Open Access

Matrix metalloproteinase‑9 in hemorrhagic transformation after acute ischemic stroke (Review)

  • Authors:
    • Pingping Guo
    • Hongmin Li
    • Xiangyu Zhang
    • Yang Liu
    • Sara Xue
    • Voon Wee Yong
    • Mengzhou Xue
  • View Affiliations

  • Published online on: June 5, 2025     https://doi.org/10.3892/mmr.2025.13590
  • Article Number: 225
  • Copyright: © Guo et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Hemorrhagic transformation (HT) is a devasting complication following acute ischemic stroke with high morbidity and mortality. The pathogenesis of HT mainly involves ischemia‑reperfusion‑induced oxidative stress, neuroinflammation, thrombolytic therapy‑associated toxicity and, most critically, blood‑brain barrier (BBB) disruption. Matrix metalloproteinase‑9 (MMP‑9) serves as a critical mediator of HT through degrading extracellular matrix components and disrupting tight junction proteins, thereby compromising BBB integrity. Thus, elaborating the underlying molecular mechanisms of MMP‑9 in destroying BBB and promoting HT is essential to improve the outcome of ischemic stroke patients. Furthermore, to provide beneficial insights for the treatment of ischemic stroke, precise understanding of the potential role of MMP‑9 as a biomarker and treatment target to predict and ameliorate the risk of HT is also necessary.

Introduction

Stroke is the second leading cause of death and the third leading cause of disability worldwide and a great burden to patients and society (1). Ischemic stroke, the most common type of stroke, occurs when cerebral artery occlusion leads to critical reduction in blood flow. Early reperfusion remains paramount in acute ischemic stroke (AIS) management. Intravenous thrombolysis with tissue plasminogen activator (tPA) is one of the most effective reperfusion therapeutic strategies and is able to rapidly restore cerebral perfusion and alleviate the neurological deficits of patients (2). Despite its clinical efficacy, tPA thrombolysis remains underused, with only 5–10% of ischemic stroke patients receiving treatment (3). This limited application primarily stems from two key constraints: the narrow 4.5-h therapeutic window and the risk of tPA-induced hemorrhagic transformation (HT) (4). Therefore, early identification of the risk of HT and timely prevention strategies might be helpful for physicians to make more useful decisions on the treatment for AIS.

The pathophysiological mechanisms of HT are complicated, involving blood-brain barrier (BBB) disruption, ischemia-reperfusion injury, oxidative stress, neuroinflammation, tPA administration and brain cell death (3). These processes interact with each other to promote the disruption of vascular integrity and BBB dysfunction (5). BBB is a highly selective functional structure that acts as a protective interface between the central nervous system (CNS) and the circulatory system, playing a critical role in maintaining the microenvironmental homeostasis of CNS (6). BBB disruption is a prominent pathophysiological feature of ischemic stroke, accompanied by worse functional prognosis and higher mortality rate (7). The components of erythrocytes, neurotoxic plasma components and pathogens enter the brain tissue through the compromised BBB, markedly contributing to the pathogenesis of HT (8).

Matrix metalloproteinases (MMPs) play a key role in the pathological process of BBB damage by mediating the degradation of extracellular matrix (ECM) and tight junction proteins (TJPs) (6,9). MMPs comprise a family of calcium-dependent zinc-containing endopeptidases and play pivotal roles in tissue remodeling and ECM degradation, including gelatin, elastins, collagens, matrix glycoproteins and proteoglycans (10). These enzymes are synthesized by neurons, astrocytes, endothelial cells, microglia and peripheral immune cells (11). Structurally, MMPs exhibit a highly conserved domain architecture consisting of four principal components: The amino-terminal signal peptide domain, the propeptide region (predomain), the catalytic domain containing zinc and variable C-terminal domains that mediate substrate recognition and interaction, including the transmembrane domain, the hemopexin-like domain and the fibronectin domain (12,13). At present, at least 26 human MMPs have been discovered, which are divided into six subgroups according to their different structures: collagenases, gelatinases, stromelysins, matrilysins, membrane-type MMPs and others (10,14). Previous studies have demonstrated that MMPs high expression is associated with BBB disruption and HT following AIS (1517). Among all MMP family members, MMP-9 has emerges as the most extensively investigated protease, with numerous animal and clinical studies establishing its crucial role in HT pathogenesis (11,1822).

The present review aimed to elaborate the mechanistic involvement of MMP-9 in HT development following AIS and evaluate its potential as biomarker and therapeutic target.

Physiopathological involvement of MMP-9 in ischemic stroke

Belonging to gelatinase B family, MMP-9 comprises a signal peptide domain, a prodomain, a catalytic domain with a Zn2+ binding site, a fibronectin domain and a hemopexin-like domain (Fig. 1) (23). The signal peptide domain mediates MMP-9 excretion from the cell (24). The predomain maintains MMP-9 in an inactive state through binding its conserved cysteine residue to Zn2+ in the enzyme's active center (25). The catalytic domain is responsible for the proteolytic activity of MMP-9 (23). The fibronectin domain acts as a modulator of substrates recognition, which is able to bind to gelatins, laminins and collagens (26). The hemopexin-like domain plays a crucial role in substrates specificity and the specific substrates mainly comprise ECM components (such as collagens, gelatin and fibronectin) and non-ECM molecules including tissue inhibitor metalloproteinases and chemokines (23,26). In the brain, MMP-9 is mainly produced by astrocytes, microglia and infiltrating leukocytes (27,28). The expression of MMP-9 is regulated by transcription factors, inflammatory factors, chemokines and growth factors (29). MMP-9 is usually secreted in an inactive form and activated by thrombin, MMP-3, tPA and reactive oxygen/nitrogen species (ROS/RNS) (2932). Notably, zinc plays a dual role in MMP-9 regulation: As a structural cofactor required for its proteolytic function at physiological levels and as a pro-oxidant stimulus that indirectly activates MMP-9 via ROS generation when accumulated in excess (33).

Following ischemic stroke, MMP-9 undergoes a dynamic change, with its expression and activity varying markedly over time (Fig. 2). The significant upregulation of MMP-9 can be found in the acute phase of ischemic stroke (within 7 days from onset). It has been demonstrated that the expression and activity of MMP-9 increases rapidly and peaks within 24 h after ischemic stroke (34,35). During this time, MMP-9 exacerbates brain injury by damaging BBB integrity, aggravating edema, enhancing neuroinflammation and brain cell death and promoting HT (36). At 7–14 days after ischemic stroke, the activity and expression of MMP-9 still remains elevated. In this delayed phase following ischemic stroke, MMP-9 facilitates the tissue repair and angiogenesis by degrading damaged ECM, thereby mitigating ischemic brain injury and prompting functional recovery (37). From weeks to months after ischemic stroke, the expression and activity of MMP-9 gradually decreases. This phase is characterized by ECM remodeling, in which impenetrable scar tissues formed by gliosis hinders the regeneration and reprojection of axons. At this time, the BBB opening mediated by MMP-9 allows cells to enter the CNS, facilitating functional recovery (38).

MMP-9 mediated blood-brain barrier dysfunction after ischemic stroke

BBB is a unique structure formed by blood vessels in the CNS, maintaining neuronal activity and blocking pathogenic damage (39). The maintenance of BBB function primarily relies on the interaction of endothelial cells, astrocytes, pericytes and ECM around the vessels (Fig. 3). Endothelial cells express high levels of TJPs, which help to prevent blood entering brain by limiting para-cellular permeability (40). TJPs mainly include the transmembrane proteins (claudin-5, claudin-3, claudin-12 and occludin) and zonula occludens proteins (ZO-1, ZO-2 and ZO-3), in which transmembrane proteins, especially claudin-5, interact with the similar proteins in the adjacent cell to form tight junctions and zonula occludens proteins connect the aforementioned proteins to the cytoskeleton (41,42). The basement membrane (BM), surrounding the lumen surface of endothelial cells, is composed of collagen IV, fibronectin, heparin sulfate and laminins secreted by endothelial cells, astrocytes and pericytes (43,44). Pericytes and astrocytic endfeet are embedded in the BM, playing a significant role in maintaining BBB integrity (45). The brain ECM consists of neuro-ECM, BM and luminal ECM. The neuro-ECM serves as the connective framework of the brain, forming a dynamic scaffold that supports neurons and glia, while both the BM and the luminal ECM are involved in forming BBB (Fig. 3) (46). These integrated cells and structures collectively promote the integrity of BBB, thereby playing a critical role in regulating the brain homeostasis and protecting the CNS.

BBB dysfunction initiates from the onset of ischemic stroke and aggravates with the sustained hyperfusion. The disruption of TJPs and ECM is the main reason underlying the increased permeability of BBB after ischemic stroke, in which MMP-9 plays a critical role (47). MMP-9 primarily disrupts BBB through proteolytic degradation of key ECM components, including collagen IV, laminin and fibronectin (48,49). Beyond its role in ECM degradation, MMP-9 also targets TJPs, with evidence demonstrating its specific proteolytic effects on occludin, claudin-5 and ZO-1 (50). In addition, MMP-9 mediates BBB disruption by degrading dystroglycan, a critical ECM receptor on astrocyte endfeet that anchors them to the BM through the high-affinity interactions with laminin (51,52).

Involvement of MMP-9 in hemorrhagic transformation after ischemic stroke

HT is a common complication of ischemic stroke, with an incidence of 10–40%, leading to increased stroke mortality (53). The classification of HT following ischemic stroke, as defined by the European Cooperative Acute Stroke Study criteria, encompasses two main categories with distinct radiographic and prognostic implications: Hemorrhagic infarction (HI) and parenchymal hematoma (PH) (54). They are further subclassified into four clinically relevant subtypes based on computed tomography characteristics: HI-1, HI-2, PH-1 and PH-2 (Table I). The clinical prognosis of HT exhibits marked heterogeneity across different subtypes. Compared with ischemic stroke patients without HT, PH-2 markedly increases the risk of early neurological deterioration, 3-month mortality and disability, whereas HI-1, HI-2 and PH-1 show no statistically significant differences in these outcomes (55). HT can be also classified into symptomatic HT and asymptomatic HT on basis of the presence or absence of neurological decline (56). While the pathogenesis of HT following ischemic stroke is complicated, current evidence points to three interconnected mechanisms, including oxidative stress induced by ischemia-reperfusion, neuroinflammation and thrombolytic therapy-associated toxicity (53,57,58). MMP-9 functions as a pivotal effector molecule in these pathological processes (Fig. 4).

Table I.

ECASS classification of hemorrhagic transformation (54).

Table I.

ECASS classification of hemorrhagic transformation (54).

TypeRadiographic features
HI-1Small scattered petechial hemorrhage at the infarct margins
HI-2Numerous confluent petechial hemorrhages within the infarction, without signs of mass effect
PH-1Hematoma within the infarction, occupying <30%, with minor signs of mass effect
PH-2Hematoma within the infarction, occupying ≥30%, with obvious mass effect

[i] ECASS, the European Cooperation Acute Stroke Study; HI, hemorrhagic infarction; PH, parenchymal hematoma.

Oxidative stress is a crucial pathological process mediating ischemia-reperfusion injury and HT, in which excess production and activation of ROS/RNS play an important role (32,59). After stroke, ischemia and hypoxia of certain brain tissue induces the release of excitotoxic substance-glutamate, which subsequently activates the calcium channel-related receptors, especially N-methyl-D-aspartate receptor, leading to the influx of calcium (22,60). Calcium influx initiates a series of deleterious cascades, such as the mitochondrial depolarization and nicotinamide adenine dinucleotide phosphate oxidase activation, subsequently triggering an excessive generation and activation of ROS/RNS (61,62). Mounting evidence indicates that oxidative stress mediates BBB dysfunction through both direct and indirect mechanisms, with MMP-9 serving as a key downstream mediator in this pathological cascade. Direct effects involve disrupting TJPs (occludin, ZO-1 and claudin-5) through oxidative modification and redistribution and activating MMP-9 to catalyze the ECM degradation (63,64). Indirectly, ROS/RNS modulate BBB permeability by oxidatively activating multiple signaling pathways and transcriptional regulators. Primally, ROS/RNS-mediated activation of the high-mobility group box1 (HMGB1)/toll-like receptor 4 (TLR4) signaling axis induces nuclear factor-κB (NF-κB)-dependent transcriptional upregulation of MMP-9, which subsequently exacerbates BBB disruption (32). Furthermore, ROS activate phosphatidylinositol-3 kinase (PI3K)/protein kinase B (Akt) and mitogen-activated protein kinase (MAPK) signaling cascades, which promote nuclear translocation of transcription factors, such as NF-κB and activator protein-1 (AP-1), thereby enhancing MMP-9 expression through specific promoter binding (65,66).

Neuroinflammation following ischemic stroke is another widely recognized contributor to BBB breakdown and HT development. After ischemic stroke, dying brain cells release damage-associated molecular pattern (DAMP) molecules, including HMGB1, heat shock protein and DNA, thereby triggering microglia and astrocytes activation and pro-inflammatory cytokines, chemokines and MMPs production (6769). The pro-inflammatory cytokines such as IL-1β, IL-6 and TNF-α play a profound role in mediating BBB breakdown (70). IL-1β enhances chemokines production, promoting neutrophil infiltration and subsequent MMP-9 release (71,72). IL-6 directly compromises BBB integrity by reducing the trans-endothelial electrical resistance value in cerebral endothelial cells (73). TNF-α activates MAPK and PI3K/Akt signaling pathways, stimulating MMP-9-mediated degradation of ECM and TJPs (74). Meanwhile, upregulated chemokines, such as monocyte chemokine protein-1 (MCP-1), macrophage inflammatory protein-1α and stromal cell-derived factor 1, mediate BBB disruption by inducing the endothelial cells retraction and intercellular gap formation (75). These pro-inflammatory factors and proteases (especially MMP-9) collectively contribute to BBB dysfunction, promoting the infiltration of peripheral immune cells, mainly neutrophils, through the compromised BBB (70). The infiltrated and activated neutrophils release more pro-inflammatory cytokines and chemokines to enhance the activation of microglia and astrocytes. Additionally, the upregulation of neutrophil-derived MMP-9 further aggravates BBB dysfunction. These interconnected pathological cascades synergistically exacerbate BBB disruption and promote the progression of HT.

Thrombolytic therapy with tPA is a well-established risk factor for HT. As the only US Food and Drug Administration-approved agent for AIS, tPA can markedly improve functional outcomes by restoring cerebral blood flow when administered within 4.5 h of symptom onset (76). However, the fibrinolytic effect of tPA simultaneously increases the risk of hemorrhagic complications. Compelling clinical evidence indicates that intravenous tPA administration in AIS patients markedly elevates HT risk, particularly when treatment is delayed beyond the 4.5-h therapeutic window, compared with non-thrombolytic treatment approaches (77). Multiple mechanisms are involved in the development of tPA-mediated HT. First, tPA promotes the formation of HT through its pharmacological effect. Beyond its fibrinolytic activity in clot dissolution, tPA also activates some extracellular proteases and mediates the dysregulation of ECM proteolysis, leading to BBB leakage (78). Second, tPA can act on the platelet-derived growth factor receptor alpha (PDGFRα) on perivascular astrocyte end feet and promote the expression of PDGF-CC, leading to the upregulation of vascular permeability (79). Last but not least, tPA can also increase the risk of HT via multiple MMP-9-dependent pathways (8083): i) tPA mobilizes and activates neutrophils, via annexin A2-dependent MAPK signaling pathway, promoting neutrophil-derived MMP-9 secretion; ii) tPA binds the low-density lipoprotein receptor-related protein receptor on endothelial cells to enhance endothelial cell-derived MMP-9 expression. It is worth mentioning that although the role of tPA in promoting HT has been well established, it is not the sole contributing factor to HT during ischemia-reperfusion. The excessive ROS/RNS production during ischemia-reperfusion also plays a critical role in HT through activating MMP-9 and mediating BBB dysfunction (53). Of particular clinical relevance, the delayed administration of tPA (>4.5 h post-ischemia) potentiates this pathological process, thereby amplifying BBB disruption and HT risk during cerebral ischemia-reperfusion (84). This pathophysiological cascade elucidates the clinical observation of elevated HT risks when thrombolysis is administered beyond the 4.5-h therapeutic window.

MMP-9 as biomarker for hemorrhagic transformation

The role of MMP-9 in mediating BBB disruption highlights its potential as a predictive biomarker for HT. Previous reviews revealed that MMP-9 concentration in peripheral blood was positively associated with larger infarct size, worse prognosis and higher HT risk after AIS (85,86). There was a clinical study supporting the aforementioned viewpoints as well. With 168 patients with ischemic stroke and 40 healthy controls included in this study, Yuan et al (87) determined the plasma MMP-9 concentrations of these patients through enzyme-linked immunosorbent assay, finding that higher plasma MMP-9 concentration was markedly associated with increased spontaneous HT risk and MMP-9 value >181.7 ng/ml within 24 h after stroke onset was an independent marker to predict HT risk in ischemic stroke patients. However, Montaner et al (16) noted that the predictive value of plasma MMP-9 level differed in different HT subtypes. The authors found that baseline plasma MMP-9 levels positively corelated to HT severity following tPA administration: Lowest MMP-9 levels in HI-1 group (lower than non-HT group), higher but almost normal levels in HI-2 group, markedly higher in PH-1 group and highest in PH-2 group, which highlights the predictive value of baseline plasma MMP-9 in severe HT. This phenomenon may be attributable to the role of MMP-9 in mediating BBB dysfunction and promoting HT following ischemic stroke. Although substantial clinical evidence has established the specific predictive role of MMP-9 for HT following ischemic stroke, most clinical trials to date rely exclusively on peripheral blood MMP-9 measurements. This methodological limitation primarily reflects the significant ethical constraints and technical difficulties involved in accurately quantifying MMP-9 concentrations in the human brain. The results from an animal study clarified that MMP-9 activity within brain was positively associated with edema and infarct volume after ischemic stroke (88). Another study performed in mice and endothelial cells cultured in vitro reported that tPA-induced HT could be markedly attenuated when suppressing the expression of brain MMP-9 (89). These experimental findings from animal models and cell cultures provide indirect evidence supporting the predictive role of brain MMP-9 in HT. Additionally, some researchers suggested that an imbalance of MMP-9 and TIMP-1 contributed to the occurrence of HT (90). TIMP-1 is secreted together with MMP-9 and inhibits its proteolytic activity. The higher MMP-9/TIMP-1 ratio usually indicates the higher MMP-9 activity and HT risk. Notably, preclinical evidence from rat AIS models revealed the parallel MMP-9/TIMP-1 ratio changes in serum and brain tissue (91), indicating their comparable predictive capacity for HT.

As an important factor regulating MMP-9 expression, MMP-9 gene polymorphism represents a promising biomarker for predicting HT risk after ischemic stroke. In a case-control study involving 1,274 ischemic stroke patients and 1,258 healthy controls, researchers investigated four critical polymorphic sites of MMP-9 (rs17156, rs3787268, rs3918241 and rs3918242), finding that rs3918242 (−1562C/T) polymorphism was markedly associated with serum MMP-9 levels (92). In another clinical study including 222 ischemic stroke patients stratified by magnetic resonance imaging findings into HT and non-HT groups, researchers genotyped the −1562C/T polymorphism using PCR-restriction fragment length polymorphism analysis, revealing that 1562C/T polymorphism was markedly associated with HT risk, with the T allele potentially serving as a predictive biomarker for HT susceptibility (93). Contrary to previous findings, another study performed in the cerebrovascular disease patients found no correlation between −1562C/T polymorphism and spontaneous HT (94). Additionally, Fernández-Cadenas et al (95) genotyped 14 single nucleotide polymorphisms (SNPs) in MMP-9 gene, demonstrating no correlation between MMP-9 genetic variations and HT. These inconsistent results can be explained by several contributing factors. First, population-specific genetic differences play a crucial role in these observed variations. A comprehensive meta-analysis revealed the correlation between MMP-9 gene −1562C/T polymorphism and stroke risk among Asians, but not among Caucasians (96). Second, the pathophysiological mechanisms underlying HT encompass a complex cascade of molecular and cellular events, including BBB disruption, inflammatory responses, oxidative stress and MMPs activation, which collectively contribute to the development and progression of this condition. Therefore, genetic polymorphisms in MMP-9 may not markedly influence the occurrence of HT, as the development of HT is mediated through complex interactions among multiple genetic, molecular and environmental factors. Third, the interaction of different genetic variants in MMP-9 gene should not be ignored. Yi et al (97) demonstrated that the increased HT risk was attributable to the synergistic interaction between rs3918242 and rs3787268 genetic variants, rather than being caused by individual variant alone. Finally, variations in sample size and statistical methodologies may also contribute to the inconsistencies in research findings.

Reduction of hemorrhagic transformation by targeting MMP-9

Reducing HT following ischemic stroke may be beneficial to extend the therapeutic window of tPA, increase eligibility for thrombolytic therapy and improve the prognosis of patients. As a key mediator in HT pathogenesis, MMP-9 may be a prospective pharmacological target for HT. Although the predictive role of MMP-9 in severe HT following ischemic stroke has been well established, whether therapies targeting MMP-9 have varying efficacy based on HT subtypes remains unknown. To date, no selective MMP-9 inhibitors have passed clinical trials. The present review evaluated clinically available neuroprotective agents that demonstrate indirect regulatory effects on MMP-9 activity or expression, aiming to elucidate their molecular pathways of MMP-9 regulation and provide evidence-based recommendations for optimizing therapeutic strategies to mitigate post-ischemic HT risk (Table II).

Table II.

List of agents targeting MMP-9 and reducing HT after stroke.

Table II.

List of agents targeting MMP-9 and reducing HT after stroke.

Authors, yearClassAgentsModelSubjectDosage/pathMain findingsTarget pathway(Refs.)
Yin et al, 2019HMG-CoA reductase InhibitorsSimvastatinMCAORat60 mg/kg/day (p.o.)Reduction of MMP-9 levels and MMP-9/TIMP-1 ratio in brain and plasma; Mitigation of tPA-induced HTRhoA/ROCK(91)
Kurzepa et al, 2006 AISHuman40 mg/day (p.o.)Reduction of serum MMP-9/TIMP-1 ratio/(102)
Liu et al, 2006 AtorvastatinMCAORat20 mg/kg, twice (s.c.)Reduction of brain MMP-9 mRNA levels (105)
Gómez-Hernández et al, 2008 NSTEACSHuman80 mg/day (p.o.)Reduction of plasma MMP-9 activity and incidence of cardiovascular events/(106)
Zhang et al, 2009 MCAORat20 mg/kg, twice (s.c.)Inhibition of brain MMP-9 upregulation and lowering of HT incidence induced by tPA; Extension of the therapeutic window of tPA for stroke to 6 h/(107)
Bellosta et al, 1998 FluvastatinMPMs/HMsCell5–100 µmol/lReduction of MMP-9 expression and activity in macrophages from mice and humans/(108)
Zheng et al, 2016Free radical scavengerEdaravoneAISHuman60 mg/day (i.v.)Reduction of HT incidence and mitigation of neurological deficits/(114)
Toyoda et al, 2004 AISHuman60 mg/day (i.v.)Alleviation of HT and decrease of mortality during the acute phage of stroke/(115)
Okamura et al, 2014 MCAORat3 mg/kg, twice (i.v.)Reduction of hemorrhage volumes after stroke/(116)
Yagi et al, 2009 MCAORat/Cell (HBECs)Rat (3 mg/kg/day, i.v.); HBECs (20 µg/ml)Reduction of endothelial-derived MMP-9 expression and activity; Attenuation of tPA-induced HTNF-κB(117)
Miyamoto et al, 2014 BCCAOMouse3 mg/kg, twice (i.v.)Reduction of brain MMP-9 expression; Preservation of BBB integrity/(118)
Harada et al, 2012 OSICell (bEnd.3)100 µMAttenuation of MMP-9 upregulation induced by the combination of ROS and tPANF-κB(119)
Chen et al, 2013Tetracycline antibioticMinocyclineOGDCell (PC12)200 nMReduction of MMP-9 expression and activityAkt(131)
Murata et al, 2008 MCAORat3 mg/kg/day (i.v.)Reduction of plasma MMP-9 levels; Lowering of tPA- induced HT risk; Extension of the therapeutic window of tPA to 6 h/(133)
Liu et al, 2021 ICHMouse90 mg/kg/day (i.p.)Reduction of brain EMMPRIN and MMP-9 expression; Alleviation of BBB disruption/(134)
Song et al, 2016 BCPRat160 mg/kg/day (i.p.)Inhibition of the NF-κB signaling pathway in spinal astrocytesNF-κB(136)
Zhang et al, 2013Thiazolidin e-dionesRosiglitazoneMCAORat2 mg/kg/day (i.p.)Reduction of HT incidence; Improvement of neurobehavioral deficits/(144)
Li et al, 2019 MCAOMouse6 mg/kg, twice (i.p.)Preservation of BBB integrity; Mitigation of tPA-induced HT/(145)
Marx et al, 2003 CADHuman8 mg/day (p.o.)Reduction of plasma MMP-9 levels/(146)
Wang et al, 2009 MCAORat2 mg/kg/day (i.g.)Prevention of collagen type IV reduction through inhibiting MMP-9 activation/(148)
Lee et al, 2009 VSMCsCell15 and 25 µMInhibition of MMP-9 activationGSK-3β(151)
Nonaka et al, 2009Phosphodie sterase III inhibitorCilostazolMCAOMouse10 mg/kg, (once i.p.)Protection against ischemic brain injury and HT/(155)
Kasahara et al, 2012 MCAOMouse0.3% in the dietReduction of brain MMP-9 activity and cerebral hemorrhage risk induced by tPA/(157)
Chuang et al, 2011 THP-1cell100 µMReduction of MMP-9 promoter activityNF-κB(158)

[i] MMP-9, matrix metalloproteinase-9; HT, hemorrhagic transformation; HMG-CoA, 3-hydroxy-3-methylglutaryl coenzyme A; MCAO, middle cerebral artery occlusion; TIMP-1, tissue inhibitor of metalloproteinase 1; RhoA, Ras homolog family member A; ROCK, Rho-associated coiled-coil containing kinases; AIS, acute ischemic stroke; NSTEACS, non-ST elevation acute coronary syndrome; MPMs, mouse peritoneal macrophages; HMs, human monocyte-derived macrophages; HBECs, human microvascular endothelial cells; NF-κB, nuclear factor-κB; BCCAO, bilateral common carotid artery occlusion; BBB, blood-brain barrier; OSI, oxidative stress in vitro model; bEnd, brain-derived endothelial cells; ROS, reactive oxygen species; OGD, oxygen and glucose deprivation; Akt, protein kinase B; ICH, intracerebral hemorrhage; EMMPRIN, extracellular matrix metalloproteinase inducer; BCP, bone cancer pain; CAD, coronary artery disease; VSMCs, vascular smooth muscle cells; GSK-3β, glycogen synthase kinase-3 β; THP-1, human monocytic leukemia cell line.

Statins

Statins, 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors, are used for the primary and secondary prevention of AIS for its role of plaque stabilization, anti-inflammatory and neuroprotective effects (98). In a previous study using an embolized rabbit model, tPA was administered 1 h following embolization. The results suggested that stains treatment markedly reduced MMP-9 levels, preserved cerebrovascular integrity and lowered the hemorrhagic incidence associated with intravenous thrombolysis (99). Another study revealed that lipophilic statins (such as simvastatin and atorvastatin) markedly reduced MMP-9/TIMP-1 ratio and suppressed MMP-9 activity in human endothelial cells (90). Furthermore, both in vivo and in vitro studies demonstrated that early administration of atorvastatin or simvastatin attenuated the risk of HT by inhibiting MMP-9 expression and preserving BBB integrity (100,101).

Simvastatin is an inhibitor of the Ras homolog family member A (RhoA)/Rho-associated coiled-coil containing kinases (ROCK) signaling pathway. In a rat model of AIS where t-PA was administered 3 h after occlusion, simvastatin pretreatment markedly alleviated tPA-induced HT (91). A randomized controlled trial has demonstrated that the protective effects of simvastatin are mediated through the suppression of MMP-9 activation and the reduction of the MMP-9/TIMP-1 ratio (102). The potentially mechanisms may be associated with the inhibition of RhoA/ROCK signaling pathway. Simvastatin has been found to inhibit geranylgeranylation of RhoA, prevent its translocation to plasma membrane and suppress the subsequent activation of ROCK (103). RhoA/ROCK pathway mediates myosin light chain phosphorylation and activation, which is necessary for vesicular transport and Cyclophilin A secretion. Cyclophilin A might promote the secretion and activation of MMP-9 via enhancing free radical generation and upregulating extracellular matrix metalloproteinase inducer (EMMPRIN) (104). Concerning atorvastatin, in the middle cerebral artery occlusion (MCAO) rat model, Liu et al (105) found that atorvastatin markedly suppressed tPA-induced MMP-9 mRNA upregulation. This finding is further supported by clinical evidence from a randomized trial in non-ST-elevation acute coronary syndrome patients, where atorvastatin markedly suppressed serum MMP-9 activity (106). A preclinical study conducted in embolic stroke rat model indicated that combined therapy of atorvastatin administered at 4 h and delayed tPA at 6 h after stroke decreased tPA-induced MMP-9 upregulation and HT, indicating that atorvastatin could help extend the therapeutic window of tPA for ischemic stroke (107). In addition, Bellosta et al (108) investigated the effects of fluvastatin on MMP-9 activity in mouse and human macrophages and found that fluvastatin markedly suppressed MMP-9 activity in a dose-dependent manner.

Despite these promising preclinical findings, clinical evidence supporting the efficacy of statins in reducing HT risk following ischemic stroke remains limited. Although stains have demonstrated an established safety in clinical practice, its potential adverse effects should not be ignored, particularly myopathy, new-onset diabetes mellitus and cognitive dysfunction (109). These adverse events may occur under specific clinical circumstances, but their absolute risks remain substantially lower than the demonstrated therapeutic benefits.

Free radical scavengers

Edaravone, a potent free radical scavenger, has been widely used in AIS patients due to its anti-inflammatory and neuroprotective properties. Clinical data have highlighted that edaravone markedly reduces neurological deficits and improve the life quality in ischemic stroke patients, with an excellent safety profile (110). A minimal proportion of edaravone-treated patients experienced mild adverse events, such as headache and dizziness, both of which were self-limiting and did not require treatment withdrawal (111). Growing evidence suggests that edaravone may play a crucial role in maintaining BBB integrity (112,113). A number of clinical studies have demonstrated its potential in reducing HT risk. A randomized controlled trial involving 65 AIS patients with diabetes showed markedly lower HT incidence in the edaravone-treated group Compared with the controls (114). Further evidence revealed that edaravone administration not only decreased the incidence of HT, but also markedly attenuated its severity in ischemic stroke patients (115). These clinical findings are supported by robust preclinical evidence. Okamura et al (116) reported that edaravone markedly reduced the hematoma volumes in a rat ischemic stroke model. Further animal study revealed that edaravone downregulated the expression and activity of MMP-9, thereby decreasing tPA-induced HT risk (117). Similar protective effects were observed in a cerebral hypoperfusion mouse model, where edaravone preserved BBB integrity through MMM-9 inhibition (118). The underlying mechanism appears to involve the suppression of the NF-κB pathway, leading to reduced MMP-9 expression and enhanced BBB stabilization (119,120). NF-κB is a crucial transcriptional regulator that becomes rapidly activated during the acute phase of ischemic stroke. This activation occurs primarily through the canonical pathway, where inflammatory stimuli, such as TNF-α, IL-1β and ROS, trigger the phosphorylation and subsequent degradation of inhibitor κB (IκB) by the IκB kinase (IKK) complex (121). The liberated NF-κB dimers (predominantly p50/p65) then translocate to the nucleus, where they bind to specific κB sites in promoter regions to upregulate the expression of numerous pro-inflammatory mediators, including cytokines (TNF-α and IL-6), chemokines (MCP-1) and MMP-9 (122). Edaravone exerts its inhibitory effect on NF-κB activation through multiple mechanisms (119,123): i) Edaravone scavenges ROS that serve as upstream activators of the NF-κB pathway; ii) Edaravone suppresses the phosphorylation and subsequent proteasomal degradation of IκBα, thereby preventing the nuclear translocation of liberated NF-κB dimers (particularly p50/p65), ultimately inhibiting their DNA-binding activity and transcriptional regulation of downstream target genes. Contrary to previous findings, a large case-control study (n=613) found increased HT incidence in ischemic stroke patients with edaravone treatment (124). These discrepancies highlight the need for further large-scale studies to clarify the precise pharmacological effects of edaravone on HT following ischemic stroke.

Minocycline

Minocycline, a semi-synthetic second-generation tetracycline antibiotic, demonstrates significant neuroprotective properties and exhibits therapeutic potential for multiple neurological disorders, such as ischemic stroke, intracerebral hemorrhage (ICH), epilepsy, multiple sclerosis, Parkinson's disease, Alzheimer's disease and spinal cord injury (125,126). Multiple randomized controlled trials have consistently established the favorable safety profile of minocycline in acute stroke management, while its therapeutic efficacy requires further validation (127129). In a clinical trial evaluating the efficacy and safety of minocycline as an adjunct to tPA therapy in ischemic stroke patients within 6 h of onset, no severe hemorrhages were observed in minocycline-treated cohort (130). Further clinical investigation demonstrated that combining minocycline with tPA appeared to mitigate thrombolytic therapy-associated complications, primarily through the inhibition of MMP-9 activity. Meanwhile, comprehensive preclinical studies have established that the neuroprotective properties of minocycline are largely attributable to its inhibitory effects on MMP-9 activity or expression. A previous study has revealed that minocycline, at concentrations ranging from 20 nM-20 µM, effectively alleviated oxygen-glucose deprivation-induced cell cytotoxicity by suppressing both MMP-9 expression and enzymatic activity (131). Minocycline also exerted a protective role in reducing ischemic lesion volume and decreasing HT risk by inhibiting MMP-9 (132). Combined administration of minocycline with tPA could lower the tPA-induced HT risk and extend the narrow treatment time windows of tPA in experimental stroke models, which was mediated by plasma MMP-9 downregulation (133). Our previous research demonstrated that the administration of minocycline could inhibit the expression of EMMPRIN, a key inflammatory mediator promoting MMPs production, thereby downregulating MMP-9 expression and alleviating BBB injury (134,135). Minocycline likely regulates EMMPRIN expression through attenuating IKK/IκBα phosphorylation, impeding NF-κB nuclear translocation and subsequently suppressing EMMPRIN transcriptional activation (136,137). While this proposed mechanism is plausible, rigorous experimental validation through approaches such as chromatin immunoprecipitation assays to demonstrate the reduced binding of NF-κB to the EMMPRIN promoter following minocycline treatment is currently lacking in the literature. In addition, minocycline also protected the integrity of BBB and reduced the risk of HT by targeting neuroinflammation via suppressing the migration and infiltration of neutrophils into the brain and inhibiting the activation of microglia (57,138).

Thiazolidinediones

Diabetes mellitus is characterized by hyperglycemia and microvascular damage of multiple organs. Clinical evidence indicates that hyperglycemia is closely associated with an increased risk of symptomatic ICH, particularly in patients with blood glucose levels >200 mg/dl at stroke onset, who might face a substantially greater risk (139). The pathophysiological basis for this clinical observation may involve hyperglycemia-induced BBB disruption. It has been demonstrated that hyperglycemia disrupts BBB homeostasis by impairing the cerebral endothelial cell function through dysregulating redox signaling, inflammatory mediators and vasoactive factors (140). Furthermore, both in vitro and in vivo studies have revealed that hyperglycemia impaired BBB integrity via suppressing TJPs expression in the neurovascular unit (141,142). Additionally, clinical data form a study enrolling 287 patients demonstrated higher HT incidence among those with stress hyperglycemia, suggesting its potential role in post-stroke HT development (143).

Considering the contribution of hyperglycemia to BBB damage and HT, antidiabetic drugs might represent promising therapeutic candidates to prevent the HT following ischemic stroke. Thiazolidinediones, as peroxisome proliferator-activated receptor (PPAR) agonists, have been extensively studied, among which rosiglitazone shows promise in lowering HT risk after ischemic stroke (144). In a rat MCAO model, rosiglitazone treatment effectively protected against BBB disruption and mitigated tPA-induced HT after stroke (145). While the precise mechanisms of the BBB protective effects of rosiglitazone remain incompletely understood, accumulating evidence suggests that MMP-9 downregulation may play a pivotal role. Clinical data from type 2 diabetic patients demonstrated a markedly reduction of MMP-9 serum levels in those treated with rosiglitazone (146). In rabbit ICH models treated with minimally invasive evacuation, rosiglitazone was shown to decrease MMP-9 expression and protect BBB integrity (147). Another animal study using rat embolic stroke models indicated that rosiglitazone treatment prevented the reduction of collagen type IV and stabilized BBB function by inhibiting MMP-9 activation (148). Furthermore, experimental studies have elucidated the molecular mechanisms by which rosiglitazone modulates MMP-9 expression. Rosiglitazone activates PPARγ-mediated transcription, which enhances adiponectin production and initiates downstream signaling pathways, leading to glycogen synthase kinase-3 β (GSK-3β) activation (149). The activated GSK-3β exerts its regulatory effects by stabilizing IκBα, thereby maintaining NF-κB in an inactive cytoplasmic state (150). This GSK-3β-mediated suppression of NF-κB nuclear translocation and DNA binding capacity results in significant downregulation of MMP-9 expression through the inhibition of promoter activity and subsequent attenuation of gene transcription (151). Notably, clinical evidence indicates that rosiglitazone therapy is associated with an increased risk of heart failure in patients with type 2 diabetes mellitus (152). Therefore, comprehensive cardiovascular evaluation, including assessment of left ventricular function and fluid status, should be routinely performed prior to treatment initiation and during follow-up monitoring.

Cilostazol

Cilostazol, a selective phosphodiesterase III inhibitor, is primarily used as an antiplatelet agent for patients with intermittent claudication or ischemic stroke. A multicenter randomized controlled trial involving patients with lacunar stroke demonstrated that cilostazol markedly reduced the incidence of recurrent ischemic stroke while maintaining a favorable safety profile, with no increase in severe or life-threatening bleeding events (153). Additionally, clinical data demonstrates that cilostazol is associated with a significant lower incidence of HT, compared with other antiplatelet drugs such as aspirin (154). Meanwhile, extensive experimental evidence from animal studies has consistently corroborated this finding. Intraperitoneally administration of cilostazol (10 mg/kg) could effectively protect against HT in a transient MCAO mouse model (155). Cilostazol might protect against tPA-induced brain edema and HT by downregulating microglia-derived MMP-9 in mice with focal cerebral ischemia (156). Consistent with the previous reports, administration of cilostazol for 7 days before ischemia markedly decreased the risk of HT following injection of tPA, which was associated with the inhibition of MMP-9 activity (157). Mechanistically, it has been revealed that cilostazol inhibited the translocation of NF-κB to nuclear and decreased the activity of MMP-9 promoter, indicating that cilostazol suppressed MMP-9 expression at transcriptional level (158). Although the detailed mechanisms underlying the inhibitory effects of cilostazol on NF-κB pathway have not been fully elucidated, AMP-activated protein kinase (AMPK) appears to play a critical role. As reported in a previous study, cilostazol promoted AMPK phosphorylation, which subsequently blocked NF-κB activation (159). Conversely, another study demonstrated that cilostazol attenuated warfarin-induced HT through upregulation of TJPs (claudin-5 and ZO-1) and VE-cadherin expression, while showing no significant effect on MMP-9 levels (160). These divergent results indicate that the protective role of cilostazol in HT involves multiple pathways, warranting systematic exploration of its underlying mechanisms.

Conclusion

MMP-9 is involved in a number of pathophysiological processes associated with BBB breakdown and HT after ischemic stroke, making it a promising predictive biomarker for HT risk. Accumulating evidence from preclinical and clinical studies indicates that pharmacological agents, such as statins, edaravone, minocycline, rosiglitazone and cilostazol, can attenuate MMP-9 expression or activity, thereby preserving BBB integrity and reducing HT incidence following ischemic stroke. The combined administration of these therapeutic agents may effectively reduce tPA-induced HT while potentially extending the therapeutic window for thrombolytic therapy in ischemic stroke.

Acknowledgements

Not applicable.

Funding

The present study was supported by grant support from National Key Research and Development Program of China (grant no. 2018YFC1312200) and the National Natural Science Foundation of China (grant nos. 82071331 and 81870942).

Availability of data and materials

Not applicable.

Authors' contributions

PG conceived and drafted the manuscript. HL and XZ performed the literature search. YL and SX revised the manuscript and created the figures. MX and VY reviewed and revised the manuscript. All authors have read and approved the final manuscript. Data authentication is not applicable.

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 

Feigin VL and Owolabi MO; World Stroke Organization-Lancet Neurology Commission Stroke Collaboration Group, : Pragmatic solutions to reduce the global burden of stroke: A orld stroke organization-lancet neurology commission. Lancet Neurol. 22:1160–1206. 2023. View Article : Google Scholar : PubMed/NCBI

2 

Hasan TF, Hasan H and Kelley RE: Overview of acute ischemic stroke evaluation and management. Biomedicines. 9:14862021. View Article : Google Scholar : PubMed/NCBI

3 

Otsu Y, Namekawa M, Toriyabe M, Ninomiya I, Hatakeyama M, Uemura M, Onodera O, Shimohata T and Kanazawa M: Strategies to prevent hemorrhagic transformation after reperfusion therapies for acute ischemic stroke: A literature review. J Neurol Sci. 419:1172172020. View Article : Google Scholar : PubMed/NCBI

4 

Goncalves A, Su EJ, Muthusamy A, Zeitelhofer M, Torrente D, Nilsson I, Protzmann J, Fredriksson L, Eriksson U, Antonetti DA and Lawrence DA: Thrombolytic tPA-induced hemorrhagic transformation of ischemic stroke is mediated by PKCβ phosphorylation of occludin. Blood. 140:388–400. 2022.PubMed/NCBI

5 

Kovács KB, Bencs V, Hudák L, Oláh L and Csiba L: Hemorrhagic transformation of ischemic strokes. Int J Mol Sci. 24:140672023. View Article : Google Scholar : PubMed/NCBI

6 

Lu W and Wen J: The relationship among H2S, neuroinflammation and MMP-9 in BBB injury following ischemic stroke. Int Immunopharmacol. 146:1139022025. View Article : Google Scholar : PubMed/NCBI

7 

Desilles JP, Rouchaud A, Labreuche J, Meseguer E, Laissy JP, Serfaty JM, Lapergue B, Klein IF, Guidoux C, Cabrejo L, et al: Blood-brain barrier disruption is associated with increased mortality after endovascular therapy. Neurology. 80:844–851. 2013. View Article : Google Scholar : PubMed/NCBI

8 

Zhao Z, Nelson AR, Betsholtz C and Zlokovic BV: Establishment and dysfunction of the blood-brain barrier. Cell. 163:1064–1078. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Rosell A, Cuadrado E, Ortega-Aznar A, Hernandez-Guillamon M, Lo EH and 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. 39:1121–1126. 2008. View Article : Google Scholar : PubMed/NCBI

10 

Kapoor C, Vaidya S, Wadhwan V, Hitesh, Kaur G and Pathak A: Seesaw of matrix metalloproteinases (MMPs). J Cancer Res Ther. 12:28–35. 2016. View Article : Google Scholar : PubMed/NCBI

11 

Misra S, Talwar P, Kumar A, Kumar P, Sagar R, Vibha D, Pandit AK, Gulati A, Kushwaha S and Prasad K: Association between matrix metalloproteinase family gene polymorphisms and risk of ischemic stroke: A systematic review and meta-analysis of 29 studies. Gene. 672:180–194. 2018. View Article : Google Scholar : PubMed/NCBI

12 

Yang Y and Rosenberg GA: Matrix metalloproteinases as therapeutic targets for stroke. Brain Res. 1623:30–38. 2015. View Article : Google Scholar : PubMed/NCBI

13 

Kimura-Ohba S and Yang Y: Oxidative DNA damage mediated by intranuclear MMP activity is associated with neuronal apoptosis in ischemic stroke. Oxid Med Cell Longev. 2016:69273282016. View Article : Google Scholar : PubMed/NCBI

14 

Cui N, Hu M and Khalil RA: Biochemical and biological attributes of matrix metalloproteinases. Prog Mol Biol Transl Sci. 147:1–73. 2017. View Article : Google Scholar : PubMed/NCBI

15 

Montaner J, Alvarez-Sabín J, Molina CA, Anglés A, Abilleira S, Arenillas J and Monasterio J: Matrix metalloproteinase expression is related to hemorrhagic transformation after cardioembolic stroke. Stroke. 32:2762–2767. 2001. View Article : Google Scholar : PubMed/NCBI

16 

Montaner J, Molina CA, Monasterio J, Abilleira S, Arenillas JF, Ribó M, Quintana M and Alvarez-Sabín J: Matrix metalloproteinase-9 pretreatment level predicts intracranial hemorrhagic complications after thrombolysis in human stroke. Circulation. 107:598–603. 2003. View Article : Google Scholar : PubMed/NCBI

17 

Li H, Ghorbani S, Ling CC, Yong VW and Xue M: The extracellular matrix as modifier of neuroinflammation and recovery in ischemic stroke and intracerebral hemorrhage. Neurobiol Dis. 186:1062822023. View Article : Google Scholar : PubMed/NCBI

18 

Wang W, Li M, Chen Q and Wang J: Hemorrhagic transformation after tissue plasminogen activator reperfusion therapy for ischemic stroke: Mechanisms, models, and biomarkers. Mol Neurobiol. 52:1572–1579. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Wang L, Wei C, Deng L, Wang Z, Song M, Xiong Y and Liu M: The accuracy of serum matrix metalloproteinase-9 for predicting hemorrhagic transformation after acute ischemic stroke: A systematic review and meta-analysis. J Stroke Cerebrovasc Dis. 27:1653–1665. 2018. View Article : Google Scholar : PubMed/NCBI

20 

Barr TL, Latour LL, Lee KY, Schaewe TJ, Luby M, Chang GS, El-Zammar Z, Alam S, Hallenbeck JM, Kidwell CS and Warach S: Blood-brain barrier disruption in humans is independently associated with increased matrix metalloproteinase-9. Stroke. 41:e123–e128. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Jha R, Battey TW, Pham L, Lorenzano S, Furie KL, Sheth KN and Kimberly WT: Fluid-attenuated inversion recovery hyperintensity correlates with matrix metalloproteinase-9 level and hemorrhagic transformation in acute ischemic stroke. Stroke. 45:1040–1045. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Zhang X, Zhang Y, Su Q, Liu Y, Li Z, Yong VW and Xue M: Ion channel dysregulation following intracerebral hemorrhage. Neurosci Bull. 40:401–414. 2024. View Article : Google Scholar : PubMed/NCBI

23 

Mondal S, Adhikari N, Banerjee S, Amin SA and Jha T: Matrix metalloproteinase-9 (MMP-9) and its inhibitors in cancer: A minireview. Eur J Med Chem. 194:1122602020. View Article : Google Scholar : PubMed/NCBI

24 

Das S, Amin SA and Jha T: Inhibitors of gelatinases (MMP-2 and MMP-9) for the management of hematological malignancies. Eur J Med Chem. 223:1136232021. View Article : Google Scholar : PubMed/NCBI

25 

Beroun A, Mitra S, Michaluk P, Pijet B, Stefaniuk M and Kaczmarek L: MMPs in learning and memory and neuropsychiatric disorders. Cell Mol Life Sci. 76:3207–3228. 2019. View Article : Google Scholar : PubMed/NCBI

26 

Cathcart J, Pulkoski-Gross A and Cao J: Targeting matrix metalloproteinases in cancer: Bringing new life to old ideas. Genes Dis. 2:26–34. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Mizoguchi H, Nakade J, Tachibana M, Ibi D, Someya E, Koike H, Kamei H, Nabeshima T, Itohara S, Takuma K, et al: Matrix metalloproteinase-9 contributes to kindled seizure development in pentylenetetrazole-treated mice by converting pro-BDNF to mature BDNF in the hippocampus. J Neurosci. 31:12963–12971. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Li YJ, Wang ZH, Zhang B, Zhe X, Wang MJ, Shi ST, Bai J, Lin T, Guo CJ, Zhang SJ, et al: Disruption of the blood-brain barrier after generalized tonic-clonic seizures correlates with cerebrospinal fluid MMP-9 levels. J Neuroinflammation. 10:802013. View Article : Google Scholar : PubMed/NCBI

29 

Bronisz E and Kurkowska-Jastrzebska I: Matrix metalloproteinase 9 in epilepsy: The role of neuroinflammation in seizure development. Mediators Inflamm. 2016:73690202016. View Article : Google Scholar : PubMed/NCBI

30 

Stawarski M, Stefaniuk M and Wlodarczyk J: Matrix metalloproteinase-9 involvement in the structural plasticity of dendritic spines. Front Neuroanat. 8:682014. View Article : Google Scholar : PubMed/NCBI

31 

Xue M, Hollenberg MD and Yong VW: Combination of thrombin and matrix metalloproteinase-9 exacerbates neurotoxicity in cell culture and intracerebral hemorrhage in mice. J Neurosci. 26:10281–10291. 2006. View Article : Google Scholar : PubMed/NCBI

32 

Chen H, He Y, Chen S, Qi S and Shen J: Therapeutic targets of oxidative/nitrosative stress and neuroinflammation in ischemic stroke: Applications for natural product efficacy with omics and systemic biology. Pharmacol Res. 158:1048772020. View Article : Google Scholar : PubMed/NCBI

33 

Qi Z, Liang J, Pan R, Dong W, Shen J, Yang Y, Zhao Y, Shi W, Luo Y, Ji X and Liu KJ: Zinc contributes to acute cerebral ischemia-induced blood-brain barrier disruption. Neurobiol Dis. 95:12–21. 2016. View Article : Google Scholar : PubMed/NCBI

34 

Li Z, Liu Y, Wei R, Yong VW and Xue M: The important role of Zinc in neurological diseases. Biomolecules. 13:282022. View Article : Google Scholar : PubMed/NCBI

35 

Foerch C, Montaner J, Furie KL, Ning MM and Lo EH: Invited article: searching for oracles? Blood biomarkers in acute stroke. Neurology. 73:393–399. 2009. View Article : Google Scholar : PubMed/NCBI

36 

Romanic AM, White RF, Arleth AJ, Ohlstein EH and Barone FC: Matrix metalloproteinase expression increases after cerebral focal ischemia in rats: Inhibition of matrix metalloproteinase-9 reduces infarct size. Stroke. 29:1020–1030. 1998. View Article : Google Scholar : PubMed/NCBI

37 

Rosenberg GA and Yang Y: Vasogenic edema due to tight junction disruption by matrix metalloproteinases in cerebral ischemia. Neurosurg Focus. 22:E42007. View Article : Google Scholar : PubMed/NCBI

38 

Zhao BQ, Wang S, Kim HY, Storrie H, Rosen BR, Mooney DJ, Wang X and Lo EH: Role of matrix metalloproteinases in delayed cortical responses after stroke. Nat Med. 12:441–445. 2006. View Article : Google Scholar : PubMed/NCBI

39 

Candelario-Jalil E, Yang Y and Rosenberg GA: Diverse roles of matrix metalloproteinases and tissue inhibitors of metalloproteinases in neuroinflammation and cerebral ischemia. Neuroscience. 158:983–994. 2009. View Article : Google Scholar : PubMed/NCBI

40 

Iadecola C and Nedergaard M: Glial regulation of the cerebral microvasculature. Nat Neurosci. 10:1369–1376. 2007. View Article : Google Scholar : PubMed/NCBI

41 

Cottarelli A, Corada M, Beznoussenko GV, Mironov AA, Globisch MA, Biswas S, Huang H, Dimberg A, Magnusson PU, Agalliu D, et al: Fgfbp1 promotes blood-brain barrier development by regulating collagen IV deposition and maintaining Wnt/β-catenin signaling. Development. 147:dev1851402020. View Article : Google Scholar : PubMed/NCBI

42 

Heinemann U and Schuetz A: structural features of tight-junction proteins. Int J Mol Sci. 20:60202019. View Article : Google Scholar : PubMed/NCBI

43 

Biswas S, Cottarelli A and Agalliu D: Neuronal and glial regulation of CNS angiogenesis and barriergenesis. Development. 147:dev1822792020. View Article : Google Scholar : PubMed/NCBI

44 

Milner R, Hung S, Wang X, Berg GI, Spatz M and del Zoppo GJ: Responses of endothelial cell and astrocyte matrix-integrin receptors to ischemia mimic those observed in the neurovascular unit. Stroke. 39:191–197. 2008. View Article : Google Scholar : PubMed/NCBI

45 

Thomsen MS, Routhe LJ and Moos T: The vascular basement membrane in the healthy and pathological brain. J Cereb Blood Flow Metab. 37:3300–3317. 2017. View Article : Google Scholar : PubMed/NCBI

46 

Kadry H, Noorani B and Cucullo L: A blood-brain barrier overview on structure, function, impairment, and biomarkers of integrity. Fluids Barriers CNS. 17:692020. View Article : Google Scholar : PubMed/NCBI

47 

Tabet A, Apra C, Stranahan AM and Anikeeva P: Changes in brain neuroimmunology following injury and disease. Front Integr Neurosci. 16:8945002022. View Article : Google Scholar : PubMed/NCBI

48 

Jiang X, Andjelkovic AV, Zhu L, Yang T, Bennett MVL, Chen J, Keep RF and Shi Y: Blood-brain barrier dysfunction and recovery after ischemic stroke. Prog Neurobiol. 163-164:144–171. 2018. View Article : Google Scholar : PubMed/NCBI

49 

Rashid ZA and Bardaweel SK: Novel matrix metalloproteinase-9 (MMP-9) inhibitors in cancer treatment. Int J Mol Sci. 24:121332023. View Article : Google Scholar : PubMed/NCBI

50 

Luchian I, Goriuc A, Sandu D and Covasa M: The role of matrix metalloproteinases (MMP-8, MMP-9, MMP-13) in periodontal and peri-implant pathological processes. Int J Mol Sci. 23:18062022. View Article : Google Scholar : PubMed/NCBI

51 

Chen X and Wang L, Wang N, Li C, Hang H, Wu G, Ren S, Jun T and Wang L: An apolipoprotein E receptor mimetic peptide decreases blood-brain barrier permeability following intracerebral hemorrhage by inhibiting the CypA/MMP-9 signaling pathway via LRP1 activation. Int Immunopharmacol. 143 (Pt 3):1130072024. View Article : Google Scholar : PubMed/NCBI

52 

Hannocks MJ, Zhang X, Gerwien H, Chashchina A, Burmeister M, Korpos E, Song J and Sorokin L: The gelatinases, MMP-2 and MMP-9, as fine tuners of neuroinflammatory processes. Matrix Biol. 75-76:102–113. 2019. View Article : Google Scholar : PubMed/NCBI

53 

Könnecke H and Bechmann I: The role of microglia and matrix metalloproteinases involvement in neuroinflammation and gliomas. Clin Dev Immunol. 2013:9141042013. View Article : Google Scholar : PubMed/NCBI

54 

Fiorelli M, Bastianello S, von Kummer R, del Zoppo GJ, Larrue V, Lesaffre E, Ringleb AP, Lorenzano S, Manelfe C and Bozzao L: Hemorrhagic transformation within 36 hours of a cerebral infarct: relationships with early clinical deterioration and 3-month outcome in the European Cooperative Acute Stroke Study I (ECASS I) cohort. Stroke. 30:2280–2284. 1999. View Article : Google Scholar : PubMed/NCBI

55 

Hacke W, Kaste M, Fieschi C, Toni D, Lesaffre E, von Kummer R, Boysen G, Bluhmki E, Höxter G, Mahagne MH, et al: Intravenous thrombolysis with recombinant tissue plasminogen activator for acute hemispheric stroke. The European Cooperative Acute Stroke Study (ECASS). JAMA. 274:1017–1025. 1995. View Article : Google Scholar : PubMed/NCBI

56 

Ande SR, Grynspan J, Aviv RI and Shankar JJS: Imaging for predicting hemorrhagic transformation of acute ischemic stroke-a narrative review. Can Assoc Radiol J. 73:194–202. 2022. View Article : Google Scholar : PubMed/NCBI

57 

Khatri P, Wechsler LR and Broderick JP: Intracranial hemorrhage associated with revascularization therapies. Stroke. 38:431–440. 2007. View Article : Google Scholar : PubMed/NCBI

58 

Ma G, Pan Z, Kong L and Du G: Neuroinflammation in hemorrhagic transformation after tissue plasminogen activator thrombolysis: Potential mechanisms, targets, therapeutic drugs and biomarkers. Int Immunopharmacol. 90:1072162021. View Article : Google Scholar : PubMed/NCBI

59 

Kanazawa M, Takahashi T, Nishizawa M and Shimohata T: Therapeutic strategies to attenuate hemorrhagic transformation after tissue plasminogen activator treatment for acute ischemic stroke. J Atheroscler Thromb. 24:240–253. 2017. View Article : Google Scholar : PubMed/NCBI

60 

Zhang Y, Khan S, Liu Y, Wu G, Yong VW and Xue M: Oxidative stress following intracerebral hemorrhage: From molecular mechanisms to therapeutic targets. Front Immunol. 13:8472462022. View Article : Google Scholar : PubMed/NCBI

61 

Zhao Y, Zhang X, Chen X and Wei Y: Neuronal injuries in cerebral infarction and ischemic stroke: From mechanisms to treatment (Review). Int J Mol Med. 49:152022. View Article : Google Scholar : PubMed/NCBI

62 

Abdullahi W, Tripathi D and Ronaldson PT: Blood-brain barrier dysfunction in ischemic stroke: targeting tight junctions and transporters for vascular protection. Am J Physiol Cell Physiol. 315:C343–C356. 2018. View Article : Google Scholar : PubMed/NCBI

63 

Fraser PA: The role of free radical generation in increasing cerebrovascular permeability. Free Radic Biol Med. 51:967–977. 2011. View Article : Google Scholar : PubMed/NCBI

64 

Sun MS, Jin H, Sun X, Huang S, Zhang FL, Guo ZN and Yang Y: Free radical damage in ischemia-reperfusion injury: An obstacle in acute ischemic stroke after revascularization therapy. Oxid Med Cell Longev. 2018:38049792018. View Article : Google Scholar : PubMed/NCBI

65 

Shuvalova M, Dmitrieva A, Belousov V and Nosov G: The role of reactive oxygen species in the regulation of the blood-brain barrier. Tissue Barriers. May 29–2024.(Epub ahead of print). View Article : Google Scholar : PubMed/NCBI

66 

Hong S, Park KK, Magae J, Ando K, Lee TS, Kwon TK, Kwak JY, Kim CH and Chang YC: Ascochlorin inhibits matrix metalloproteinase-9 expression by suppressing activator protein-1-mediated gene expression through the ERK1/2 signaling pathway: Inhibitory effects of ascochlorin on the invasion of renal carcinoma cells. J Biol Chem. 280:25202–25209. 2005. View Article : Google Scholar : PubMed/NCBI

67 

Lee GH, Jin SW, Kim SJ, Pham TH, Choi JH and Jeong HG: Tetrabromobisphenol A induces MMP-9 expression via NADPH Oxidase and the activation of ROS, MAPK and Akt pathways in human breast cancer MCF-7 cells. Toxicol Res. 35:93–101. 2019. View Article : Google Scholar : PubMed/NCBI

68 

Banjara M and Ghosh C: Sterile Neuroinflammation and strategies for therapeutic intervention. Int J Inflam. 2017:83859612017.PubMed/NCBI

69 

Gülke E, Gelderblom M and Magnus T: Danger signals in stroke and their role on microglia activation after ischemia. Ther Adv Neurol Disord. 11:17562864187742542018. View Article : Google Scholar : PubMed/NCBI

70 

Alsbrook DL, Di Napoli M, Bhatia K, Biller J, Andalib S, Hinduja A, Rodrigues R, Rodriguez M, Sabbagh SY, Selim M, et al: Neuroinflammation in acute ischemic and hemorrhagic stroke. Curr Neurol Neurosci Rep. 23:407–431. 2023. View Article : Google Scholar : PubMed/NCBI

71 

Yang C, Hawkins KE, Doré S and Candelario-Jalil E: Neuroinflammatory mechanisms of blood-brain barrier damage in ischemic stroke. Am J Physiol Cell Physiol. 316:C135–C153. 2019. View Article : Google Scholar : PubMed/NCBI

72 

McColl BW, Rothwell NJ and Allan SM: Systemic inflammation alters the kinetics of cerebrovascular tight junction disruption after experimental stroke in mice. J Neurosci. 28:9451–9462. 2008. View Article : Google Scholar : PubMed/NCBI

73 

McColl BW, Rothwell NJ and Allan SM: Systemic inflammatory stimulus potentiates the acute phase and CXC chemokine responses to experimental stroke and exacerbates brain damage via interleukin-1- and neutrophil-dependent mechanisms. J Neurosci. 27:4403–4412. 2007. View Article : Google Scholar : PubMed/NCBI

74 

de Vries HE, Blom-Roosemalen MC, van Oosten M, de Boer AG, van Berkel TJ, Breimer DD and Kuiper J: The influence of cytokines on the integrity of the blood-brain barrier in vitro. J Neuroimmunol. 64:37–43. 1996. View Article : Google Scholar : PubMed/NCBI

75 

Takata F, Dohgu S, Matsumoto J, Takahashi H, Machida T, Wakigawa T, Harada E, Miyaji H, Koga M, Nishioku T, et al: Brain pericytes among cells constituting the blood-brain barrier are highly sensitive to tumor necrosis factor-α, releasing matrix metalloproteinase-9 and migrating in vitro. J Neuroinflammation. 8:1062011. View Article : Google Scholar : PubMed/NCBI

76 

Dimitrijevic OB, Stamatovic SM, Keep RF and Andjelkovic AV: Effects of the chemokine CCL2 on blood-brain barrier permeability during ischemia-reperfusion injury. J Cereb Blood Flow Metab. 26:797–810. 2006. View Article : Google Scholar : PubMed/NCBI

77 

Kim JS: tPA Helpers in the treatment of acute ischemic stroke: Are they ready for clinical use? J Stroke. 21:160–174. 2019. View Article : Google Scholar : PubMed/NCBI

78 

Lees KR, Bluhmki E, von Kummer R, Brott TG, Toni D, Grotta JC, Albers GW, Kaste M, Marler JR, Hamilton SA, et al: Time to treatment with intravenous alteplase and outcome in stroke: An updated pooled analysis of ECASS, ATLANTIS, NINDS, and EPITHET trials. Lancet. 375:1695–1703. 2010. View Article : Google Scholar : PubMed/NCBI

79 

Fan X, Jiang Y, Yu Z, Yuan J, Sun X, Xiang S, Lo EH and Wang X: Combination approaches to attenuate hemorrhagic transformation after tPA thrombolytic therapy in patients with poststroke hyperglycemia/diabetes. Adv Pharmacol. 71:391–410. 2014. View Article : Google Scholar : PubMed/NCBI

80 

Su EJ, Fredriksson L, Geyer M, Folestad E, Cale J, Andrae J, Gao Y, Pietras K, Mann K, Yepes M, et al: Activation of PDGF-CC by tissue plasminogen activator impairs blood-brain barrier integrity during ischemic stroke. Nat Med. 14:731–737. 2008. View Article : Google Scholar : PubMed/NCBI

81 

Cuadrado E, Ortega L, Hernández-Guillamon M, Penalba A, Fernández-Cadenas I, Rosell A and Montaner J: Tissue plasminogen activator (t-PA) promotes neutrophil degranulation and MMP-9 release. J Leukoc Biol. 84:207–214. 2008. View Article : Google Scholar : PubMed/NCBI

82 

Wang X, Lee SR, Arai K, Lee SR, Tsuji K, Rebeck GW and Lo EH: Lipoprotein receptor-mediated induction of matrix metalloproteinase by tissue plasminogen activator. Nat Med. 9:1313–1317. 2003. View Article : Google Scholar : PubMed/NCBI

83 

Cheng T, Petraglia AL, Li Z, Thiyagarajan M, Zhong Z, Wu Z, Liu D, Maggirwar SB, Deane R, Fernández JA, et al: Activated protein C inhibits tissue plasminogen activator-induced brain hemorrhage. Nat Med. 12:1278–1285. 2006. View Article : Google Scholar : PubMed/NCBI

84 

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

85 

Mashaqi S, Mansour HM, Alameddin H, Combs D, Patel S, Estep L and Parthasarathy S: Matrix metalloproteinase-9 as a messenger in the cross talk between obstructive sleep apnea and comorbid systemic hypertension, cardiac remodeling, and ischemic stroke: A literature review. J Clin Sleep Med. 17:567–591. 2021. View Article : Google Scholar : PubMed/NCBI

86 

di Biase L, Bonura A, Pecoraro PM, Carbone SP and Di Lazzaro V: Unlocking the potential of stroke blood biomarkers: Early diagnosis, ischemic vs. haemorrhagic differentiation and haemorrhagic transformation risk: A comprehensive review. Int J Mol Sci. 24:115452023. View Article : Google Scholar : PubMed/NCBI

87 

Yuan R, Tan S, Wang D, Wu S, Cao X, Zhang S, Wu B and Liu M: Predictive value of plasma matrix metalloproteinase-9 concentrations for spontaneous haemorrhagic transformation in patients with acute ischaemic stroke: A cohort study in Chinese patients. J Clin Neurosci. 58:108–112. 2018. View Article : Google Scholar : PubMed/NCBI

88 

Arkelius K, Wendt TS, Andersson H, Arnou A, Gottschalk M, Gonzales RJ and Ansar S: LOX-1 and MMP-9 inhibition attenuates the detrimental effects of delayed rt-PA therapy and improves outcomes after acute ischemic stroke. Circ Res. 134:954–969. 2024. View Article : Google Scholar : PubMed/NCBI

89 

Sun X, Liu Z, Zhou L, Ma R, Zhang X, Wang T, Fu F and Wang Y: Escin avoids hemorrhagic transformation in ischemic stroke by protecting BBB through the AMPK/Cav-1/MMP-9 pathway. Phytomedicine. 120:1550712023. View Article : Google Scholar : PubMed/NCBI

90 

Izidoro-Toledo TC, Guimaraes DA, Belo VA, Gerlach RF and Tanus-Santos JE: Effects of statins on matrix metalloproteinases and their endogenous inhibitors in human endothelial cells. Naunyn Schmiedebergs Arch Pharmacol. 383:547–554. 2011. View Article : Google Scholar : PubMed/NCBI

91 

Yin B, Li DD, Xu SY, Huang H, Lin J, Sheng HS, Fang JH, Song JN and Zhang M: Simvastatin pretreatment ameliorates t-PA-induced hemorrhage transformation and MMP-9/TIMP-1 imbalance in thromboembolic cerebral ischemic rats. Neuropsychiatr Dis Treat. 15:1993–2002. 2019. View Article : Google Scholar : PubMed/NCBI

92 

Li Y, Chen L, Yao S, Chen J, Hu W, Wang M, Chen S, Chen X, Li S, Gu X, et al: Association of polymorphisms of the matrix metalloproteinase 9 gene with ischaemic stroke in a southern Chinese population. Cell Physiol Biochem. 49:2188–2199. 2018. View Article : Google Scholar : PubMed/NCBI

93 

Zhang X, Cao X, Xu X, Li A and Xu Y: Correlation between the −1562C/T polymorphism in the matrix metalloproteinase-9 gene and hemorrhagic transformation of ischemic stroke. Exp Ther Med. 9:1043–1047. 2015. View Article : Google Scholar : PubMed/NCBI

94 

Szczudlik P and Borratyńska A: Association between the −1562 C/T MMP-9 polymorphism and cerebrovascular disease in a Polish population. Neurol Neurochir Pol. 44:350–357. 2010. View Article : Google Scholar : PubMed/NCBI

95 

Fernández-Cadenas I, Del Río-Espínola A, Carrera C, Domingues-Montanari S, Mendióroz M, Delgado P, Rosell A, Ribó M, Giralt D, Quintana M, et al: Role of the MMP-9 gene in hemorrhagic transformations after tissue-type plasminogen activator treatment in stroke patients. Stroke. 43:1398–1400. 2012. View Article : Google Scholar : PubMed/NCBI

96 

Wang B, Wang Y and Zhao L: MMP-9 gene rs3918242 polymorphism increases risk of stroke: A meta-analysis. J Cell Biochem. 119:9801–9808. 2018. View Article : Google Scholar : PubMed/NCBI

97 

Yi X, Sui G, Zhou Q, Wang C, Lin J, Chai Z and Zhou J: Variants in matrix metalloproteinase-9 gene are associated with hemorrhagic transformation in acute ischemic stroke patients with atherothrombosis, small artery disease, and cardioembolic stroke. Brain Behav. 9:e012942019. View Article : Google Scholar : PubMed/NCBI

98 

Kytö V, Åivo J and Ruuskanen JO: Intensity of statin therapy after ischaemic stroke and long-term outcomes: A nationwide cohort study. Stroke Vasc Neurol. 10:142–145. 2024. View Article : Google Scholar : PubMed/NCBI

99 

Lapchak PA and Han MK: The 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor simvastatin reduces thrombolytic-induced intracerebral hemorrhage in embolized rabbits. Brain Res. 1303:144–150. 2009. View Article : Google Scholar : PubMed/NCBI

100 

Reuter B, Rodemer C, Grudzenski S, Meairs S, Bugert P, Hennerici MG and Fatar M: Effect of simvastatin on MMPs and TIMPs in human brain endothelial cells and experimental stroke. Transl Stroke Res. 6:156–159. 2015. View Article : Google Scholar : PubMed/NCBI

101 

Fang X, Tao D, Shen J, Wang Y, Dong X and Ji X: Neuroprotective effects and dynamic expressions of MMP9 and TIMP1 associated with atorvastatin pretreatment in ischemia-reperfusion rats. Neurosci Lett. 603:60–65. 2015. View Article : Google Scholar : PubMed/NCBI

102 

Kurzepa J, Szczepanska-Szerej A, Stryjecka-Zimmer M, Malecka-Massalska T and Stelmasiak Z: Simvastatin could prevent increase of the serum MMP-9/TIMP-1 ratio in acute ischaemic stroke. Folia Biol (Praha). 52:181–183. 2006. View Article : Google Scholar : PubMed/NCBI

103 

Turner NA, Aley PK, Hall KT, Warburton P, Galloway S, Midgley L, O'Regan DJ, Wood IC, Ball SG and Porter KE: Simvastatin inhibits TNFalpha-induced invasion of human cardiac myofibroblasts via both MMP-9-dependent and -independent mechanisms. J Mol Cell Cardiol. 43:168–176. 2007. View Article : Google Scholar : PubMed/NCBI

104 

Skrzypiec-Spring M, Kaczorowski M, Rak-Pasikowska A, Sapa-Wojciechowska A, Kujawa K, Żuryń A, Bil-Lula I, Hałoń A and Szeląg A: RhoA/ROCK pathway is upregulated in experimental autoimmune myocarditis and is inhibited by simvastatin at the stage of myosin light chain phosphorylation. Biomedicines. 12:5962024. View Article : Google Scholar : PubMed/NCBI

105 

Liu XS, Zhang ZG, Zhang L, Morris DC, Kapke A, Lu M and Chopp M: Atorvastatin downregulates tissue plasminogen activator-aggravated genes mediating coagulation and vascular permeability in single cerebral endothelial cells captured by laser microdissection. J Cereb Blood Flow Metab. 26:787–796. 2006. View Article : Google Scholar : PubMed/NCBI

106 

Gómez-Hernández A, Sánchez-Galán E, Ortego M, Martín-Ventura JL, Blanco-Colio LM, Tarín-Vicente N, Jiménez-Nacher JJ, López-Bescos L, Egido J and Tuñón J: Effect of intensive atorvastatin therapy on prostaglandin E2 levels and metalloproteinase-9 activity in the plasma of patients with non-ST-elevation acute coronary syndrome. Am J Cardiol. 102:12–18. 2008. View Article : Google Scholar : PubMed/NCBI

107 

Zhang L, Chopp M, Jia L, Cui Y, Lu M and Zhang ZG: Atorvastatin extends the therapeutic window for tPA to 6 h after the onset of embolic stroke in rats. J Cereb Blood Flow Metab. 29:1816–1824. 2009. View Article : Google Scholar : PubMed/NCBI

108 

Bellosta S, Via D, Canavesi M, Pfister P, Fumagalli R, Paoletti R and Bernini F: HMG-CoA reductase inhibitors reduce MMP-9 secretion by macrophages. Arterioscler Thromb Vasc Biol. 18:1671–1678. 1998. View Article : Google Scholar : PubMed/NCBI

109 

Collins R, Reith C, Emberson J, Armitage J, Baigent C, Blackwell L, Blumenthal R, Danesh J, Smith GD, DeMets D, et al: Interpretation of the evidence for the efficacy and safety of statin therapy. Lancet. 388:2532–2561. 2016. View Article : Google Scholar : PubMed/NCBI

110 

Sun Z, Xu Q, Gao G, Zhao M and Sun C: Clinical observation in edaravone treatment for acute cerebral infarction. Niger J Clin Pract. 22:1324–1327. 2019. View Article : Google Scholar : PubMed/NCBI

111 

Batino LKJ, Escabillas CG and Navarro JC: Edaravone's safety profile in acute ischemic stroke. Brain Behav. 14:e701582024. View Article : Google Scholar : PubMed/NCBI

112 

Liu J, Jiang Y, Zhang G, Lin Z and Du S: Protective effect of edaravone on blood-brain barrier by affecting NRF-2/HO-1 signaling pathway. Exp Ther Med. 18:2437–2442. 2019.PubMed/NCBI

113 

Barna L, Walter FR, Harazin A, Bocsik A, Kincses A, Tubak V, Jósvay K, Zvara Á, Campos-Bedolla P and Deli MA: Simvastatin, edaravone and dexamethasone protect against kainate-induced brain endothelial cell damage. Fluids Barriers CNS. 17:52020. View Article : Google Scholar : PubMed/NCBI

114 

Zheng J and Chen X: Edaravone offers neuroprotection for acute diabetic stroke patients. Ir J Med Sci. 185:819–824. 2016. View Article : Google Scholar : PubMed/NCBI

115 

Toyoda K, Fujii K, Kamouchi M, Nakane H, Arihiro S, Okada Y, Ibayashi S and Iida M: Free radical scavenger, edaravone, in stroke with internal carotid artery occlusion. J Neurol Sci. 221:11–17. 2004. View Article : Google Scholar : PubMed/NCBI

116 

Okamura K, Tsubokawa T, Johshita H, Miyazaki H and Shiokawa Y: Edaravone, a free radical scavenger, attenuates cerebral infarction and hemorrhagic infarction in rats with hyperglycemia. Neurol Res. 36:65–69. 2014. View Article : Google Scholar : PubMed/NCBI

117 

Yagi K, Kitazato KT, Uno M, Tada Y, Kinouchi T, Shimada K and Nagahiro S: Edaravone, a free radical scavenger, inhibits MMP-9-related brain hemorrhage in rats treated with tissue plasminogen activator. Stroke. 40:626–631. 2009. View Article : Google Scholar : PubMed/NCBI

118 

Miyamoto N, Pham LD, Maki T, Liang AC and Arai K: A radical scavenger edaravone inhibits matrix metalloproteinase-9 upregulation and blood-brain barrier breakdown in a mouse model of prolonged cerebral hypoperfusion. Neurosci Lett. 573:40–45. 2014. View Article : Google Scholar : PubMed/NCBI

119 

Harada K, Suzuki Y, Yamakawa K, Kawakami J and Umemura K: Combination of reactive oxygen species and tissue-type plasminogen activator enhances the induction of gelatinase B in brain endothelial cells. Int J Neurosci. 122:53–59. 2012. View Article : Google Scholar : PubMed/NCBI

120 

Yang CC, Hsiao LD, Tseng HC, Kuo CM and Yang CM: Pristimerin inhibits MMP-9 expression and cell migration through attenuating NOX/ROS-dependent NF-κB activation in rat brain astrocytes challenged with LPS. J Inflamm Res. 13:325–341. 2020. View Article : Google Scholar : PubMed/NCBI

121 

Zhang H and Sun SC: NF-κB in inflammation and renal diseases. Cell Biosci. 5:632015. View Article : Google Scholar : PubMed/NCBI

122 

Ridder DA and Schwaninger M: NF-κB signaling in cerebral ischemia. Neuroscience. 158:995–1006. 2009. View Article : Google Scholar : PubMed/NCBI

123 

Amirshahrokhi K and Imani M: Edaravone reduces brain injury in hepatic encephalopathy by upregulation of Nrf2/HO-1 and inhibition of NF-κB, iNOS/NO and inflammatory cytokines. Mol Biol Rep. 52:2222025. View Article : Google Scholar : PubMed/NCBI

124 

Mishina M, Komaba Y, Kobayashi S, Kominami S, Fukuchi T, Mizunari T, Teramoto A and Katayama Y: Administration of free radical scavenger edaravone associated with higher frequency of hemorrhagic transformation in patients with cardiogenic embolism. Neurol Med Chir (Tokyo). 48:292–297. 2008. View Article : Google Scholar : PubMed/NCBI

125 

Liao TV, Forehand CC, Hess DC and Fagan SC: Minocycline repurposing in critical illness: Focus on stroke. Curr Top Med Chem. 13:2283–2290. 2013. View Article : Google Scholar : PubMed/NCBI

126 

Singh T, Thapliyal S, Bhatia S, Singh V, Singh M, Singh H, Kumar A and Mishra A: Reconnoitering the transformative journey of minocycline from an antibiotic to an antiepileptic drug. Life Sci. 293:1203462022. View Article : Google Scholar : PubMed/NCBI

127 

Chang JJ, Kim-Tenser M, Emanuel BA, Jones GM, Chapple K, Alikhani A, Sanossian N, Mack WJ, Tsivgoulis G, Alexandrov AV and Pourmotabbed T: Minocycline and matrix metalloproteinase inhibition in acute intracerebral hemorrhage: A pilot study. Eur J Neurol. 24:1384–1391. 2017. View Article : Google Scholar : PubMed/NCBI

128 

Fouda AY, Newsome AS, Spellicy S, Waller JL, Zhi W, Hess DC, Ergul A, Edwards DJ, Fagan SC and Switzer JA: Minocycline in acute cerebral hemorrhage: an early phase randomized trial. Stroke. 48:2885–2887. 2017. View Article : Google Scholar : PubMed/NCBI

129 

Kohler E, Prentice DA, Bates TR, Hankey GJ, Claxton A, van Heerden J and Blacker D: Intravenous minocycline in acute stroke: A randomized, controlled pilot study and meta-analysis. Stroke. 44:2493–2499. 2013. View Article : Google Scholar : PubMed/NCBI

130 

Fagan SC, Waller JL, Nichols FT, Edwards DJ, Pettigrew LC, Clark WM, Hall CE, Switzer JA, Ergul A and Hess DC: Minocycline to improve neurologic outcome in stroke (MINOS): A dose-finding study. Stroke. 41:2283–2287. 2010. View Article : Google Scholar : PubMed/NCBI

131 

Chen X, Chen S, Jiang Y, Zhu C, Wu A, Ma X, Peng F, Ma L, Zhu D, Wang Q and Pi R: Minocycline reduces oxygen-glucose deprivation-induced PC12 cell cytotoxicity via matrix metalloproteinase-9, integrin β1 and phosphorylated Akt modulation. Neurol Sci. 34:1391–1396. 2013. View Article : Google Scholar : PubMed/NCBI

132 

Knecht T, Borlongan C and Dela Peña I: Combination therapy for ischemic stroke: Novel approaches to lengthen therapeutic window of tissue plasminogen activator. Brain Circ. 4:99–108. 2018. View Article : Google Scholar : PubMed/NCBI

133 

Murata Y, Rosell A, Scannevin RH, Rhodes KJ, Wang X and Lo EH: Extension of the thrombolytic time window with minocycline in experimental stroke. Stroke. 39:3372–3377. 2008. View Article : Google Scholar : PubMed/NCBI

134 

Liu Y, Li Z, Khan S, Zhang R, Wei R, Zhang Y, Xue M and Yong VW: Neuroprotection of minocycline by inhibition of extracellular matrix metalloproteinase inducer expression following intracerebral hemorrhage in mice. Neurosci Lett. 764:1362972021. View Article : Google Scholar : PubMed/NCBI

135 

Liu Y, Mu Y, Li Z, Yong VW and Xue M: Extracellular matrix metalloproteinase inducer in brain ischemia and intracerebral hemorrhage. Front Immunol. 13:9864692022. View Article : Google Scholar : PubMed/NCBI

136 

Song ZP, Xiong BR, Guan XH, Cao F, Manyande A, Zhou YQ, Zheng H and Tian YK: Minocycline attenuates bone cancer pain in rats by inhibiting NF-κB in spinal astrocytes. Acta Pharmacol Sin. 37:753–762. 2016. View Article : Google Scholar : PubMed/NCBI

137 

Fan Y, Meng S, Wang Y, Cao J and Wang C: Visfatin/PBEF/Nampt induces EMMPRIN and MMP-9 production in macrophages via the NAMPT-MAPK (p38, ERK1/2)-NF-κB signaling pathway. Int J Mol Med. 27:607–615. 2011.PubMed/NCBI

138 

Chen Y, Won SJ, Xu Y and Swanson RA: Targeting microglial activation in stroke therapy: Pharmacological tools and gender effects. Curr Med Chem. 21:2146–2155. 2014. View Article : Google Scholar : PubMed/NCBI

139 

Kase CS, Furlan AJ, Wechsler LR, Higashida RT, Rowley HA, Hart RG, Molinari GF, Frederick LS, Roberts HC, Gebel JM, et al: Cerebral hemorrhage after intra-arterial thrombolysis for ischemic stroke: the PROACT II trial. Neurology. 57:1603–1610. 2001. View Article : Google Scholar : PubMed/NCBI

140 

Arcambal A, Taïlé J, Rondeau P, Viranaïcken W, Meilhac O and Gonthier MP: Hyperglycemia modulates redox, inflammatory and vasoactive markers through specific signaling pathways in cerebral endothelial cells: Insights on insulin protective action. Free Radic Biol Med. 130:59–70. 2019. View Article : Google Scholar : PubMed/NCBI

141 

Liu Y, Zhang H, Wang S, Guo Y, Fang X, Zheng B, Gao W, Yu H, Chen Z, Roman RJ and Fan F: Reduced pericyte and tight junction coverage in old diabetic rats are associated with hyperglycemia-induced cerebrovascular pericyte dysfunction. Am J Physiolo Heart Circ Physiol. 320:H549–H562. 2021. View Article : Google Scholar : PubMed/NCBI

142 

Rom S, Heldt NA, Gajghate S, Seliga A, Reichenbach NL and Persidsky Y: Hyperglycemia and advanced glycation end products disrupt BBB and promote occludin and claudin-5 protein secretion on extracellular microvesicles. Sci Rep. 10:72742020. View Article : Google Scholar : PubMed/NCBI

143 

Yuan C, Chen S, Ruan Y, Liu Y, Cheng H, Zeng Y, Chen Y, Cheng Q, Huang G, He W and He J: The stress hyperglycemia ratio is associated with hemorrhagic transformation in patients with acute ischemic stroke. Clin Interv Aging. 16:431–442. 2021. View Article : Google Scholar : PubMed/NCBI

144 

Zhang FH, Lin YH, Huang HG, Sun JZ, Wen SQ and Lou M: Rosiglitazone attenuates hyperglycemia-enhanced hemorrhagic transformation after transient focal ischemia in rats. Neuroscience. 250:651–657. 2013. View Article : Google Scholar : PubMed/NCBI

145 

Li Y, Zhu ZY, Lu BW, Huang TT, Zhang YM, Zhou NY, Xuan W, Chen ZA, Wen DX, Yu WF and Li PY: Rosiglitazone ameliorates tissue plasminogen activator-induced brain hemorrhage after stroke. CNS Neurosci Ther. 25:1343–1352. 2019. View Article : Google Scholar : PubMed/NCBI

146 

Marx N, Froehlich J, Siam L, Ittner J, Wierse G, Schmidt A, Scharnagl H, Hombach V and Koenig W: Antidiabetic PPAR gamma-activator rosiglitazone reduces MMP-9 serum levels in type 2 diabetic patients with coronary artery disease. Arterioscler Thromb Vasc Biol. 23:283–288. 2003. View Article : Google Scholar : PubMed/NCBI

147 

Wu G, Wu J, Jiao Y, Wang L, Wang F and Zhang Y: Rosiglitazone infusion therapy following minimally invasive surgery for intracerebral hemorrhage evacuation decreases matrix metalloproteinase-9 and blood-brain barrier disruption in rabbits. BMC Neurol. 15:372015. View Article : Google Scholar : PubMed/NCBI

148 

Wang CX, Ding X, Noor R, Pegg C, He C and Shuaib A: Rosiglitazone alone or in combination with tissue plasminogen activator improves ischemic brain injury in an embolic model in rats. J Cereb Blood Flow Metab. 29:1683–1694. 2009. View Article : Google Scholar : PubMed/NCBI

149 

Zhou M, Xu A, Lam KS, Tam PK, Che CM, Chan L, Lee IK, Wu D and Wang Y: Rosiglitazone promotes fatty acyl CoA accumulation and excessive glycogen storage in livers of mice without adiponectin. J Hepatol. 53:1108–1116. 2010. View Article : Google Scholar : PubMed/NCBI

150 

Medunjanin S, Schleithoff L, Fiegehenn C, Weinert S, Zuschratter W and Braun-Dullaeus RC: GSK-3β controls NF-kappaB activity via IKKγ/NEMO. Sci Rep. 6:385532016. View Article : Google Scholar : PubMed/NCBI

151 

Lee CS, Kwon YW, Yang HM, Kim SH, Kim TY, Hur J, Park KW, Cho HJ, Kang HJ, Park YB and Kim HS: New mechanism of rosiglitazone to reduce neointimal hyperplasia: Activation of glycogen synthase kinase-3beta followed by inhibition of MMP-9. Arterioscler Thromb Vasc Biol. 29:472–479. 2009. View Article : Google Scholar : PubMed/NCBI

152 

Home PD, Pocock SJ, Beck-Nielsen H, Curtis PS, Gomis R, Hanefeld M, Jones NP, Komajda M and McMurray JJ; RECORD Study Team, : Rosiglitazone evaluated for cardiovascular outcomes in oral agent combination therapy for type 2 diabetes (RECORD): A multicentre, randomised, open-label trial. Lancet. 373:2125–2135. 2009. View Article : Google Scholar : PubMed/NCBI

153 

Nishiyama Y, Kimura K, Otsuka T, Toyoda K, Uchiyama S, Hoshino H, Sakai N, Okada Y, Origasa H, Naritomi H, et al: Dual antiplatelet therapy with cilostazol for secondary prevention in lacunar stroke: Subanalysis of the CSPS.com trial. Stroke. 54:697–705. 2023. View Article : Google Scholar : PubMed/NCBI

154 

Uchiyama S: Results of the Cilostazol Stroke Prevention Study II (CSPS II): A randomized controlled trial for the comparison of cilostazol and aspirin in stroke patients. Rinsho Shinkeigaku. 50:832–834. 2010.(In Japanese). View Article : Google Scholar : PubMed/NCBI

155 

Nonaka Y, Tsuruma K, Shimazawa M, Yoshimura S, Iwama T and Hara H: Cilostazol protects against hemorrhagic transformation in mice transient focal cerebral ischemia-induced brain damage. Neurosci Lett. 452:156–161. 2009. View Article : Google Scholar : PubMed/NCBI

156 

Hase Y, Okamoto Y, Fujita Y, Kitamura A, Nakabayashi H, Ito H, Maki T, Washida K, Takahashi R and Ihara M: Cilostazol, a phosphodiesterase inhibitor, prevents no-reflow and hemorrhage in mice with focal cerebral ischemia. Exp Neurol. 233:523–533. 2012. View Article : Google Scholar : PubMed/NCBI

157 

Kasahara Y, Nakagomi T, Matsuyama T, Stern D and Taguchi A: Cilostazol reduces the risk of hemorrhagic infarction after administration of tissue-type plasminogen activator in a murine stroke model. Stroke. 43:499–506. 2012. View Article : Google Scholar : PubMed/NCBI

158 

Chuang SY, Yang SH, Chen TY and Pang JH: Cilostazol inhibits matrix invasion and modulates the gene expressions of MMP-9 and TIMP-1 in PMA-differentiated THP-1 cells. Eur J Pharmacol. 670:419–426. 2011. View Article : Google Scholar : PubMed/NCBI

159 

da Motta NA and de Brito FC: Cilostazol exerts antiplatelet and anti-inflammatory effects through AMPK activation and NF-kB inhibition on hypercholesterolemic rats. Fundam Clin Pharmacol. 30:327–337. 2016. View Article : Google Scholar : PubMed/NCBI

160 

Kitashoji A, Egashira Y, Mishiro K, Suzuki Y, Ito H, Tsuruma K, Shimazawa M and Hara H: Cilostazol ameliorates warfarin-induced hemorrhagic transformation after cerebral ischemia in mice. Stroke. 44:2862–2868. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2025
Volume 32 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Guo P, Li H, Zhang X, Liu Y, Xue S, Yong VW and Xue M: Matrix metalloproteinase‑9 in hemorrhagic transformation after acute ischemic stroke (Review). Mol Med Rep 32: 225, 2025.
APA
Guo, P., Li, H., Zhang, X., Liu, Y., Xue, S., Yong, V.W., & Xue, M. (2025). Matrix metalloproteinase‑9 in hemorrhagic transformation after acute ischemic stroke (Review). Molecular Medicine Reports, 32, 225. https://doi.org/10.3892/mmr.2025.13590
MLA
Guo, P., Li, H., Zhang, X., Liu, Y., Xue, S., Yong, V. W., Xue, M."Matrix metalloproteinase‑9 in hemorrhagic transformation after acute ischemic stroke (Review)". Molecular Medicine Reports 32.2 (2025): 225.
Chicago
Guo, P., Li, H., Zhang, X., Liu, Y., Xue, S., Yong, V. W., Xue, M."Matrix metalloproteinase‑9 in hemorrhagic transformation after acute ischemic stroke (Review)". Molecular Medicine Reports 32, no. 2 (2025): 225. https://doi.org/10.3892/mmr.2025.13590