Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Oncology Letters
      • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Biomedical Reports
      • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • Information for Authors
    • Information for Reviewers
    • Information for Librarians
    • Information for Advertisers
    • Conferences
  • Language Editing
Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • For Authors
    • For Reviewers
    • For Librarians
    • For Advertisers
    • Conferences
  • Language Editing
Login Register Submit
  • This site uses cookies
  • You can change your cookie settings at any time by following the instructions in our Cookie Policy. To find out more, you may read our Privacy Policy.

    I agree
Search articles by DOI, keyword, author or affiliation
Search
Advanced Search
presentation
Molecular Medicine Reports
Join Editorial Board Propose a Special Issue
Print ISSN: 1791-2997 Online ISSN: 1791-3004
Journal Cover
March-2026 Volume 33 Issue 3

Full Size Image

Sign up for eToc alerts
Recommend to Library

Journals

International Journal of Molecular Medicine

International Journal of Molecular Medicine

International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.

International Journal of Oncology

International Journal of Oncology

International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.

Molecular Medicine Reports

Molecular Medicine Reports

Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.

Oncology Reports

Oncology Reports

Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.

Oncology Letters

Oncology Letters

Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.

Biomedical Reports

Biomedical Reports

Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.

Molecular and Clinical Oncology

Molecular and Clinical Oncology

International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.

World Academy of Sciences Journal

World Academy of Sciences Journal

Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.

International Journal of Functional Nutrition

International Journal of Functional Nutrition

Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.

International Journal of Epigenetics

International Journal of Epigenetics

Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.

Medicine International

Medicine International

An International Open Access Journal Devoted to General Medicine.

Journal Cover
March-2026 Volume 33 Issue 3

Full Size Image

Sign up for eToc alerts
Recommend to Library

  • Article
  • Citations
    • Cite This Article
    • Download Citation
    • Create Citation Alert
    • Remove Citation Alert
    • Cited By
  • Similar Articles
    • Related Articles (in Spandidos Publications)
    • Similar Articles (Google Scholar)
    • Similar Articles (PubMed)
  • Download PDF
  • Download XML
  • View XML
Review Open Access

Interleukin‑6 and ischemic stroke: From mechanisms to clinical prospects (Review)

  • Authors:
    • Xinyue Wang
    • Xiaohang Zhang
    • Jianing Lin
    • Ping Lin
  • View Affiliations / Copyright

    Affiliations: Department of Neurology, Xinhua Hospital Affiliated to Dalian University, Dalian, Liaoning 116021, P.R. China, Department of Rehabilitation, Xinhua Hospital Affiliated to Dalian University, Dalian, Liaoning 116021, P.R. China
    Copyright: © Wang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
  • Article Number: 80
    |
    Published online on: January 5, 2026
       https://doi.org/10.3892/mmr.2026.13790
  • Expand metrics +
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Metrics: Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )
Cited By (CrossRef): 0 citations Loading Articles...

This article is mentioned in:


Abstract

Neuroinflammation is a central component of the pathophysiology of ischemic stroke (IS). Suppressing excessive inflammatory responses after stroke can markedly improve patient outcomes. Interleukin‑6 (IL‑6), a key mediator of the inflammatory cascade, serves a notable role in the pathological process of acute IS through multiple mechanisms. Elevated serum IL‑6 levels serve as an important biomarker for predicting the onset and recurrence of IS and are closely associated with disease severity and prognosis. Anti‑inflammatory interventions are notably important during the acute phase and secondary prevention of stroke. Currently, therapeutic strategies targeting the IL‑6/IL‑6R signaling axis are under investigation and have shown promising clinical potential. The present review summarizes the important role of IL‑6 in neuroinflammation associated with IS, its association with disease severity and prognosis and previous advances in anti‑inflammatory therapeutic strategies targeting the IL‑6/IL‑6R pathway during both the acute phase and secondary prevention of IS.

Introduction

A 2024 study in ‘The Lancet’ reported that, in 2021, stroke accounted for almost 50% of cardiovascular disease-related mortalities in the Western Pacific region, making it a leading cause of mortality. As populations age, stroke-related mortalities and disabilities are projected to rise, imposing an increasing burden on healthcare systems (1,2). Ischemic stroke (IS) represents 70–80% of all strokes. Mounting evidence suggests that persistent systemic inflammation after IS promotes infarct expansion, worsens neurological deficits and aggravates brain injury (3–5). Studies on post-stroke inflammation indicate that attenuating acute inflammatory responses can limit cerebral damage, while anti-inflammatory therapies may reduce recurrence in patients with residual inflammation (6,7). Interleukin (IL)-6, a key mediator of the inflammatory cascade following IS, serves a central role in its pathophysiological progression (8). Advances in neuroimmunology in the field of IS mean that elucidating the mechanism of action of IL-6, investigating its association with IS and developing IL-6-targeted interventions could lead to new methods for improving the outcomes for patients with IS.

Pathological mechanism of acute IS (AIS)

Following IS, the brain rapidly initiates the ischemic cascade, in which disrupted energy metabolism triggers excitotoxicity and oxidative stress, leading to neuronal injury and the release of damage-associated molecular patterns (DAMPs) (9,10). DAMPs activate downstream signaling through pattern recognition receptors (PRRs), inducing microglial M1 polarization and the production of pro-inflammatory cytokines, including TNF-α and IL-6 (11,12). Concurrently, activated neutrophils release reactive oxygen species (ROS), amplifying inflammation and tissue damage (13,14). These processes collectively disrupt the blood-brain barrier (BBB), cause microcirculatory failure and promote vasogenic cerebral edema. Neuroinflammation is not confined to the infarcted region but propagates throughout the brain, persistently impairing neurological recovery (15). In the acute phase of IS, widespread pathological alterations, such as excitotoxicity, oxidative stress and neuronal apoptosis, as well as systemic responses, including autonomic dysfunction, hypothalamic/pituitary/adrenal (HPA) axis overactivation and immune dysregulation, further aggravate IS-induced injury (9,16) (Fig. 1).

AIS-related pathological alterations
within and beyond the brain. (A) Impaired cerebral energy
metabolism. (B) Excitotoxic effects of Glu at synaptic sites. (C)
Extracerebral pathological alterations, including autonomic
dysfunction, hyperactivation of the HPA axis and systemic immune
dysregulation. (D) Increased neuronal apoptosis. (E) Oxidative
stress-induced neuronal injury. (F) Excessive neuroinflammation.
Created with BioRender.com and used with permission under
BioRender's Academic License. ATP, adenosine triphosphate; Glu,
glutamate; ROS, reactive oxygen species; DAMPs, damage-associated
molecular patterns; HPA, hypothalamic/pituitary/adrenal axis; AIS,
acute ischemic stroke; HPA, hypothalamic/pituitary/adrenal.

Figure 1.

AIS-related pathological alterations within and beyond the brain. (A) Impaired cerebral energy metabolism. (B) Excitotoxic effects of Glu at synaptic sites. (C) Extracerebral pathological alterations, including autonomic dysfunction, hyperactivation of the HPA axis and systemic immune dysregulation. (D) Increased neuronal apoptosis. (E) Oxidative stress-induced neuronal injury. (F) Excessive neuroinflammation. Created with BioRender.com and used with permission under BioRender's Academic License. ATP, adenosine triphosphate; Glu, glutamate; ROS, reactive oxygen species; DAMPs, damage-associated molecular patterns; HPA, hypothalamic/pituitary/adrenal axis; AIS, acute ischemic stroke; HPA, hypothalamic/pituitary/adrenal.

Notably, inflammation exerts a dual role in regulating IS-induced neuronal injury: Excessive early responses exacerbate ischemic damage, whereas moderate inflammation during later phases facilitates debris clearance, glial scar formation and neuroregeneration (17,18). Hence, timely modulation of acute inflammation may slow infarct core expansion and alleviate ischemic brain injury (19).

IL-6

IL-6 is a globular glycoprotein composed of 184 amino acids, with a molecular weight of 21–30 kDa. IL-6 contains two N-glycosylation sites and four conserved cysteine residues that form a stable antiparallel α-helical structure, maintained by hydrophobic interactions and disulfide bonds, which are important for receptor binding and signal transduction. IL-6 has three receptor-binding sites, one for IL-6 receptor (IL-6R) and two for glycoprotein 130 (gp130), and is primarily secreted by macrophages and activated T cells, although endothelial cells and hepatocytes also contribute to its production (20,21).

IL-6 mediates three distinct signaling modes via two types of receptors. The membrane-bound IL-6R (mIL-6R) activates the classical pathway, which exerts regenerative and anti-inflammatory effects, whereas the soluble IL-6R (sIL-6R) triggers trans-signaling, which is predominantly pro-inflammatory (22,23). Both pathways require gp130 for downstream activation (24,25). Because gp130 is ubiquitously expressed, trans-signaling can occur in nearly all cell types, amplifying inflammation (26). Additionally, in trans-presentation, IL-6 bound to dendritic cells is presented to naïve CD4+ T cells, inducing their differentiation into pro-inflammatory T helper 17 cells (27). Notably, neurons respond to IL-6 only in the presence of sIL-6R (26).

Through the Janus kinase (JAK)/STAT pathway and other related pathways, IL-6 regulates cell proliferation, differentiation and immune homeostasis, serving as an important bridge between innate and adaptive immunity (8). Under physiological conditions, IL-6 levels in the central nervous system are minimal but rise sharply during AIS (28). Notable cellular sources of IL-6 include neurons within the ischemic core, endothelial cells, microglia and astrocytes (29). Endothelial cell-derived IL-6 and chemokines recruit neutrophils, which cleave mIL-6R to generate sIL-6R, thereby amplifying endothelial trans-signaling and inflammatory responses (22). Consequently, serum IL-6 levels are markedly elevated in patients with AIS (30), and clinical studies have demonstrated a strong association between IL-6 concentration and stroke risk (31). The present review provides a detailed overview of the pathological mechanisms by which IL-6 contributes to AIS progression.

IL-6 participates in the pathological mechanism of AIS

IL-6 in oxidative stress

Oxidative stress refers to a disruption of redox homeostasis, characterized by excessive oxidant activity leading to cellular injury. Cerebral ischemia and reperfusion provoke cellular responses that generate excessive ROS (32). When ROS production surpasses cellular antioxidant capacity, it triggers lipid peroxidation and protein and DNA oxidation, also activating redox-sensitive kinases such as IKKβ, thereby initiating inflammatory and cell death signaling pathways (33).

Under ischemic conditions, ROS have been shown to promote neuroinflammation and activate the NACHT, LRR and PYD domains-containing protein 3 (NLRP3) inflammasome, while also inducing IL-6 expression through NF-κB activation in vascular smooth muscle cells (34). In turn, IL-6 upregulates NADPH oxidase 4 via the JAK/STAT3 pathway, sustaining ROS production (35). This ROS/IL-6 positive feedback loop amplifies oxidative stress and aggravates ischemic brain injury.

IL-6 in inflammatory responses

Following cerebral ischemia, DAMPs released from injured neurons activate inflammatory cascades through PRRs (6). PRR-mediated activation of Toll-like receptor (TLR) 2 and TLR4 initiates myeloid differentiation primary response protein MYD88 (MYD88)- and TIR domain-containing adapter molecule 1 (TRIF)-dependent signaling pathways, leading to NF-κB activation and the subsequent secretion of pro-inflammatory cytokines, such as IL-1β and IL-6 (6,11,15). Activated microglia and astrocytes further amplify the inflammatory response by releasing IL-6, which stimulates the JAK/STAT3 pathway (11). This results in STAT3 phosphorylation and nuclear translocation, driving the transcription of pro-inflammatory genes and aggravating neuronal injury and repair impairment (Fig. 2).

Inflammatory response in AIS. (A)
Following cerebral blood flow interruption, ischemic neurons
release DAMPs, which activate resident glial cells and induce the
secretion of numerous inflammatory mediators. This cascade
upregulates chemokines and adhesion molecules, leading to BBB
disruption. (B) DAMPs activate TLR2 and TLR4, triggering MYD88- and
TRIF-dependent signaling pathways that converge on NF-κB
activation, thereby promoting the expression of inflammatory
cytokines such as IL-1β, IL-6 and TNF-α. (C) Pro-inflammatory
cytokines activate the JAK/STAT3 pathway, amplifying the
inflammatory response and driving microglial polarization toward
the M1 phenotype. (D) IL-6 further acts on vascular endothelial
cells to promote atherosclerotic processes. Created with
BioRender.com and used with permission under BioRender's Academic
License. AIS, acute ischemic stroke; DAMPs, damage-associated
molecular patterns; BBB, blood-brain barrier; TLR, Toll-like
receptor; TRIF, TIR domain-containing adapter molecule 1; JAK,
Janus kinase.

Figure 2.

Inflammatory response in AIS. (A) Following cerebral blood flow interruption, ischemic neurons release DAMPs, which activate resident glial cells and induce the secretion of numerous inflammatory mediators. This cascade upregulates chemokines and adhesion molecules, leading to BBB disruption. (B) DAMPs activate TLR2 and TLR4, triggering MYD88- and TRIF-dependent signaling pathways that converge on NF-κB activation, thereby promoting the expression of inflammatory cytokines such as IL-1β, IL-6 and TNF-α. (C) Pro-inflammatory cytokines activate the JAK/STAT3 pathway, amplifying the inflammatory response and driving microglial polarization toward the M1 phenotype. (D) IL-6 further acts on vascular endothelial cells to promote atherosclerotic processes. Created with BioRender.com and used with permission under BioRender's Academic License. AIS, acute ischemic stroke; DAMPs, damage-associated molecular patterns; BBB, blood-brain barrier; TLR, Toll-like receptor; TRIF, TIR domain-containing adapter molecule 1; JAK, Janus kinase.

In AIS, endothelial injury and BBB disruption promote sustained NF-κB activation, which enhances IL-6 production. Elevated IL-6 in turn further amplifies NF-κB-mediated inflammatory signaling, forming a positive feedback loop that exacerbates neuroinflammation and tissue damage (36). Peripheral immune cells infiltrate through the disrupted BBB, propagating local inflammation into a widespread cerebral response (15). IL-6 promotes microglial M1 polarization via the JAK1/2/STAT1/3 pathway and upregulates endothelial adhesion molecules, enhancing neutrophil recruitment and amplifying local inflammation. Furthermore, IL-6 induces endothelial cadherin disassembly and complement C5a receptor expression, further compromising BBB integrity (37). It also activates platelets, facilitating aggregation and contributing to thrombosis by impairing endothelial function (38,39). Activated M1-type microglia secrete pro-inflammatory cytokines, including IL-1β, IL-6 and TNF-α, during the acute phase of IS. These cytokines have a combined destructive effect, promoting neuronal death, disrupting the BBB and causing overall brain tissue damage (40). Clinically, serum IL-6 levels are independently associated with infarct volume (41).

Over the long term, as a key pro-inflammatory mediator, IL-6 accelerates atherosclerotic plaque progression by activating vascular endothelial cells, forming a notable pathological basis for large-artery atherosclerotic stroke (42,43). Through its classical signaling pathway, IL-6 also stimulates hepatic synthesis of C-reactive protein (CRP); both IL-6 and CRP serve as established biomarkers for systemic inflammation and atherosclerotic cardiovascular disease (ASCVD) risk assessments.

IL-6 in neuronal death

In a hypoxia-ischemia (HI) rat model, small interfering RNA-mediated silencing of the IL-6 gene markedly inhibited the activation of the pro-apoptotic protein Bax and the apoptotic executor protein caspase-3, thereby reducing brain injury and neuronal apoptosis (44). In both middle cerebral artery occlusion (MCAO) and oxygen-glucose deprivation/reoxygenation models, targeting IL-6R was found to block IL-6 signaling, resulting in reduced expression of the pro-apoptotic proteins Bax and caspase-3 and upregulation of the anti-apoptotic protein Bcl-2. This was shown to ultimately lead to the inhibition of neuronal apoptosis (45). Following IS, inflammatory factors induce neuronal and glial cell apoptosis by regulating apoptosis-related genes and signaling pathways, and this apoptosis is one of the key mechanisms underlying brain injury (44,45).

IL-6 in extracerebral pathophysiology

Following AIS, activation of the HPA axis promotes the release of corticotropin-releasing hormone (CRH) and adrenocorticotropic hormone, markedly increasing cortisol (CORT) levels, which disrupt BBB integrity, facilitate inflammatory cytokine entry into brain tissue and exacerbate neuroinflammation (46). Stroke also induces sympathetic dominance and systemic immune dysregulation, characterized by increased gut permeability, translocation of bacterial components into the bloodstream and dysfunction of peripheral immune organs, such as the bone marrow and thymus (16).

Under physiological conditions, CORT exerts anti-inflammatory effects; however, chronic HPA axis hyperactivation leads to excessive CORT release, resulting in sustained activation of microglia and astrocytes. Elevated IL-6 suppresses HPA axis negative feedback, maintaining its hyperactive state (46). IL-6 also promotes CORT secretion either by directly stimulating hypothalamic CRH release or by enhancing pituitary sensitivity to CRH (47). Prolonged CORT upregulation may also induce glucocorticoid receptor resistance, leading to increased secretion of pro-inflammatory cytokines such as IL-6 (48).

Patients with diabetes frequently exhibit HPA axis dysregulation and elevated CORT levels (49). A study performed by Kim et al (50) reported that in diabetic mice subjected to MCAO, the HPA axis was markedly overactivated, with mice displaying elevated IL-6, IL-1β and TNF-α expression. Treatment with metyrapone, a glucocorticoid synthesis inhibitor, subsequently reduced IL-6 expression and infarct volume. These findings suggest that under diabetic conditions, IL-6 may exacerbate stroke-related inflammation by modulating the HPA axis, providing an extracerebral mechanism linking metabolic and neuroinflammatory pathways and offering theoretical support for IL-6-targeted strategies to improve stroke outcomes in diabetic patients.

Role of IL-6 in IS

IL-6 and the risk of IS

IL-6 serves a notable role in atherosclerotic development and progression, as well as in the pathogenesis of associated complications. Persistent elevation of IL-6 is a notable risk factor for atherosclerotic and thrombotic vascular events (42). Clinical and imaging studies have demonstrated the potential of IL-6 in predicting carotid plaque severity, instability and progression (51,52). As a key pro-inflammatory cytokine, circulating IL-6 reflects chronic low-grade inflammation that promotes atherothrombosis. Elevated IL-6 concentrations are consistently associated with increased risks of coronary artery disease, stroke and myocardial infarction (52–54). However, the standardization of IL-6 measurement remains a core bottleneck in clinical practice, constraining its application (8). Unified testing standards must be established; otherwise, the adoption of IL-6 measurements in routine clinical practice will remain limited.

In IS, IL-6 demonstrates notable prognostic potential. Large-scale prospective studies, including the Reasons for Geographic and Racial Differences in Stroke cohort, have shown a dose-dependent relationship between baseline IL-6 levels and future IS risk, independent of conventional vascular risk factors (55,56). In AIS, higher IL-6 levels are associated with multiple acute infarctions (43), greater recurrence risk (57) and worse functional outcomes (58,59). Kaplan-Meier analyses further reveal that IL-6 levels above specific thresholds at AIS onset predict higher rates of stroke recurrence (30). Individual-participant data meta-analyses have corroborated these findings, showing that IL-6 independently predicts post-stroke vascular events regardless of underlying atherosclerosis (60).

Genetic evidence indicates that reduced IL-6 pathway activity markedly lowers the risk of IS, particularly large artery atherosclerosis (LAA) and small artery occlusion (SAO) subtypes, as well as coronary and peripheral artery disease, and this protective association, consistently observed in both European and East Asian populations, is also linked to a lower incidence of type 2 diabetes (61). Furthermore, clinical findings of IL-6 inhibition with ziltivekimab align with these genetic observations, providing mechanistic support for ongoing phase III trials and highlighting IL-6 as a promising therapeutic target for cardiovascular-metabolic disease prevention and treatment (62). Table I (43,51,54–61) lists the relevant studies.

Table I.

Association between IL-6 and IS risk.

Table I.

Association between IL-6 and IS risk.

First author, yearStudy type (study ID)Population/nFollow-up duration, yearsFindings(Refs.)
Jenny et al, 2019Case-cohort study (REGARDS)IS/557; CohS/951N/AHigher baseline IL-6 levels are notably associated with future IS risk.(55)
Georgakis et al, 2020Mendelian randomization (MEGASTROKE)IS/34,217; GP/404,630N/AGenetic downregulation of IL-6 signaling reduces the risk of IS, particularly in the LAA and SAO subtypes, but is not associated with the CE subtype.(61)
Xu et al, 2022Observational (CNSR-III)AIS + TIA/9,7331.00For patients with LAA and SAO-type IS, elevated IL-6 levels are closely associated with residual recurrence risk after secondary prevention.(58)
Li et al, 2022Observational (CNSR-III)AIS + TIA/10,4721.00Acute-phase IL-6 levels have been demonstrated to be associated with an elevated risk of recurrence and unfavorable functional outcomes.(59)
Papadopoulos et al, 2022Prospective cohort and meta-analysisGP/27,41112.40IL-6 levels are linearly associated with IS risk.(56)
Kamtchum-Tatuene et al, 2022Prospective cohort (CHS)GP/4,3345.00IL-6 is an independent predictor of carotid plaque severity, fragility and progression.(51)
Mo et al, 2024Prospective cohort (CNSR-III)sICAD/1,919N/AHigher IL-6 levels are markedly associated with the incidence of MAIs.(43)
Jiang and Fan, 2024Prospective cohortAIS/3050.25IL-6 levels are closely related to recurrence and have good predictive value.(57)
Kitagawa et al, 2025post-hoc analysis (TWMU-CVD)CVD/4714.60Serum IL-6 levels can predict IS and cardiovascular events in patients with vascular risk factors.(54)
McCabe et al, 2025 Individual-participant data meta-analysisIS + TIA/10,148N/AIL-6 was associated with post-stroke recurrence irrespective of atherosclerosis.(60)

[i] REGARDS, Reasons for Geographic and Racial Differences in Stroke; CHS, cardiovascular health study; TWMU-CVD, Tokyo Women's Medical University Cardiovascular Disease Study; CNSR-III, China National Stroke Registry-III; sICAD, symptomatic intracranial atherosclerotic disease; AIS, acute ischemic stroke; IS, ischemic stroke; CohS, cohort sample; GP, general population; CVD, cardiovascular disease; NA, not available; LAA, large artery atherosclerosis; SAO, small artery occlusion; CE, cardioembolic stroke; MAIs, multiple acute infarctions; TIA, transient ischemic attack.

IL-6 and the severity and prognosis of AIS

The intensity of the inflammatory response in AIS is closely associated with disease severity. Numerous studies have provided evidence that higher serum or cerebrospinal fluid levels of pro-inflammatory cytokines, particularly IL-6, associate with greater neurological deficits, larger infarct volumes and worse functional outcomes (63–77). Compared with healthy individuals, patients with AIS show markedly elevated serum IL-6 levels, and IL-6-based prediction models for AIS demonstrate strong diagnostic and prognostic performance (63). Experimental evidence also indicates that IL-6 measurements can help estimate stroke onset time (64). Ischemia, hypoxia, oxidative stress, vascular occlusion and inflammation itself can all induce IL-6 upregulation, which in turn promotes hepatic acute-phase response protein synthesis, leukocyte recruitment and thrombogenesis, thereby exacerbating ischemic injury (65). Prospective studies have further validated the clinical relevance of IL-6. Elevated IL-6 levels reflect acute disease severity and independently predict short-term functional outcomes (66–70). In patients undergoing endovascular therapy (EVT) for large-vessel occlusion, both circulating and intrathrombus IL-6 concentrations are notably associated with poor prognosis (71). Longitudinal studies have demonstrated that dynamic IL-6 changes mirror the intensity and extent of post-stroke inflammation: Patients with lower IL-6 levels before thrombolysis achieve improved 90-day outcomes compared with patients with high IL-6 levels, suggesting that IL-6 may serve as a potential marker of neurorecovery (72). After reperfusion, a decline in patient IL-6 levels indicates successful recanalization and neurological stabilization, whereas persistent elevation of IL-6 reflects failed or incomplete reperfusion (73).

IL-6 also exhibits early warning value for early neurological deterioration (END). Serial monitoring shows that increased IL-6 levels associate with worsening National Institutes of Health Stroke Scale scores and adverse neurological evolution (74–77). In EVT-treated patients, elevated postoperative IL-6 independently predicts END, underscoring its role as a quantitative biomarker for early IS risk stratification and treatment optimization (77). Collectively, serum IL-6 represents a reliable indicator of AIS severity and prognosis, and early dynamic monitoring of IL-6 levels may facilitate the identification of high-risk patients and guide individualized therapeutic strategies. Table II (41,66–77) lists the relevant studies.

Table II.

Association of IL-6 with the severity and prognosis of AIS.

Table II.

Association of IL-6 with the severity and prognosis of AIS.

First author, yearPopulation/nIL-6 measurement timeFindings(Refs.)
Li et al, 2019AIS/180Within 72 h after symptom onsetAcute-phase IL-6 level is an independent risk predictor, associated with stroke severity, short-term outcome and infarct volume.(68)
Alfieri et al, 2020AIS/176; GP/176;Within 24 h after admissionPre-stroke disability (baseline mRS ≥3) is associated with higher levels of ferritin, IL-6, hs-CRP, WBC, ESR and glucose.(67)
Hervella et al, 2020AIS 4295At admission and 24 h laterAt 24 h, IL-6 levels were markedly higher in PNR and PWER groups than in the PER group.(73)
Mosarrezaii et al, 2020AIS/60Day 1 and 5 after onsetIL-6 level is associated with AIS severity and may serve as a prognostic indicator.(66)
Purroy et al, 2021AIS/332Within 24 h after onsetSerum IL-6 level is independently associated with infarct volume.(41)
Deng et al, 2021LVO with EVT/210Days 1, 2, 3 and 7 after EVTIL-6 may be a potential marker for END after EVT.(77)
Pawluk et al, 2022AIS 125Before thrombolysis, and atSerum IL-6 levels are notably lower in patients with promising outcomes. 24 h and 7 days(72)
Sabir Rashi et al, 2022AIS 101Within 24 h of admissionIL-6 levels are markedly higher in patients with poor functional outcomes.(76)
Gu et al, 2023AIS/7,053 (CNSR-III)Within 24 h of admissionThe association between acute-phase IL-6 and 90 day mRS is mainly driven by post-stroke inflammation leading to disability.(70)
Jia et al, 2024AIS/6931 (CNSR-III)Within 24 h of admissionHigh IL-6 levels are notably associated with poor 3-month outcomes in patients with AIS.(69)
Yi et al, 2024AISa/4031 (CNSR-III)Within 24 h of admissionElevated IL-6 is markedly associated with in-hospital neurological deterioration and improves risk prediction when added to traditional models.(75)
Dargazanli et al, 2025LVO with EVT/75In intracranial blood samples before thrombectomyElevated intracranial IL-6 levels are linked to worse functional outcomes.(71)
Băcilă et al, 2025AIS/56Days 1 and 7 after admissionIL-6 is an important biomarker for predicting AIS severity and functional. recovery(74)

a Patients who arrive at the hospital within 24 h after the onset of illness and have a National Institutes of Health Stroke Scale score ≤5. CNSR-III, China National Stroke Registry-III; AIS, acute ischemic stroke; GP, general population; EVT, endovascular treatment; NA, not available; PNR, patients not receiving reperfusion therapy; PER, patients with effective reperfusion; PWER, patients without effective reperfusion; END, early neurological deterioration; LVO, large vessel occlusion; mRS, modified Rankin Scale; hs-CRP, high-sensitivity C-reactive protein; WBC, white blood cells; ESR, erythrocyte sedimentation rate.

Therapeutic studies targeting the IL-6/IL-6R axis

Biologic agents targeting the IL-6/IL-6R axis are generally divided into three classes: Anti-IL-6R antibodies, anti-IL-6 antibodies and soluble gp130 (sgp130)-Fc fusion proteins. Representative IL-6R inhibitors include tocilizumab, sarilumab and satralizumab, while siltuximab, ziltivekimab and pacibekitug selectively target IL-6 itself (78).

Basic research

Animal studies have shown that targeting the IL-6/IL-6R axis is therapeutically beneficial. In MCAO mouse models, sgp130-Fc administration improved long-term survival outcomes by blocking IL-6 trans-signaling (79). Tocilizumab administered during the acute phase of IS was shown to reduce infarct volume, with female mice requiring higher doses compared with males due to elevated sIL-6R levels (80). Inhibition of IL-6 trans-signaling also slows or reverses atherosclerotic plaque formation (81). Table III (79–82) lists the relevant studies.

Table III.

Therapeutic studies targeting the IL-6/IL-6R axis.

Table III.

Therapeutic studies targeting the IL-6/IL-6R axis.

First author, yearIschemia modelStrainInterventionResults(Refs.)
Hall et al, 2025tMCAO (60 min, filament)C57BL/6sgp130Inhibiting IL-6 trans-signaling via sgp130 markedly improves long-term functional outcomes after stroke in mice, with dosage differences observed between males and females.(79)
Hudobenko et al, 2019tMCAO (60 min, filament)C57BL/6JTocilizumabBlocking IL-6R may offer a new therapeutic approach for stroke, requiring higher doses in females.(80)
Ministrini et al, 2025pMCAO (photo-chemically)C57BL/6JAnti-mouse IL-6 mAbDirect IL-6 inhibition slows arterial thrombosis by reducing collagen-induced platelet activation, especially under chronic low-grade inflammation.(82)
Schuett et al, 2012ApoEC57BL/6J Ldlr−/−sgp130-FcAs a selective inhibitor of IL-6 trans-signaling, sgp130-Fc markedly reduces the development and progression of atherosclerosis and can reverse established lesions.(81)

[i] IL-6R, IL-6 receptor; tMCAO, transient middle cerebral artery occlusion; pMCAO, permanent middle cerebral artery occlusion; ApoE, apolipoprotein E; mAb, monoclonal antibody; sgp130, soluble glycoprotein 130; Ldlr−/−, low-density lipoprotein receptor knockout.

Clinical studies

Phase II clinical evidence further supports the therapeutic potential of IL-6 pathway inhibition. The IL-6R Inhibition in Stroke (IRIS) trial demonstrated that in patients with anterior circulation large-vessel occlusion-induced AIS, EVT combined with tocilizumab within 24 h of onset markedly reduced infarct core volume and may have improved functional outcomes (83). The Assessing the Effect of Anti-IL-6 Treatment in Myocardial Infarction trial showed that a single dose of tocilizumab attenuated myocardial injury in patients with acute ST-segment elevation myocardial infarction without serious adverse events (84). In the RESCUE trial, treatment with the fully human monoclonal antibody ziltivekimab markedly reduced the levels of atherosclerosis-related inflammatory and thrombotic biomarkers in patients with moderate-to-severe chronic kidney disease, likely through inhibition of collagen-induced platelet activation (82,85). Furthermore, the TRANQUILITY trial demonstrated that quarterly administration of the IL-6 inhibitor pacibekitug effectively lowered high-sensitivity (hs)-CRP levels, prompting a phase III trial for ASCVD (86). The ongoing ZEUS trial, with an expected completion date during 2026, aims to determine whether ziltivekimab can reduce the incidence of cardiovascular events in the high-risk population that is patients with ASCVD (87,88). Table IV (83–86) lists the relevant studies.

Table IV.

Therapeutic clinical phase II and III randomized controlled trials targeting the IL-6/IL-6 receptor axis.

Table IV.

Therapeutic clinical phase II and III randomized controlled trials targeting the IL-6/IL-6 receptor axis.

First author, yearPhaseAnti-IL-6 mAbStudy populationStudy (ID)Results(Refs.)
Ridker et al, 2021IIZiltivekimabModerate-to-severe CKD with high CV riskRESCUE (NCT03926117)This drug can notably reduce multiple inflammatory and thrombotic biomarkers associated with atherosclerosis and displays good safety.(85)
Huse et al, 2022IITocilizumabAcute STEMI undergoing PCI within 6 hASSAIL-MI (NCT03004703)By reducing the number of circulating neutrophils and weakening their function, it may help improve myocardial salvage in patients with STEMI.(84)
Chu et al, 2024IITocilizumabAIS with anterior circulation LVOIRIS (NCT06238024)Combining tocilizumab with EVT can reduce infarct size in patients with anterior circulation LVO within 24 h of onset and may improve functional outcomes.(83)
Tourmaline Bio, 2025IIPacibekitugModerate-to-severe CKD with hs-CRP ≥2 mg/lTRANQUILITY (NCT06362759)Marked reductions in hs-CRP can be achieved through quarterly dosing.(86)
IIIZiltivekimabModerate-to-severe CKD with hs-CRP ≥2 mg/lZEUS (NCT05021835)Ongoing.N/A

[i] mAb, monoclonal antibody; ApoE, apolipoprotein E; CKD, chronic kidney disease; AIS, acute ischemic stroke; LVO, large vessel occlusion; CV, cardiovascular; STEMI, ST-segment elevation myocardial infarction; PCI, percutaneous coronary intervention; hs-CRP, high-sensitivity C-reactive protein; EVT, endovascular thrombectomy.

Conclusions and future perspectives

IL-6 exerts a prototypical dual effect in neuroinflammation, with its biological outcomes determined by the dynamic balance between the anti-inflammatory classical signaling pathway, mediated by mIL-6R/gp130 signaling and the pro-inflammatory trans-signaling pathway comprising sIL-6R/gp130 signaling. Notably, sgp130-Fc selectively binds the IL-6/-sIL-6R binary complex, resulting in blockage of trans-signaling without interfering with classical IL-6 signaling (89,90). This property makes sgp130-Fc a promising precision therapeutic target. IL-6 antagonists have been shown to protect BBB integrity, reduce cerebral edema and inflammatory injury and attenuate systemic inflammation by downregulating CRP expression and inhibiting the JAK/STAT3 pathway (91). Clinically, serum IL-6 levels rise sharply within the first week after AIS, whereas sgp130 levels decrease markedly, with sIL-6R levels showing no notable change (92). This imbalance increases the ratio of pro-inflammatory IL-6/sIL-6R binary (B) complexes to inactive IL-6/sIL-6R/sgp130-Fc ternary (T) complexes (B/T ratio). A biomarker study has shown that elevated B/T ratios are independently associated with higher future stroke risks in non-atrial fibrillation populations, suggesting the potential of this ratio as a biomarker for individualized anti-IL-6 therapy (93).

Numerous clinical trials have demonstrated that reducing IL-6 levels can slow the progression of atherosclerosis, prevent recurrent strokes and reduce the severity of ischemia-reperfusion injury following stroke recanalization (51,58,61,85,94). The CANTOS trial further demonstrated that IL-1β inhibition indirectly suppresses IL-6 signaling, reducing cardiovascular event incidence independent of lipid lowering (94). Similarly, colchicine, by inhibiting NLRP3 inflammasome activation and subsequently reducing IL-6 levels, markedly decreases IS incidence and the occurrence of major adverse cardiovascular events in secondary prevention without increasing bleeding risk (95,96). Reperfusion therapies themselves can exacerbate inflammation. Tissue plasminogen activator, while restoring perfusion, may disrupt the BBB and increase hemorrhagic transformation risk through IL-6-driven inflammatory cascades (97). In mechanical thrombectomy (MT), postoperative systemic inflammation has been identified as a notable determinant of poor patient outcomes (98). Patients that develop systemic inflammatory response syndrome after MT exhibit higher mortality and worse neurological recovery compared with patients that do not (99). The IRIS trial demonstrated that adjunctive IL-6R blockade with tocilizumab during EVT markedly reduced infarct core size and improved neurological outcomes (83).

Genetic evidence also supports the notion that decreased IL-6 levels are associated with improved patient outcomes for a number of diseases: Attenuation of IL-6 signaling reduces the risk of poor IS outcomes among patients with rheumatoid arthritis (100). Genetic variability further influences IL-6 bioactivity. The 3′ untranslated region polymorphism rs2228145 on the C allele of IL-6R markedly increases sIL-6R levels, enhancing the buffering capacity of patients against IL-6-mediated inflammation (61). As serum sgp130 concentrations generally exceed sIL-6R levels, the latter becomes the rate-limiting component of this neutralization system (101). Carriers of the rs2228145-C allele exhibit protective effects against cardiovascular and autoimmune diseases akin to treatment with tocilizumab (102). Mendelian randomization studies confirm that genetically-reduced IL-6 signaling lowers atherosclerotic and IS risk, particularly in LAA and SAO subtypes, although this potentially occurs at the cost of increased infection susceptibility (61,103).

Due to the low serum concentration, short half-life and multifactorial regulation of IL-6, including factors such as circadian rhythm, glucose level, age and comorbidities (104,105), precise monitoring and individualized assessment of IL-6 levels are necessary. Future studies should integrate differences in model sex, genetic polymorphisms, dynamic B/T ratio monitoring and hs-CRP trajectories to develop adaptive dosing algorithms, facilitating the transition of IL-6-related indicators from biomarkers to therapeutic targets. The present review also had certain limitations regarding the previous studies discussed, including small cohort sizes across individual studies, inconsistent timing of IL-6 testing and assay variability.

In summary, accumulating mechanistic and clinical evidence positions the IL-6/IL-6R axis as a notable link between neuroinflammation, atherosclerosis and ischemic injury. Targeted IL-6/IL-6R inhibition holds promise not only for neuroprotection in AIS but also for secondary cardiovascular prevention (83–86). Future clinical translation should establish unified standards for IL-6 detection, and focus on identifying appropriate patient subgroups for testing, optimizing treatment timing and dosage and balancing the anti-inflammatory efficacy of IL-6/IL-6R inhibition with immune safety to realize the full therapeutic potential of IL-6-targeted interventions in stroke management.

Acknowledgements

Not applicable.

Funding

The present review was funded by the Dalian Medical Science Research Program (grant no. 2211046).

Availability of data and materials

Not applicable.

Authors' contributions

XW was responsible for the conceptualization of the research project and drafting of the initial manuscript. XZ contributed to study conceptualization and reviewed the manuscript. JL contributed to the conclusion and provided critical insights. PL contributed towards conceptualization of the research project, as well as reviewing and revising the manuscript. Data authentication is not applicable. All authors read and approved the final version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Wang H, Yu X, Guo J, Ma S, Liu Y, Hu Y, Li J, Song Y and Zou Z: Burden of cardiovascular disease among the Western Pacific region and its association with human resources for health, 1990–2021: A systematic analysis of the global burden of disease study 2021. Lancet Reg Health West Pac. 51:1011952024.PubMed/NCBI

2 

Ji C, Ge X, Zhang J and Tong H: The stroke burden in china and its long-term trends: Insights from the global burden of disease (GBD) study 1990–2021. Nutr Metab Cardiovasc Dis. 35:1038482025. View Article : Google Scholar : PubMed/NCBI

3 

Fu Y, Liu Q, Anrather J and Shi FD: Immune interventions in stroke. Nat Rev Neurol. 11:524–535. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Li M, Li Z, Yao Y, Jin WN, Wood K, Liu Q, Shi FD and Hao J: Astrocyte-derived interleukin-15 exacerbates ischemic brain injury via propagation of cellular immunity. Proc Natl Acad Sci USA. 114:E396–E405. 2017.PubMed/NCBI

5 

Zhang F, Yan C, Wei C, Yao Y, Ma X, Gong Z, Liu S, Zang D, Chen J, Shi FD and Hao J: Vinpocetine inhibits NF-κB-dependent inflammation in acute ischemic stroke patients. Transl Stroke Res. 9:174–184. 2018. View Article : Google Scholar : PubMed/NCBI

6 

Endres M, Moro MA, Nolte CH, Dames C, Buckwalter MS and Meisel A: Immune pathways in etiology, acute phase, and chronic sequelae of ischemic stroke. Circ Res. 130:1167–1186. 2022. View Article : Google Scholar : PubMed/NCBI

7 

Zietz A, Gorey S, Kelly PJ, Katan M and McCabe JJ: Targeting inflammation to reduce recurrent stroke. Int J Stroke. 19:379–387. 2024. View Article : Google Scholar : PubMed/NCBI

8 

Pawluk H, Woźniak A, Tafelska-Kaczmarek A, Kosinska A, Pawluk M, Sergot K, Grochowalska R and Kołodziejska R: The role of IL-6 in ischemic stroke. Biomolecules. 15:4702025. View Article : Google Scholar : PubMed/NCBI

9 

Cui P, McCullough LD and Hao J: Brain to periphery in acute ischemic stroke: Mechanisms and clinical significance. Front Neuroendocrinol. 63:1009322021. View Article : Google Scholar : PubMed/NCBI

10 

Stanzione R, Forte M, Cotugno M, Bianchi F, Marchitti S and Rubattu S: Role of DAMPs and of leukocytes infiltration in ischemic stroke: Insights from animal models and translation to the human disease. Cell Mol Neurobiol. 42:545–556. 2022. View Article : Google Scholar : PubMed/NCBI

11 

Kumari S, Dhapola R, Sharma P, Nagar P, Medhi B and HariKrishnaReddy D: The impact of cytokines in neuroinflammation-mediated stroke. Cytokine Growth Factor Rev. 78:105–119. 2024. View Article : Google Scholar : PubMed/NCBI

12 

Lai TW, Zhang S and Wang YT: Excitotoxicity and stroke: Identifying novel targets for neuroprotection. Prog Neurobiol. 115:157–188. 2014. View Article : Google Scholar : PubMed/NCBI

13 

Tang C, Wang C, Zhang Y, Xue L, Li Y, Ju C and Zhang C: Recognition, intervention, and monitoring of neutrophils in acute ischemic stroke. Nano Lett. 19:4470–4477. 2019. View Article : Google Scholar : PubMed/NCBI

14 

Dhanesha N, Patel RB, Doddapattar P, Ghatge M, Flora GD, Jain M, Thedens D, Olalde H, Kumskova M, Leira EC and Chauhan AK: PKM2 promotes neutrophil activation and cerebral thromboinflammation: Therapeutic implications for ischemic stroke. Blood. 139:1234–1245. 2022. View Article : Google Scholar : PubMed/NCBI

15 

Shi K, Tian DC, Li ZG, Ducruet AF, Lawton MT and Shi FD: Global brain inflammation in stroke. Lancet Neurol. 18:1058–1066. 2019. View Article : Google Scholar : PubMed/NCBI

16 

Iadecola C, Buckwalter MS and Anrather J: Immune responses to stroke: Mechanisms, modulation, and therapeutic potential. J Clin Invest. 130:2777–2788. 2020. View Article : Google Scholar : PubMed/NCBI

17 

Wang Q, Tang XN and Yenari MA: The inflammatory response in stroke. J Neuroimmunol. 184:53–68. 2007. View Article : Google Scholar : PubMed/NCBI

18 

Jayaraj RL, Azimullah S, Beiram R, Jalal FY and Rosenberg GA: Neuroinflammation: Friend and foe for ischemic stroke. J Neuroinflammation. 16:1422019. View Article : Google Scholar : PubMed/NCBI

19 

Lu D, Chen WQ, Wang YP, Duan WY, Guo L, Wang L, Liu L, Xu AD and Wang YJ: Scientific Statements on Brain Cytoprotection in Ischemic Stroke—A Scientific Statement from the Chinese Stroke Association. Chin J Stroke. 19:938–55. 2024.(In Chinese).

20 

Tanaka T and Kishimoto T: The biology and medical implications of interleukin-6. Cancer Immunol Res. 2:288–294. 2014. View Article : Google Scholar : PubMed/NCBI

21 

Taher MY, Davies DM and Maher J: The role of the interleukin (IL)-6/IL-6 receptor axis in cancer. Biochem Soc Trans. 46:1449–1462. 2018. View Article : Google Scholar : PubMed/NCBI

22 

Scheller J, Chalaris A, Schmidt-Arras D and Rose-John S: The pro- and anti-inflammatory properties of the cytokine interleukin-6. Biochim Biophys Acta. 1813:878–888. 2011. View Article : Google Scholar : PubMed/NCBI

23 

Montgomery A, Tam F, Gursche C, Cheneval C, Besler K, Enns W, Manku S, Rey K, Hanson PJ, Rose-John S, et al: Overlapping and distinct biological effects of IL-6 classic and trans-signaling in vascular endothelial cells. Am J Physiol Cell Physiol. 320:C554–C565. 2021. View Article : Google Scholar : PubMed/NCBI

24 

Mihara M, Hashizume M, Yoshida H, Suzuki M and Shiina M: IL-6/IL-6 receptor system and its role in physiological and pathological conditions. Clin Sci (Lond). 122:143–159. 2012. View Article : Google Scholar : PubMed/NCBI

25 

Heo TH, Wahler J and Suh N: Potential therapeutic implications of IL-6/IL-6R/gp130-targeting agents in breast cancer. Oncotarget. 7:15460–15473. 2016. View Article : Google Scholar : PubMed/NCBI

26 

März P, Cheng JG, Gadient RA, Patterson PH, Stoyan T, Otten U and Rose-John S: Sympathetic neurons can produce and respond to interleukin 6. Proc Natl Acad Sci USA. 95:3251–3256. 1998. View Article : Google Scholar : PubMed/NCBI

27 

Heink S, Yogev N, Garbers C, Herwerth M, Aly L, Gasperi C, Husterer V, Croxford AL, Möller-Hackbarth K, Bartsch HS, et al: Trans-presentation of IL-6 by dendritic cells is required for the priming of pathogenic T(H)17 cells. Nat Immunol. 18:74–85. 2017. View Article : Google Scholar : PubMed/NCBI

28 

Gu Y, He M, Zhou X, Liu J, Hou N, Bin T, Zhang Y, Li T and Chen J: Endogenous IL-6 of mesenchymal stem cell improves behavioral outcome of hypoxic-ischemic brain damage neonatal rats by supressing apoptosis in astrocyte. Sci Rep. 6:185872016. View Article : Google Scholar : PubMed/NCBI

29 

Zhu H, Hu S, Li Y, Sun Y, Xiong X, Hu X, Chen J and Qiu S: Interleukins and ischemic stroke. Front Immunol. 13:8284472022. View Article : Google Scholar : PubMed/NCBI

30 

Pacinella G, Ciaccio AM, Casuccio A, Daidone M, Pecoraro R, Di Bona D, Del Cuore A, Puleo MG, Di Raimondo D, Di Chiara T, et al: Genetic polymorphisms and cytokine levels in ischemic stroke: Associations with subtypes and prognosis. Expert Rev Clin Immunol. 21:961–976. 2025. View Article : Google Scholar : PubMed/NCBI

31 

Huang X, Zhang M, Wang J and Hu F: Association between interleukin-6 levels and stroke: A systematic review and meta-analysis. J Int Med Res. 52:30006052412746262024. View Article : Google Scholar : PubMed/NCBI

32 

Sanderson TH, Reynolds CA, Kumar R, Przyklenk K and Hüttemann M: Molecular mechanisms of ischemia-reperfusion injury in brain: Pivotal role of the mitochondrial membrane potential in reactive oxygen species generation. Mol Neurobiol. 47:9–23. 2013. View Article : Google Scholar : PubMed/NCBI

33 

Shirley R, Ord EN and Work LM: Oxidative stress and the use of antioxidants in stroke. Antioxidants (Basel). 3:472–501. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Dong X, Wu D, Zhang Y, Jia L, Pan X, Sun J and Pan LL: Cathelicidin modulates vascular smooth muscle cell phenotypic switching through ROS/IL-6 pathway. Antioxidants (Basel). 9:4912020. View Article : Google Scholar : PubMed/NCBI

35 

Yan M, Sun Z, Zhang S, Yang G, Jiang X, Wang G, Li R, Wang Q and Tian X: SOCS modulates JAK-STAT pathway as a novel target to mediate the occurrence of neuroinflammation: Molecular details and treatment options. Brain Res Bull. 213:1109882024. View Article : Google Scholar : PubMed/NCBI

36 

Zhang SR, Li CT, Yang Y and Zhang LH: Oxidative stress, inflammation and autophagy in cerebral ischemic injury: mechanisms and therapeutic strategies. Chin J Pharmacol Toxicol. 32:651–660. 2018.(In Chinese).

37 

Kang S and Kishimoto T: Interplay between interleukin-6 signaling and the vascular endothelium in cytokine storms. Exp Mol Med. 53:1116–1123. 2021. View Article : Google Scholar : PubMed/NCBI

38 

Henein MY, Vancheri S, Longo G and Vancheri F: The role of inflammation in cardiovascular disease. Int J Mol Sci. 23:129062022. View Article : Google Scholar : PubMed/NCBI

39 

Webb CE, Vautrinot J and Hers I: IL-6 as a mediator of platelet hyper-responsiveness. Cells. 14:7662025. View Article : Google Scholar : PubMed/NCBI

40 

Zhang W, Tian T, Gong SX, Huang WQ, Zhou QY, Wang AP and Tian Y: Microglia-associated neuroinflammation is a potential therapeutic target for ischemic stroke. Neural Regen Res. 16:6–11. 2021. View Article : Google Scholar : PubMed/NCBI

41 

Purroy F, Farré-Rodriguez J, Mauri-Capdevila G, Vicente-Pascual M and Farré J: Basal IL-6 and S100b levels are associated with infarct volume. Acta Neurol Scand. 144:517–523. 2021. View Article : Google Scholar : PubMed/NCBI

42 

Ohta M, Kihara T, Toriuchi K, Aoki H, Iwaki S, Kakita H, Yamada Y and Aoyama M: IL-6 promotes cell adhesion in human endothelial cells via microRNA-126-3p suppression. Exp Cell Res. 393:1120942020. View Article : Google Scholar : PubMed/NCBI

43 

Mo J, Chen Z, Wang M, Cheng A, Li J, Pan Y, Jiang Y, Jing J, Wang Y, Pu Y and Li Z: Association between interleukin-6 and multiple acute infarctions in symptomatic intracranial atherosclerotic disease. Curr Neurovasc Res. 21:292–299. 2024. View Article : Google Scholar : PubMed/NCBI

44 

Yang X, Liao KH, Deng IB and Zhang LC: Knockdown of interleukin-6 plays a neuroprotective role against hypoxia-ischemia in neonatal rats via inhibition of caspase 3 and Bcl-2-associated X protein signaling pathway. Ibrain. 8:413–428. 2022. View Article : Google Scholar : PubMed/NCBI

45 

Zhan L, Mu Z, Jiang H, Zhang S, Pang Y, Jin H, Chen J, Jia C and Guo H: MiR-21-5p protects against ischemic stroke by targeting IL-6R. Ann Transl Med. 11:1012023. View Article : Google Scholar : PubMed/NCBI

46 

Radak D, Resanovic I and Isenovic ER: Changes in hypothalamus-pituitary-adrenal axis following transient ischemic attack. Angiology. 65:723–732. 2014. View Article : Google Scholar : PubMed/NCBI

47 

Crofford LJ, Kalogeras KT, Mastorakos G, Magiakou MA, Wells J, Kanik KS, Gold PW, Chrousos GP and Wilder RL: Circadian relationships between interleukin (IL)-6 and hypothalamic-pituitary-adrenal axis hormones: Failure of IL-6 to cause sustained hypercortisolism in patients with early untreated rheumatoid arthritis. J Clin Endocrinol Metab. 82:1279–1283. 1997. View Article : Google Scholar : PubMed/NCBI

48 

Hassamal S: Chronic stress, neuroinflammation, and depression: An overview of pathophysiological mechanisms and emerging anti-inflammatories. Front Psychiatry. 14:11309892023. View Article : Google Scholar : PubMed/NCBI

49 

Roy M, Collier B and Roy A: Hypothalamic-pituitary-adrenal axis dysregulation among diabetic outpatients. Psychiatry Res. 31:31–37. 1990. View Article : Google Scholar : PubMed/NCBI

50 

Kim S, Park ES, Chen PR and Kim E: Dysregulated hypothalamic-pituitary-adrenal axis is associated with increased inflammation and worse outcomes after ischemic stroke in diabetic mice. Front Immunol. 13:8648582022. View Article : Google Scholar : PubMed/NCBI

51 

Kamtchum-Tatuene J, Saba L, Heldner MR, Poorthuis MHF, de Borst GJ, Rundek T, Kakkos SK, Chaturvedi S, Topakian R, Polak JF, et al: Interleukin-6 predicts carotid plaque severity, vulnerability, and progression. Circ Res. 131:e22–e33. 2022. View Article : Google Scholar : PubMed/NCBI

52 

Mehta NN, deGoma E and Shapiro MD: IL-6 and cardiovascular risk: A narrative review. Curr Atheroscler Rep. 27:122024. View Article : Google Scholar : PubMed/NCBI

53 

Lundin JI, Peters U, Hu Y, Ammous F, Benjamin EJ, Bis JC, Brody JA, Cushman M, Fuller H, Gignoux C, et al: Epigenetic mechanisms underlying variation of IL-6, a well-established inflammation biomarker and risk factor for cardiovascular disease. Atherosclerosis. 407:1202192025. View Article : Google Scholar : PubMed/NCBI

54 

Kitagawa K, Toi S, Yoshizawa H, Sato Y and Todo K: Association between serum levels of interleukin-6 and stroke, cardiovascular events, and Alzheimer's disease dementia. J Atheroscler Thromb. 32:1390–1399. 2025. View Article : Google Scholar : PubMed/NCBI

55 

Jenny NS, Callas PW, Judd SE, McClure LA, Kissela B, Zakai NA and Cushman M: Inflammatory cytokines and ischemic stroke risk: The REGARDS cohort. Neurology. 92:e2375–e2384. 2019. View Article : Google Scholar : PubMed/NCBI

56 

Papadopoulos A, Palaiopanos K, Björkbacka H, Peters A, de Lemos JA, Seshadri S, Dichgans M and Georgakis MK: Circulating interleukin-6 levels and incident ischemic stroke: A systematic review and meta-analysis of prospective studies. Neurology. 98:e1002–e1012. 2022. View Article : Google Scholar : PubMed/NCBI

57 

Jiang Y and Fan T: IL-6 and stroke recurrence in ischemic stroke. Biomark Med. 18:739–747. 2024. View Article : Google Scholar : PubMed/NCBI

58 

Xu J, Mo J, Zhang X, Chen Z, Pan Y, Yan H, Meng X and Wang Y: Nontraditional risk factors for residual recurrence risk in patients with ischemic stroke of different etiologies. Cerebrovasc Dis. 51:630–638. 2022. View Article : Google Scholar : PubMed/NCBI

59 

Li J, Lin J, Pan Y, Wang M, Meng X, Li H, Wang Y, Zhao X, Qin H, Liu L, et al: Interleukin-6 and YKL-40 predicted recurrent stroke after ischemic stroke or TIA: analysis of 6 inflammation biomarkers in a prospective cohort study. J Neuroinflammation. 19:1312022. View Article : Google Scholar : PubMed/NCBI

60 

McCabe JJ, Harris K, Walsh C, Gorey S, Arnold M, De Marchis GM, Hervella P, Iglesias-Rey R, Jern C, Katan M, et al: Interleukin-6, C-reactive protein, and vascular recurrence after stroke with and without atherosclerosis. Stroke. 56:2588–2596. 2025. View Article : Google Scholar : PubMed/NCBI

61 

Georgakis MK, Malik R, Gill D, Franceschini N, Sudlow CLM and Dichgans M; INVENT Consortium, CHARGE Inflammation Working Group, : Interleukin-6 signaling effects on ischemic stroke and other cardiovascular outcomes: A mendelian randomization study. Circ Genom Precis Med. 13:e0028722020. View Article : Google Scholar : PubMed/NCBI

62 

Zhang L, Omarov M, Xu L, deGoma E, Natarajan P and Georgakis MK: IL6 genetic perturbation mimicking IL-6 inhibition is associated with lower cardiometabolic risk. Nat Cardiovasc Res. 4:1172–1186. 2025. View Article : Google Scholar : PubMed/NCBI

63 

Cui JJ, Zhang CZ, Zhu B, Xu ZP and Liu YY: Serum miR-21-5p, IL-6, and IL-8 Expression Levels and Predictive Model Construction in Patients with Cerebral Infarction. Chin J Gerontology. 43:4103–4106. 2023.(In Chinese).

64 

Kowalski RG, Ledreux A, Violette JE, Paustian W, Sillau S, Thompson JA, Neumann RT, Ornelas D, Monte AA, Dylla L, et al: Circulating interleukin-6 levels and timing of acute ischemic stroke onset. Ann Clin Transl Neurol. 12:1926–1931. 2025. View Article : Google Scholar : PubMed/NCBI

65 

Su JH, Luo MY, Liang N, Gong SX, Chen W, Huang WQ, Tian Y and Wang AP: Interleukin-6: A novel target for cardio-cerebrovascular diseases. Front Pharmacol. 12:7450612021. View Article : Google Scholar : PubMed/NCBI

66 

Mosarrezaii A, Amiri-Nikpour MR, Mehryar HR, Choobi Anzali B, Nourooz-Zadeh S, Babaei S and Farrokhi H: Investigating the relationship between interleukin-6 serum levels and outcome in acute ischemic CVA. Brain Behav. 10:e016682020. View Article : Google Scholar : PubMed/NCBI

67 

Alfieri DF, Lehmann MF, Flauzino T, de Araújo MCM, Pivoto N, Tirolla RM, Simão ANC, Maes M and Reiche EMV: Immune-inflammatory, metabolic, oxidative, and nitrosative stress biomarkers predict acute ischemic stroke and short-term outcome. Neurotox Res. 38:330–343. 2020. View Article : Google Scholar : PubMed/NCBI

68 

Li X, Lin S, Chen X, Huang W, Li Q, Zhang H, Chen X, Yang S, Jin K and Shao B: The prognostic value of serum cytokines in patients with acute ischemic stroke. Aging Dis. 10:544–556. 2019. View Article : Google Scholar : PubMed/NCBI

69 

Jia WL, Jiang YY, Jiang Y, Meng X, Li H, Zhao XQ, Wang YL, Wang YJ, Gu HQ and Li ZX: Associations between admission levels of multiple biomarkers and subsequent worse outcomes in acute ischemic stroke patients. J Cereb Blood Flow Metab. 44:742–756. 2024. View Article : Google Scholar : PubMed/NCBI

70 

Gu HQ, Yang KX, Li JJ, Lin JX, Jing J, Xiong YY, Zhao XQ, Wang YL, Liu LP, Meng X, et al: Mediation effect of stroke recurrence in the association between post-stroke interleukin-6 and functional disability. CNS Neurosci Ther. 29:3579–3587. 2023. View Article : Google Scholar : PubMed/NCBI

71 

Dargazanli C, Blaquière M, Moynier M, de Bock F, Labreuche J, Ter Schiphorst A, Derraz I, Radu RA, Gascou G, Lefevre PH, et al: Inflammation biomarkers in the intracranial blood are associated with outcome in patients with ischemic stroke. J Neurointerv Surg. 17:159–166. 2025. View Article : Google Scholar : PubMed/NCBI

72 

Pawluk H, Kołodziejska R, Grześk G, Kozakiewicz M, Woźniak A, Pawluk M, Kosinska A, Grześk M, Wojtasik J and Kozera G: Selected mediators of inflammation in patients with acute ischemic stroke. Int J Mol Sci. 23:106142022. View Article : Google Scholar : PubMed/NCBI

73 

Hervella P, Rodríguez-Castro E, Rodríguez-Yáñez M, Arias S, Santamaría-Cadavid M, López-Dequidt I, Estany-Gestal A, Maqueda E, López-Loureiro I, Sobrino T, et al: Intra- and extra-hospital improvement in ischemic stroke patients: Influence of reperfusion therapy and molecular mechanisms. Sci Rep. 10:35132020. View Article : Google Scholar : PubMed/NCBI

74 

Băcilă CI, Vlădoiu MG, Văleanu M, Moga DF and Pumnea PM: The role of IL-6 and TNF-alpha biomarkers in predicting disability outcomes in acute ischemic stroke patients. Life Basel). 15:472025. View Article : Google Scholar : PubMed/NCBI

75 

Yi L, Li ZX, Jiang YY, Jiang Y, Meng X, Li H, Zhao XQ, Wang YL, Liu LP, Wang YJ and Gu HQ: Inflammatory marker profiles and in-hospital neurological deterioration in patients with acute minor ischemic stroke. CNS Neurosci Ther. 30:e146482024. View Article : Google Scholar : PubMed/NCBI

76 

Sabir Rashid A, Huang-Link Y, Johnsson M, Wetterhäll S and Gauffin H: Predictors of early neurological deterioration and functional outcome in acute ischemic stroke: The importance of large artery disease, hyperglycemia and inflammatory blood biomarkers. Neuropsychiatr Dis Treat. 18:1993–2002. 2022. View Article : Google Scholar : PubMed/NCBI

77 

Deng QW, Huang S, Li S, Zhai Q, Zhang Q, Wang ZJ, Chen WX, Sun H, Lu M and Zhou J: Inflammatory factors as potential markers of early neurological deterioration in acute ischemic stroke patients receiving endovascular therapy-the AISRNA study. J Inflamm Res. 14:4399–4407. 2021. View Article : Google Scholar : PubMed/NCBI

78 

Grebenciucova E and VanHaerents S: Interleukin 6: At the interface of human health and disease. Front Immunol. 14:12555332023. View Article : Google Scholar : PubMed/NCBI

79 

Hall C, Nguyen DT, Mendoza K, Tan C and Chauhan A: Inhibition of IL-6 trans-signaling promotes post-stroke functional recovery in a sex and dose-dependent manner. J Neuroinflammation. 22:522025. View Article : Google Scholar : PubMed/NCBI

80 

Hudobenko J, Chauhan A and McCullough L: Abstract 128: Amelioration of ischemic stroke damage through inhibition of interleukin-6 signaling with tocilizumab requires sex specific dosing. Stroke. 50 (Suppl 1):A1282019. View Article : Google Scholar

81 

Schuett H, Oestreich R, Waetzig GH, Annema W, Luchtefeld M, Hillmer A, Bavendiek U, von Felden J, Divchev D, Kempf T, et al: Transsignaling of interleukin-6 crucially contributes to atherosclerosis in mice. Arterioscler Thromb Vasc Biol. 32:281–290. 2012. View Article : Google Scholar : PubMed/NCBI

82 

Ministrini S, Liberale L, Puspitasari YM, Han J, Kirmes K, Unkelbach LP, Tirandi A, Niederberger R, Bengs S, Beer JH, et al: Direct interleukin-6 inhibition blunts arterial thrombosis by reducing collagen-mediated platelet activation. Arterioscler Thromb Vasc Biol. 45:1432–1439. 2025. View Article : Google Scholar : PubMed/NCBI

83 

Chu X, Ma Z, Liu Y, Sun J, Wang N, Li C, Feng X, Li J, Wang B, Zhou C, et al: IRIS, a randomised, double-blind, placebo-controlled trial of interleukin-6 receptor inhibition undergoing endovascular treatment in acute anterior circulation ischaemic stroke: Study rationale and design. Stroke Vasc Neurol. 10:514–519. 2025. View Article : Google Scholar : PubMed/NCBI

84 

Huse C, Anstensrud AK, Michelsen AE, Ueland T, Broch K, Woxholt S, Yang K, Sharma K, Tøllefsen IM, Bendz B, et al: Interleukin-6 inhibition in ST-elevation myocardial infarction: Immune cell profile in the randomised ASSAIL-MI trial. EBioMedicine. 80:1040132022. View Article : Google Scholar : PubMed/NCBI

85 

Ridker PM, Devalaraja M, Baeres FMM, Engelmann MDM, Hovingh GK, Ivkovic M, Lo L, Kling D, Pergola P, Raj D, et al: IL-6 inhibition with ziltivekimab in patients at high atherosclerotic risk (RESCUE): A double-blind, randomised, placebo-controlled, phase 2 trial. Lancet. 397:2060–2069. 2021. View Article : Google Scholar : PubMed/NCBI

86 

GlobeNewswire, . Tourmaline Bio Announces Positive Topline Results from the Ongoing Phase 2 TRANQUILITY Trial Evaluating Pacibekitug in Patients with Elevated High-Sensitivity C-reactive Protein and Chronic Kidney Disease. Tourmaline Inc.; https://www.globenewswire.com/news-release/2025/05/20/3084742/0/en/Tourmaline-Bio-Announces-Positive-Topline-Results-from-the-Ongoing-Phase-2-TRANQUILITY-Trial-Evaluating-Pacibekitug-in-Patients-with-Elevated-High-Sensitivity-C-reactive-Protein-an.htmlAugust 7–2025

87 

Ridker PM: From RESCUE to ZEUS: will interleukin-6 inhibition with ziltivekimab prove effective for cardiovascular event reduction? Cardiovasc Res. 117:e138–e140. 2021. View Article : Google Scholar : PubMed/NCBI

88 

Ridker PM and Rane M: Interleukin-6 signaling and anti-interleukin-6 therapeutics in cardiovascular disease. Circ Res. 128:1728–1746. 2021. View Article : Google Scholar : PubMed/NCBI

89 

Rose-John S, Jenkins BJ, Garbers C, Moll JM and Scheller J: Targeting IL-6 trans-signalling: Past, present and future prospects. Nat Rev Immunol. 23:666–681. 2023. View Article : Google Scholar : PubMed/NCBI

90 

Jostock T, Müllberg J, Ozbek S, Atreya R, Blinn G, Voltz N, Fischer M, Neurath MF and Rose-John S: Soluble gp130 is the natural inhibitor of soluble interleukin-6 receptor transsignaling responses. Eur J Biochem. 268:160–167. 2001. View Article : Google Scholar : PubMed/NCBI

91 

Weiss A and Ding Y: Beyond reperfusion: Adjunctive therapies targeting inflammation, edema, and blood-brain barrier dysfunction in ischemic stroke. Cerebrovasc Dis. 1–10. 2025.(Epub ahead of print). View Article : Google Scholar : PubMed/NCBI

92 

Acalovschi D, Wiest T, Hartmann M, Farahmi M, Mansmann U, Auffarth GU, Grau AJ, Green FR, Grond-Ginsbach C and Schwaninger M: Multiple levels of regulation of the interleukin-6 system in stroke. Stroke. 34:1864–1869. 2003. View Article : Google Scholar : PubMed/NCBI

93 

Ziegler L, Wallén H, Aspberg S, de Faire U and Gigante B: IL6 trans-signaling associates with ischemic stroke but not with atrial fibrillation. BMC Neurol. 21:3062021. View Article : Google Scholar : PubMed/NCBI

94 

Ridker PM, Libby P, MacFadyen JG, Thuren T, Ballantyne C, Fonseca F, Koenig W, Shimokawa H, Everett BM and Glynn RJ: Modulation of the interleukin-6 signalling pathway and incidence rates of atherosclerotic events and all-cause mortality: Analyses from the canakinumab anti-inflammatory thrombosis outcomes study (CANTOS). Eur Heart J. 39:3499–3507. 2018. View Article : Google Scholar : PubMed/NCBI

95 

Zhu S, Pan W, Yao Y and Shi K: The efficacy of colchicine compared to placebo for preventing ischemic stroke among individuals with established atherosclerotic cardiovascular diseases: A systematic review and meta-analysis. Scand Cardiovasc J. 59:24411122025. View Article : Google Scholar : PubMed/NCBI

96 

Fiolet ATL, Poorthuis MHF, Opstal TSJ, Amarenco P, Boczar KE, Buysschaert I, Budgeon C, Chan NC, Cornel JH, Jolly SS, et al: Colchicine for secondary prevention of ischaemic stroke and atherosclerotic events: A meta-analysis of randomised trials. EClinicalMedicine. 76:1028352024. View Article : Google Scholar : PubMed/NCBI

97 

Shi K, Zou M, Jia DM, Shi S, Yang X, Liu Q, Dong JF, Sheth KN, Wang X and Shi FD: tPA mobilizes immune cells that exacerbate hemorrhagic transformation in stroke. Circ Res. 128:62–75. 2021. View Article : Google Scholar : PubMed/NCBI

98 

Wang H, Tian X, Liao Z, Yue X, Sun L, Li X, Zou M and Ding J: Inflammatory biomarkers may be associated with poor outcomes after mechanical thrombectomy. Thromb J. 22:582024. View Article : Google Scholar : PubMed/NCBI

99 

Jafari M, Katlowitz K, De la Garza C, Sellers A, Moore S, Hall H, Desai A, Singh V and Damani R: Impact of systemic inflammatory response syndrome on acute ischemic stroke patients treated with mechanical thrombectomy. J Neurol Sci. 430:1199882021. View Article : Google Scholar : PubMed/NCBI

100 

Jia Y, Zhang K, Shi M, Guo D, Yang P, Bu X, Chen J, Wang A, Xu T, He J, et al: Associations of rheumatoid factor, rheumatoid arthritis, and interleukin-6 inhibitor with the prognosis of ischemic stroke: A prospective multicenter cohort study and mendelian randomization analysis. Transl Stroke Res. 15:750–760. 2024. View Article : Google Scholar : PubMed/NCBI

101 

Scheller J and Rose-John S: The interleukin 6 pathway and atherosclerosis. Lancet. 380:3382012. View Article : Google Scholar : PubMed/NCBI

102 

Zhang M, Bai Y, Wang Y, Cui H, Tang M, Wang L, Wang X and Gu D: Cumulative evidence for associations between genetic variants in interleukin 6 receptor gene and human diseases and phenotypes. Front Immunol. 13:8607032022. View Article : Google Scholar : PubMed/NCBI

103 

Georgakis MK, Malik R, Li X, Gill D, Levin MG, Vy HMT, Judy R, Ritchie M, Verma SS; Regeneron Genetics Center, ; et al: Genetically downregulated interleukin-6 signaling is associated with a favorable cardiometabolic profile: A phenome-wide association study. Circulation. 143:1177–1780. 2021. View Article : Google Scholar : PubMed/NCBI

104 

Said EA, Al-Reesi I, Al-Shizawi N, Jaju S, Al-Balushi MS, Koh CY, Al-Jabri AA and Jeyaseelan L: Defining IL-6 levels in healthy individuals: A meta-analysis. J Med Virol. 93:3915–3924. 2021. View Article : Google Scholar : PubMed/NCBI

105 

Docherty S, Harley R, McAuley JJ, Crowe LAN, Pedret C, Kirwan PD, Siebert S and Millar NL: The effect of exercise on cytokines: Implications for musculoskeletal health: A narrative review. BMC Sports Sci Med Rehabil. 14:52022. View Article : Google Scholar : PubMed/NCBI

Related Articles

  • Abstract
  • View
  • Download
  • Twitter
Copy and paste a formatted citation
Spandidos Publications style
Wang X, Zhang X, Lin J and Lin P: Interleukin‑6 and ischemic stroke: From mechanisms to clinical prospects (Review). Mol Med Rep 33: 80, 2026.
APA
Wang, X., Zhang, X., Lin, J., & Lin, P. (2026). Interleukin‑6 and ischemic stroke: From mechanisms to clinical prospects (Review). Molecular Medicine Reports, 33, 80. https://doi.org/10.3892/mmr.2026.13790
MLA
Wang, X., Zhang, X., Lin, J., Lin, P."Interleukin‑6 and ischemic stroke: From mechanisms to clinical prospects (Review)". Molecular Medicine Reports 33.3 (2026): 80.
Chicago
Wang, X., Zhang, X., Lin, J., Lin, P."Interleukin‑6 and ischemic stroke: From mechanisms to clinical prospects (Review)". Molecular Medicine Reports 33, no. 3 (2026): 80. https://doi.org/10.3892/mmr.2026.13790
Copy and paste a formatted citation
x
Spandidos Publications style
Wang X, Zhang X, Lin J and Lin P: Interleukin‑6 and ischemic stroke: From mechanisms to clinical prospects (Review). Mol Med Rep 33: 80, 2026.
APA
Wang, X., Zhang, X., Lin, J., & Lin, P. (2026). Interleukin‑6 and ischemic stroke: From mechanisms to clinical prospects (Review). Molecular Medicine Reports, 33, 80. https://doi.org/10.3892/mmr.2026.13790
MLA
Wang, X., Zhang, X., Lin, J., Lin, P."Interleukin‑6 and ischemic stroke: From mechanisms to clinical prospects (Review)". Molecular Medicine Reports 33.3 (2026): 80.
Chicago
Wang, X., Zhang, X., Lin, J., Lin, P."Interleukin‑6 and ischemic stroke: From mechanisms to clinical prospects (Review)". Molecular Medicine Reports 33, no. 3 (2026): 80. https://doi.org/10.3892/mmr.2026.13790
Follow us
  • Twitter
  • LinkedIn
  • Facebook
About
  • Spandidos Publications
  • Careers
  • Cookie Policy
  • Privacy Policy
How can we help?
  • Help
  • Live Chat
  • Contact
  • Email to our Support Team