XJ, Shen Y, Wang WS, et al. The significance of calcium ions in cerebral ischemia-reperfusion injury: mechanisms and intervention strategies. Front Mol Biosci 2025;12:1585758 View Article PubMed/NCBI
  38. Salman M, Ismael S, Li L, Ahmed HA, Puchowicz MA, Ishrat T. Endothelial Thioredoxin-Interacting Protein Depletion Reduces Hemorrhagic Transformation in Hyperglycemic Mice after Embolic Stroke and Thrombolytic Therapy. Pharmaceuticals (Basel) 2021;14(10):983 View Article PubMed/NCBI
  39. Asahi M, Asahi K, Jung JC, del Zoppo GJ, Fini ME, Lo EH. Role for matrix metalloproteinase 9 after focal cerebral ischemia: effects of gene knockout and enzyme inhibition with BB-94. J Cereb Blood Flow Metab 2000;20(12):1681-1689 View Article PubMed/NCBI
  40. Brkic M, Balusu S, Libert C, Vandenbroucke RE. Friends or Foes: Matrix Metalloproteinases and Their Multifaceted Roles in Neurodegenerative Diseases. Mediators Inflamm 2015;2015:620581 View Article PubMed/NCBI
  41. Pagano E, Elias JE, Schneditz G, Saveljeva S, Holland LM, Borrelli F, et al. Activation of the GPR35 pathway drives angiogenesis in the tumour microenvironment. Gut 2022;71(3):509-520 View Article PubMed/NCBI
  42. Esnault S, Kelly EA, Johnson SH, DeLain LP, Haedt MJ, Noll AL, et al. Matrix Metalloproteinase-9-Dependent Release of IL-1β by Human Eosinophils. Mediators Inflamm 2019;2019:7479107 View Article PubMed/NCBI
  43. Lakhan SE, Avramut M. Matrix metalloproteinases in neuropathic pain and migraine: friends, enemies, and therapeutic targets. Pain Res Treat 2012;2012:952906 View Article PubMed/NCBI
  44. Yang X, Xiong T, Li S. Role of long noncoding RNAs in angiogenesis-related cerebrovascular disorders and regenerative medicine: a narrative review. Regenerative Medicine Reports 2024;1(2):156-171 View Article
  45. Li S, Qiu N, Ni A, Hamblin MH, Yin KJ. Role of regulatory non-coding RNAs in traumatic brain injury. Neurochem Int 2024;172:105643 View Article PubMed/NCBI
  46. Pan J, Qu M, Li Y, Wang L, Zhang L, Wang Y, et al. MicroRNA-126-3p/-5p Overexpression Attenuates Blood-Brain Barrier Disruption in a Mouse Model of Middle Cerebral Artery Occlusion. Stroke 2020;51(2):619-627 View Article PubMed/NCBI
  47. Qu M, Pan J, Wang L, Zhou P, Song Y, Wang S, et al. MicroRNA-126 Regulates Angiogenesis and Neurogenesis in a Mouse Model of Focal Cerebral Ischemia. Mol Ther Nucleic Acids 2019;16:15-25 View Article PubMed/NCBI
  48. Turner RJ, Sharp FR. Implications of MMP9 for Blood Brain Barrier Disruption and Hemorrhagic Transformation Following Ischemic Stroke. Front Cell Neurosci 2016;10:56 View Article PubMed/NCBI
  49. Xie K, Cai Y, Yang P, Du F, Wu K. Upregulating microRNA-874-3p inhibits CXCL12 expression to promote angiogenesis and suppress inflammatory response in ischemic stroke. Am J Physiol Cell Physiol 2020;319(3):C579-C588 View Article PubMed/NCBI
  50. Butovsky O, Jedrychowski MP, Cialic R, Krasemann S, Murugaiyan G, Fanek Z, et al. Targeting miR-155 restores abnormal microglia and attenuates disease in SOD1 mice. Ann Neurol 2015;77(1):75-99 View Article PubMed/NCBI
  51. Huang X, Li S, Qiu N, Ni A, Xiong T, Xue J, et al. Sex and Age-Dependent Effects of miR-15a/16-1 Antagomir on Ischemic Stroke Outcomes. Int J Mol Sci 2024;25(21):11765 View Article PubMed/NCBI
  52. Burguillos MA, Svensson M, Schulte T, Boza-Serrano A, Garcia-Quintanilla A, Kavanagh E, et al. Microglia-Secreted Galectin-3 Acts as a Toll-like Receptor 4 Ligand and Contributes to Microglial Activation. Cell Rep 2015;10(9):1626-1638 View Article PubMed/NCBI
  53. Liu FT, Stowell SR. The role of galectins in immunity and infection. Nat Rev Immunol 2023;23(8):479-494 View Article PubMed/NCBI
  54. Shan L, Xu K, Ji L, Zeng Q, Liu Y, Wu Y, et al. Injured sensory neurons-derived galectin-3 contributes to neuropathic pain via programming microglia in the spinal dorsal horn. Brain Behav Immun 2024;117:80-99 View Article PubMed/NCBI
  55. Zeng J, Zhu L, Liu J, Zhu T, Xie Z, Sun X, et al. Metformin Protects against Oxidative Stress Injury Induced by Ischemia/Reperfusion via Regulation of the lncRNA-H19/miR-148a-3p/Rock2 Axis. Oxid Med Cell Longev 2019;2019:8768327 View Article PubMed/NCBI
  56. Papadopoulos A, Palaiopanos K, Björkbacka H, Peters A, de Lemos JA, Seshadri S, et al. Circulating Interleukin-6 Levels and Incident Ischemic Stroke: A Systematic Review and Meta-analysis of Prospective Studies. Neurology 2022;98(10):e1002-e1012 View Article PubMed/NCBI
  57. Ritzel RM, Patel AR, Grenier JM, Crapser J, Verma R, Jellison ER, et al. Functional differences between microglia and monocytes after ischemic stroke. J Neuroinflammation 2015;12:106 View Article PubMed/NCBI
  58. Zhang W, Li S, Peng W, Bai L, Zheng Z, Yu J. Correlation of serum IL-6, TNF-α, MMP-9 and CRP levels with prognosis in patients with hypertensive cerebral hemorrhage. Med & Pharm J Chin PLA 2020;32(3):105-108
  59. Yan R, Qiu X, Dai Y, Jiang Y, Gu H, Jiang Y, et al. Association between PPARγ polymorphisms and neurological functional disability of ischemic stroke. J Cereb Blood Flow Metab 2025;45(2):328-339 View Article PubMed/NCBI
  60. Xu X, Chen Q, Li S, Xiong T. Peroxisome proliferator-activated receptors as biomarkers in cerebrovascular diseases: A narrative review. NeuroMarkers 2025;2(1):100035 View Article
  61. Jiang C, Ting AT, Seed B. PPAR-gamma agonists inhibit production of monocyte inflammatory cytokines. Nature 1998;391(6662):82-86 View Article PubMed/NCBI
  62. Libby P, Buring JE, Badimon L, Hansson GK, Deanfield J, Bittencourt MS, et al. Atherosclerosis. Nat Rev Dis Primers 2019;5(1):56 View Article PubMed/NCBI
  63. Wang G, Jing J, Li J, Pan Y, Yan H, Meng X, et al. Association of elevated hs-CRP and multiple infarctions with outcomes of minor stroke or TIA: subgroup analysis of CHANCE randomised clinical trial. Stroke Vasc Neurol 2021;6(1):80-86 View Article PubMed/NCBI
  64. Wu DM, Liu JP, Liu J, Ge WH, Wu SZ, Zeng CJ, et al. Immune pathway activation in neurons triggers neural damage after stroke. Cell Rep 2023;42(11):113368 View Article PubMed/NCBI
  65. Kim GS, Yang L, Zhang G, Zhao H, Selim M, McCullough LD, et al. Critical role of sphingosine-1-phosphate receptor-2 in the disruption of cerebrovascular integrity in experimental stroke. Nat Commun 2015;6:7893 View Article PubMed/NCBI

About this Article

Cite this article
Yang Y, Zhu H, Xiong T, Li S. Inflammatory Biomarkers in Ischemic Stroke: Mechanisms, Clinical Applications, and Future Directions. Neurosurgical Subspecialties. Published online: Oct 4, 2025. doi: 10.14218/NSSS.2025.00029.
Copy        Export to RIS        Export to EndNote
Article History
Received Revised Accepted Published
June 7, 2025 September 1, 2025 September 2, 2025 October 4, 2025
DOI http://dx.doi.org/10.14218/NSSS.2025.00029
  • Neurosurgical Subspecialties
  • pISSN 0000-0000
  • eISSN 3067-6150
Back to Top

Inflammatory Biomarkers in Ischemic Stroke: Mechanisms, Clinical Applications, and Future Directions

Yi Yang, Hong Zhu, Tianqing Xiong, Shun Li
  • Reset Zoom
  • Download TIFF