Skip to main content
Journal of Cellular and Molecular Medicine logoLink to Journal of Cellular and Molecular Medicine
. 2020 Jul 13;24(16):8918–8929. doi: 10.1111/jcmm.15641

Osteopontin as a candidate of therapeutic application for the acute brain injury

Yunxiang Zhou 1, Yihan Yao 1, Lesang Sheng 1, Jianmin Zhang 2,3,4, John H Zhang 5,6, Anwen Shao 2,
PMCID: PMC7417697  PMID: 32657030

Abstract

Acute brain injury is the leading cause of human death and disability worldwide, which includes intracerebral haemorrhage, subarachnoid haemorrhage, cerebral ischaemia, traumatic brain injury and hypoxia‐ischaemia brain injury. Currently, clinical treatments for neurological dysfunction of acute brain injury have not been satisfactory. Osteopontin (OPN) is a complex adhesion protein and cytokine that interacts with multiple receptors including integrins and CD44 variants, exhibiting mostly neuroprotective roles and showing therapeutic potential for acute brain injury. OPN‐induced tissue remodelling and functional repair mainly rely on its positive roles in the coordination of pro‐inflammatory and anti‐inflammatory responses, blood‐brain barrier maintenance and anti‐apoptotic actions, as well as other mechanisms such as affecting the chemotaxis and proliferation of nerve cells. The blood OPN strongly parallel with the OPN induced in the brain and can be used as a novel biomarker of the susceptibility, severity and outcome of acute brain injury. In the present review, we summarized the molecular signalling mechanisms of OPN as well as its overall role in different kinds of acute brain injury.

Keywords: apoptosis, intracerebral haemorrhage, neuroprotection, osteopontin, stroke, subarachnoid haemorrhage, traumatic brain injury

1. INTRODUCTION

Acute brain injury, exemplified by stroke, traumatic brain injury (TBI) and hypoxia‐ischaemia brain injury, is the leading cause of human death and disability worldwide. 1 , 2 , 3 , 4 , 5 Stroke, which represents the primary reason for permanent disability in adults, can be divided into two types: ischaemic stroke, typically occurring in the setting of atherothrombosis, and haemorrhagic stroke, mainly due to the rupture of cerebral arteries. 6 , 7 , 8 The latter further consists of subarachnoid haemorrhage (SAH) and intracerebral haemorrhage (ICH) and accounts for approximately 10%‐20% of strokes yet has higher mortality vs the former. 9 , 10 , 11 , 12 , 13 TBI refers to sudden damage caused by mechanical force, occurring in traffic accidents, blast, wars, violence, terrorism, falls and sporting activity. 14 TBI is currently the major source of fatality in young adults, with an annual global economic loss of approximately US$ 400 billion. 1 , 2 , 14 , 15 , 16 Hypoxic‐ischaemic brain injury is another frequent, fatal and crippling neurologic disease, particularly perinatal hypoxia‐ischaemia remains the dominating cause of acute brain injury in the neonate. 17 , 18 , 19 These acute brain injuries impose a heavy socio‐economic burden, whereas effective therapies are still scarce. Notably, acute neurologic disorders share many common features and processes within the pathophysiology. 20 Although pathogenic mechanisms involved in acute brain injury have been studied extensively, which include cellular apoptosis, neuroinflammation, blood‐brain barrier (BBB) disruption, 21 the prognosis of patients remains poor under current therapeutic strategies. 1 New treatments targeting acute brain injury are urgently needed.

Osteopontin (OPN), a highly phosphorylated glycoprotein, is a complex adhesion protein and cytokine that interacts with multiple receptors including integrins and CD44 variants. 22 OPN has been found in various tissues, including the brain, and plays an important role in cellular processes such as adhesion, motility and survival. 23 Altered expression patterns of OPN have been observed in pathological conditions such as multiple sclerosis, atherosclerosis, myocardial infarction and cancers. 24 , 25 Under normal conditions, OPN expression is weak in the brain, while under pathological conditions including Alzheimer's disease, Parkinson's disease, TBI, stroke and hypoxia‐ischaemia brain injury, it is significantly increased in macrophages/microglia and astrocytes and exerts neuroprotective effects. 26 , 27 , 28 , 29 , 30 In this review, we will highlight the molecular signalling pathways involved in neuroprotective part of OPN as well as its value as a potential therapeutic target, biomarker and predictor; we will also discuss the potential reason why exogenous OPN is not effective in some experimental models and put forth the limitations of current OPN research.

2. GENERAL FEATURES OF OPN

Osteopontin is a highly phosphorylated extracellular matrix glycoprotein that is rich in aspartic acid and has acidic characteristics consisting of approximately 314 amino acids with a molecular weight ranging between 44 and 75 kD. 31 , 32 OPN is initially found in osteoblasts and is later independently identified as secreted phosphoprotein 1 associated with neoplastic transformation and early T lymphocyte activation 1. 33 , 34 , 35 The multiplicity of functions ascribed to OPN may reflect the presence of various isoforms, post‐translational modifications, proteolytic processing, and diversity of cell types that OPN can interact with. 32 , 36

OPN gene is located in the small integrin‐binding ligand, N‐linked glycoproteins (SIBLING) cluster on chromosome 4 (4q13) in the human genome and on mouse chromosome 5. 37 The gene contains seven exons, six of which are translated in the full‐length isoform OPN‐a. 38 Alternative translation and splicing result in two splice variants with deletion of exon 5 (OPN‐b) or deletion of exon 4 (OPN‐c), which correlates with cancer progression and poor prognosis. 38 , 39 Besides important isoforms, OPN is subject to significant post‐translational modifications, including phosphorylation, sulfation, glycosylation and transglutamination, and regulation of these modifications represents the potential to control OPN function. 32 , 36

When cleaved by thrombin, OPN transforms into two types, the N‐terminal fragment (trOPN‐N) and C‐terminal fragment (trOPN‐C). 40 TrOPN‐N contains several highly conserved cell adhesive motifs including an Arg‐Gly‐Asp (RGD) sequence which binds to integrins such as αvβ1, αvβ3, αvβ5, α8β1 and α5β1, as well as a cryptic Ser‐Val‐Val‐Tyr‐Gly‐Leu‐Arg (SVVYGLR)‐containing domain that is only exposed after thrombin cleavage and binds to α4β1, α9β1 and α4β7 integrins, 22 whereas trOPN‐C binds to CD44 variants. 41 Moreover, mouse OPN cleaved by matrix metalloproteinase (MMP) 3/7 produces LRSKSRSFQVSDEQY, a novel α9β1 integrin‐binding motif in the C‐terminal fragment. 41 In the human bone marrow, trOPN‐N constitutes the predominant form and acts as a chemotactic factor promoting hematopoietic progenitor cell homing as well as T cell‐derived IFN‐γ secretion through its binding to α9β1 and α4β1 integrins. 42 In T cells, trOPN‐N upregulates IL‐17, whereas OPN‐C induces IL‐10 downregulation by selectively interacting with CD44 isoforms. 43 In carotid specimens, inflammation severity is only associated with trOPN‐N expression, not with full‐length OPN or trOPN‐C. 44 Thus, different terminal fragments may perform different functions.

3. THE ROLES OF OSTEOPONTIN IN ACUTE BRAIN INJURIES AND OTHER DISEASES

Osteopontin can be secreted by multiple tissues as well as body fluids and serves as a regulator in various biological mechanisms including osteoclast function, wound healing, cell migration, immune response, insulin resistance and cellular processes. 24 , 45 , 46 Expression levels of OPN vary in different cell types; however, the mechanism underlying the release of OPN is not yet fully understood. 23 In the central nervous system, OPN expression is weak under normal conditions and can be distinctly upregulated in response to either injuries or inflammation. 47 Accumulating evidence has demonstrated that OPN plays a significant role in neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease and multiple sclerosis, 26 , 27 , 48 as well as acute brain injury including TBI, stroke, and hypoxia‐ischaemia brain injury. 49 , 50 , 51 , 52

Different acute brain injury models share many common features and processes within the pathophysiology. To date, most studies suggest a neuroprotective role of OPN in these neurological diseases. 49 , 53 , 54 , 55 , 56 , 57 , 58 , 59 , 60 , 61 Furthermore, numerous studies have demonstrated that intracerebroventricular injection or intranasal administration of exogenous OPN possesses neuroprotective roles and improves neurological outcome following ischaemic stroke and haemorrhagic stroke 53 , 54 , 55 , 56 , 57 , 58 (Table 1). As regards TBI, although studies have reported endogenous OPN positively influenced synapse reorganization and improved functional recovery, 49 , 59 the study by Jullienne et al 62 showed exogenous administration of OPN treatment following TBI did not alter lesion characteristics. Moreover, inconsistent therapeutic effects were found in the experimental neonatal hypoxia‐ischaemia stroke treated with exogenous OPN. 60 , 61 , 63 , 64

Table 1.

The main findings of OPN‐relevant therapeutic potential in acute brain injuries

Disease Rat models Agents and methods Main findings References
Ischaemic stroke Male SD rats, 250‐300 g, MACO R‐OPN, injected stereotaxically into the striatum, 1 h post‐MACO, 100 ng Reduced the mean infarct volume, extended the therapeutic window at least to 12 h post‐MACO Jin et al 58
Male SD rats, 250‐300 g, MACO OPN peptide, intranasally, 100 ng Reduced mean infarct volume, ameliorated neurological deficits, suppressed the induction of iNOS; the RGD motif in OPN peptide and endogenous αvβ3 integrin are essential for the neuroprotective effects Jin et al 50
Male SD rats, 240‐280 g, MACO Hyperbaric oxygen preconditioning Improved neurological outcome, promoted expression of OPN, reduced the expression of IL‐1β/NFκB and augmented Akt phosphorylation Hu et al 93
Male C57Bl/6 mice, 8 to 10 wk, transient MACO R‐OPN, intracerebroventricularly, immediately before and immediately after surgery, 50 ng Reduced infarct size, increased phosphorylation of Akt and p42/p44 MAPK Meller et al 53
Hypoxia‐ischaemia brain injury 7‐day‐old rat pups, unilateral ligation of the RCA followed by hypoxia R‐OPN, intracerebroventricularly, 1 h post‐HI, 0.03 μg and 0.1 μg Reduced apoptotic cell, cleaved caspase‐3 and infarct volume, ameliorated bodyweight loss, improved long‐term neurological impairment Chen et al 61
Postnatal day 10 SD rat pups, unilateral ligation of the RCA followed by hypoxia R‐OPN, intranasally, 1 h post‐HI, 5 μg Attenuated BBB permeability and brain oedema Dixon et al 60
Postnatal day 9 C57BL/6 mice pups, permanent occlusion of the RCA followed by hypoxia R‐OPN peptide, intranasally (350 or 2100 ng), intraperitoneally (10 mg/kg) or intracerebrally (100 ng), at several points in time Did not exert neuroprotective effects Bonestroo et al 63
Postnatal day 5 C57BL/6J mice pups, ligation of the LCA followed by hypoxia Full‐length OPN protein and thrombin‐cleaved OPN, intranasally (1.2 μg, immediately before and after HI) and intracerebroventricularly (5 μg, immediately before HI) Did not exert neuroprotective effects Albertsson et al 64
SAH Male SD rats, 300‐350 g, endovascular perforation model Vitamin D3, intranasally Attenuated BBB disruption through endogenous upregulation of OPN and subsequent CD44 and P‐gp glycosylation signals in brain endothelial cells Enkhjargal et al 127
Male SD rats, 300‐370 g, endovascular perforation model R‐OPN, intracerebroventricularly, 1 h before surgery, 0.1 μg Improved BBB disruption, increased MAPK phosphatase‐1, inactivated MAPK, reduced vascular endothelial growth factor‐A Suzuki et al 125
Male SD rats, 280‐320 g, endovascular perforation model R‐OPN, intranasally, minutes post‐SAH, 5 μg Improved neuronal cell survival, brain oedema and neurological scores, increased p‐FAK and p‐Akt expressions, decreased caspase‐3 cleavage Topkoru et al 55
Male SD rats, 250‐300 g, endovascular perforation model Ephedra sinica extract, orally, immediately after the surgery, 15 mg/kg Upregulated osteopontin signal, reduced the expressions of MMP‐9, alleviated the BBB disruption, improved neurological functions Zuo et al 132
Male SD rats, 300‐370 g, endovascular perforation model R‐OPN, intracerebroventricularly, 1 h pre‐SAH, 0.02 and 0.1 g Ameliorated bodyweight loss, neurologic impairment, brain oedema and BBB disruption, inhibited NFκB and MMP‐9, maintained MMP‐1 and ZO‐1 Suzuki et al 56
Male SD rats, 300‐370 g, endovascular perforation model R‐OPN, Intracerebroventricularly, 1 hour pre‐surgery or 5 hours post‐surgery, 0.01 μg, 0.02 μg or 0.1 μg Prevented vasospasm, improved neurological impairments, inhibited MAPKs, caldesmon and HSP 27 Suzuki et al 148
Male SD rats, 300‐350 g, endovascular perforation model R‐OPN, intranasally (1 h, 3 h, 6 h post‐SAH, 5 μg) and intracerebroventricularly (3 h post‐SAH, 0.1 μg) Stabilized the phenotype of vascular smooth muscle, dilated cerebral arteries, improved neurological outcome Wu et al 57
Male SD rats, 300‐375 g, the double injection model R‐OPN, intracerebroventricularly, nearly 30 min after the first SAH, 0.3 μg or 0.1 μg Improved neurological scores vasospasm, reduced cleaved caspase‐3, Bax and apoptosis, increased p‐Akt and Bcl‐2 He et al 109
ICH Male SD rats, 280‐320 g, a collagenase model R‐OPN, intranasally, 1 h after ICH, 1μg, 3 μg or 9 μg Attenuated brain inflammation and brain oedema improved neurological functions via integrin‐β1 induced inhibition of JAK2/STAT1 pathway Gong et al 91
CD‐1 mice, 30‐40 g, a collagenase model R‐OPN, intracerebroventricularly, 20 min pre‐ICH, 10 ng, 50 ng, or 100 ng Improved neurological scores and brain water content Wu et al 86

Male SD rats, 280‐320 g, injection of autologous blood into the right

basal ganglia

R‐OPN, intracerebroventricularly, 1 h post‐ICH, 0.1 μg Reduced neurological deficits, rotarod latencies and brain water content, increased p‐Akt expression and decreased p‐GSK‐3β, Bax/Bcl‐2 ratio and cleaved caspase‐3 Zhang et al 99
TBI Male SD rats, 275‐375 g (9 to 12 wks old), moderate‐to‐severe controlled cortical impact R‐OPN, intranasally, 1 h post‐TBI, 5 μg Did not improve neurological score, lesion volumes, BBB dysfunction, or vascular characteristics; increased the microglial and HO‐1 response Jullienne et al 62
Healthy rats Male SD rats, 270‐320 g R‐TNC, r‐OPN, or both were injected into a cisterna magna R‐OPN had no effect on the vessel diameter but could reverse prolonged contractions of rat basilar arteries induced by r‐TNC Suzuki et al 146

Abbreviations: BBB, blood‐brain barrier; FAK, focal adhesion kinase; GSK‐3β, glycogen synthase kinase 3 beta; HO‐1, haem oxygenase 1; ICH, intracerebral haemorrhage; IL, interleukin; JAK2/STAT1, Janus kinase/signal transducers and activators of transcription 1; LCA, left carotid artery; MACO, middle cerebral artery occlusion; MAPK, mitogen‐activated protein kinase; MMP, matrix metalloproteinase; NFκB, factor‐κ‐gene binding; OPN, osteopontin; RCA, right carotid artery; RGD, Arg‐Gly‐Asp; r‐OPN, recombinant; SAH, subarachnoid haemorrhage; TNC, tenascin‐C; ZO‐1, zona occludens‐1

The aforementioned experimental hypoxia‐ischaemia strokes were performed on neonatal mice or neonatal rats. 60 , 61 , 63 , 64 Accordingly, an age‐dependent manner of the neuroprotective effects of exogenous OPN has been introduced, in which OPN potentiates injury in the neonatal brain while resisting injury in the adult brain due to the different integrin subunit expression levels and distribution at different neuronal development stages 64 Additionally, the above models of the neuroprotective group 60 , 61 and the non‐neuroprotective group 63 , 64 were established on rat pups and mice pups, respectively, and consistently, the animal heterogeneity of OPN efficacy has been proposed. 62 Jullienne et al 62 were the first to test the function of exogenous OPN after experimental TBI on adult male SD rats (similar ages to the models in cited experimental stroke researches); although several potential beneficial alterations were observed, exogenous OPN did not substantially improve lesion characteristics. It is worthy of note that the protective effects of OPN may be overridden by the hyperacute cerebral injury but sufficient to attenuate delayed thalamic neurodegeneration. 65

Indeed, the regulation and function of OPN may be specific to each pathophysiological condition and may be context‐dependent, spatiotemporal‐dependent or cell‐dependent, which perhaps explain the conflicting results regarding the efficacy of exogenous OPN in distinct disorders. 25 , 27 , 62 , 63 , 64 , 65 , 67 , 68 , 69 Notably, OPN is proposed to play a dichotomous role in the neuroinflammation 70 , 71 (see Section 4.1 for details) and can propel the progress of various autoimmune diseases such as multiple sclerosis. 25 , 67 Thus, the ability to maintain the balance of anti‐inflammatory and pro‐inflammatory responses and to coordinate these signals with other inputs received by the cell (eg T cell‐independent neurodegeneration in ischaemic brain injury vs T cell‐mediated aggressive signals in multiple sclerosis contribute to the altered efficacy of OPN 65 ) may define the ultimate role of OPN. Crucially, as mentioned above, different isoforms, different terminal fragments, various post‐translational modifications of OPN may yield different impact on acute brain injury. 32 , 43 , 44

4. THE CANDIDATE MECHANISMS OF THE NEUROPROTECTIVE EFFECTS OF OSTEOPONTIN IN ACUTE BRAIN INJURY

The pleiotropic effects of OPN in acute brain injury are reflected in its multifunctional regulation of various physiological processes, such as inflammation, apoptosis, BBB reconstruction and neurogenesis, and some of the mechanisms are interrelated and overlapping. In the present section, we will summarize current research findings concerning the OPN signalling in acute brain injury and integrate knowledge about its underlying mechanisms of neuroprotection and therapeutic potential. Some potential mechanisms for its beneficial effects are summarized in Figure 1, and promoting these signalling pathways may elicit a neuroprotective role of OPN.

Figure 1.

Figure 1

Some neuroprotective signalling pathways induced by osteopontin following acute brain injury. OPN exerts multiple roles in acute brain injury via interaction with integrins and CD44, which is reflected in its multifunctional regulation of various physiological processes, such as inflammation, apoptosis and BBB reconstruction. The mechanisms in these processes are interrelated and overlapping. AK2/STAT1, Janus kinase/signal transducers and activators of transcription 1; Ang‐1, angiopoietin‐1; BBB, blood‐brain barrier; ERK, extracellular signal‐regulated kinase; FAK, focal adhesion kinase; GSK‐3β, glycogen synthase kinase 3 beta; IL, interleukin; iNOS, inducible nitric oxide synthase; JNK, c‐Jun N‐terminal kinase; MAPK, mitogen‐activated protein kinase; MKP‐1, MAPK phosphatase‐1; MMP, matrix metalloproteinase; NFκB, factor‐κ‐gene binding; P‐gp, P‐glucoprotein; Rac‐1, Ras‐related C3 botulinum toxin substrate 1; ROS, reactive oxygen species; TIMP‐1, tissue Inhibitor of MMP‐1; VEGF, vascular endothelial growth factor

4.1. OPN and neuroinflammation

Inflammation plays a crucial role in the pathogenesis of acute brain injury. 9 , 16 , 72 , 73 Potentially exacerbating secondary brain injury via inflammatory cascade in the acute stage, whereas beneficially promoting tissue remodelling and functional repair, inflammatory response induced by acute brain injury is suggested to be a double‐edged sword. 74 By early inhibition of inflammatory cascade, the coordination of pro‐inflammatory and anti‐inflammatory responses leads to the alleviation of the brain injury and better patient outcome. 75 Interestingly, OPN is also indicated to exert dual roles in neuroinflammation. 70 Many studies have highlighted the pro‐inflammatory role of OPN in the pathogenesis of various autoimmune diseases, such as multiple sclerosis. 25 , 65 , 67 , 76 During acute brain injury, although some researchers have reported that OPN exacerbates cerebral injury via neuroinflammation, 71 OPN is also considered a promising target for anti‐inflammation. 58 , 65 , 77 Thus, considering the important role OPN plays in the initiation of inflammation 70 and its overall neuroprotective role, 49 , 53 , 54 , 55 , 56 , 57 , 58 , 59 , 60 , 61 OPN may maintain inflammation homeostasis with a negative feedback mechanism.

An important role of OPN in the inflammatory response is to trigger various leucocytes to cause a functional response and induce cytokine secretion, thereby forming an entire immune response. 78 Activated microglia/macrophages and astrocytes are the main cellular sources of OPN induction in the central nervous system. 27 Kang et al 79 showed that microglia/macrophages and astrocytes induced OPN upregulation at different stages after brain injury. And then, the expression of OPN inversely recruited, activated and polarized additional microglial/macrophages and astrocytes in the lesional and perilesional area, which was based on the interaction of OPN with αvβ3 integrin and/or interaction with CD44. 79 , 80 , 81 , 82 , 83 These receptors can also be upregulated after brain injury. 52 The activated macrophages/microglia and astrocytes, as well as the induced OPN and its receptors, contribute to the succedent cytokine secretion, removal of the necrotic tissue, extracellular matrix formation, tissue remodelling, angiogenesis and gliosis. 52 , 80 , 84 , 85

Inducible nitric oxide synthase (iNOS) exerts vital roles in inflammation response and is identified to be excitotoxic and neurotoxic. 77 Moreover, iNOS‐derived nitric oxide is known to activate MMP‐9, which is involved in neuroinflammation, cell death and the BBB disruption. 86 , 87 , 88 Previous studies have demonstrated that OPN is involved in iNOS pathway. 50 , 89 , 90 Induced OPN provides a dose‐dependent pattern suppressing iNOS expression after acute brain injury 65 , 80 by increasing expression of integrin‐β1 and then inhibiting the Janus kinase/signal transducers and activators of transcription 1 (JAK/STAT1) pathway, which is closely linked to iNOS expression. 86 , 91 Ladwig et al 81 suggested that OPN downregulates iNOS by shifting the microglia phenotype towards an M2 to reduce iNOS‐expressing M1 cells. In addition to iNOS pathway, OPN also downregulates MMP‐9 expression by inhibiting interleukin (IL)‐1β/nuclear factor‐κ‐gene binding (NFκB) pathway, through which IL‐1β activates NFκB and subsequently mediates the induction of MMP‐9. 56 , 89 , 92 , 93

4.2. OPN and apoptosis

Apoptosis is a highly complex energy‐dependent programmed cell death process closely in correlation with neuron development and homeostasis under normal physiological conditions. 94 , 95 Nevertheless, apoptosis also participates in the process of neurological disorders under pathological conditions and is considered as a key player for the progression and prognosis of patients. 96 , 97 , 98 Previous studies have suggested that OPN ameliorates apoptosis and promotes cell survival during brain injury through several molecular signalling pathways, which may suggest a potential therapeutic strategy for treating acute brain injury. 61 , 99 , 100 And accumulating studies indicate that OPN exerts direct anti‐apoptotic actions via interaction with integrins or CD44. 53 , 65 , 101 , 102

Among all the proteins involved in the initiation and execution of apoptosis, the caspases stand out as being crucial mediators of this process. 103 , 104 , 105 Furthermore, of all the caspases, caspase‐3 is probably the best understood and required for a large portion of distinct apoptotic processes and cell deaths. 104 , 106 , 107 And not until caspase‐3 zymogen is cleaved by an initiator caspase following the initiation of apoptosis signal does it have activity. 106 Thus, suppression of caspase‐3 cleavage may be effective to inhibit neuronal apoptosis. 55 , 77 , 108 Consistently, experimental studies focusing on stroke and hypoxia‐ischaemia neonatal brain injury found a remarkable reduction of caspase‐3 cleavage following the OPN administration and suggested that OPN‐induced neuroprotection was associated with the inhibition of caspase‐3 activity. 61 However, the underlying mechanisms of OPN‐induced caspase‐3 inhibition remain not entirely clear. Topkoru et al performed nasal administration of recombinant OPN 30 minutes after SAH induction on rat models, and they not only found a robust decrease of caspase‐3 cleavage but also a notable increase of phosphorylated focal adhesion kinase (FAK) and phosphorylated Akt, in line with the decline of brain oedema and improvement of neuronal cell survival and neurological status. 55 Similarly, Zhang et al 99 intracerebroventricularly injected recombinant OPN 1 h after ICH induced on rat models and found increased phosphorylated Akt expression and decreased phosphorylated glycogen synthase kinase 3 beta (GSK‐3β), Bax/Bcl‐2 ratio and cleaved caspase‐3, consistent with attenuated brain water content and cell death. Moreover, Meller et al and He et al also found a reduction of cleaved caspase‐3 accompanied by increased phosphorylated Akt expression and a better neurological function. 53 , 109

The PI3K/Akt signalling pathway is involved in numerous cellular processes, including apoptosis, BBB disruption, neurogenesis and angiogenesis. 60 , 109 , 110 Inducing the phosphorylation of FAK via integrin receptor or CD44 receptor, OPN subsequently activates the PI3K/Akt signalling pathway. 60 , 99 , 111 Then, phosphorylated Akt suppresses the pro‐apoptotic proteins including Bax and Bad and upregulates the anti‐apoptotic proteins such as Bcl‐2. 99 , 109 These molecules play important roles in the caspase‐dependent pathway, in which caspase‐3 is considered to be a common downstream protein, and the regulation of phosphorylated Akt results in the suppression of caspase‐3 and apoptosis. 61 , 112 In addition, phosphorylated Akt induces the phosphorylation and subsequent inactivation of GSK‐3β, which is also pro‐apoptotic and has been found to aggravate brain injury in experimental ICH, ischaemic stroke and traumatic brain injury. 113 , 114 , 115 Additionally, inhibitors of FAK and PI3K administrated to the rat models could abolish the protective effects of OPN. 55 , 99 Thus, the PI3K/Akt pathway may play a critical role in the OPN‐induced anti‐apoptotic actions.

Besides, some proteins involved in the process of apoptosis after acute brain injury such as FAK, Bcl‐2, PI3K and Akt may overlap with autophagic pathways. 116 , 117 Emerging studies suggest that OPN enhances autophagy and reduces apoptosis after experimental SAH through FAK signalling, resulting in the attenuation of early brain injury and improvement of long‐term outcome. 51

4.3. OPN and BBB disruption

The BBB is built up by specialized monolayer endothelial cells which are connected by tight junctions without fenestrations. 118 The endothelial barrier is also supplemented with a large number of pericytes, which share the common basal membrane with the endothelial cells. 118 Besides, the abluminal surface of the microvascular basement membranes is covered by astrocytic perivascular end‐feet. 119 This major barrier plays a significant role in maintaining brain haemostasis and protecting the brain from disease and injury. Dysfunction of the BBB has been described as one of the independent risk factors for poor prognosis after acute brain injury, which may amplify inflammation and lead to further parenchyma damage and oedema by allowing more blood‐borne cells and substances to flow into the brain parenchyma. 120 , 121 , 122 , 123 Previous studies have demonstrated that OPN is significantly induced and locates at reactive capillary endothelial cells and astrocytes during the recovery of BBB function, 124 , 125 , 126 suggesting an important role for OPN in BBB reconstruction. And still, OPN exerts effects of BBB maintenance via interaction with integrins and CD44 receptors. 127

As mentioned above, MMP‐9 is involved in aggravation of BBB disruption in addition to facilitating the inflammatory cascade and cell death. 86 , 87 , 88 The balanced interaction between MMP‐9 and its corresponding inhibitor, tissue inhibitor of metalloproteinase‐1 (TIMP‐1), determines the severity of BBB disruption. 128 Induced by acute brain injury, oxidative stress and IL‐1β then activate NF‐κB, which directly upregulates MMP‐9 and inhibits TIMP‐1 levels in the brain. 56 , 129 There is plenty of evidence that MMP‐9 plays a role in degrading the extracellular matrix of cerebral microvessel basal lamina including laminin, fibronectin, collagen IV and zona occludens‐1 (ZO‐1) and causing BBB disruption. 130 , 131 , 132 ZO‐1 belongs to endothelial tight junction‐related proteins, loss of which will increase BBB permeability. 133 The inhibition of IL‐1β/NFκB pathway and reduction of oxidative stress have been widely reported by exogenous OPN administration, through which OPN downregulates MMP‐9 expression and upregulates TIMP‐1 to protect cerebral microvessel basal lamina and subsequently maintain BBB integrity. 56 , 89 , 92 , 93 Furthermore, the FAK/PI3K signalling pathway is also involved in BBB dysfunction following experimental brain injury. 60 It has been demonstrated that exogenous OPN promotes the phosphorylation of FAK and subsequently activates the PI3K, 60 , 99 , 111 leading to the induction of Ras‐related C3 botulinum toxin substrate 1 (Rac‐1) expression and the preservation of BBB integrity. 60 , 134

Interestingly, endogenous OPN induced after acute brain injury may ameliorate BBB disruption through different mechanisms. Mitogen‐activated protein kinase (MAPK) can influence the expression of vascular endothelial growth factor (VEGF)‐A, which is a potent inducer of vascular permeability. 135 , 136 Endogenous OPN induction after acute brain injury keeps angiopoietin (Ang)‐1 levels within the normal range, which inhibits the effect of VEGF‐A with a robust anti‐permeability property. 137 And blockage of endogenous OPN reduces Ang‐1 and MAPK phosphatase‐1, an endogenous MAPKs inhibitor, then activates MAPKs including p38, c‐Jun N‐terminal kinase, and extracellular signal‐regulated kinase 1/2, resulting in the induction of VEGF‐A and the exacerbation of BBB permeability. 125 In addition, endogenous OPN has been shown to induce reactive astrocyte polarization, which is pivotal to the complete neovessel coverage by astrocyte end‐feet and the integrity of BBB. 138 Moreover, Enkhjargal et al 127 demonstrated that brain OPN protected BBB against disruption via the CD44/P‐glucoprotein glycosylation pathway in the vascular endothelial cells.

4.4. Additional aspects of OPN

Previous studies also revealed that OPN exerted positive roles in the proliferation, survival and differentiation of various cells, including cerebrovascular smooth muscle cells, 138 , 139 , 140 neural progenitor cells 119 , 141 and neural stem cells. 142 Moreover, adhesive and chemotactic properties of OPN exert function in the lateral migration of neuroblasts from the subventricular zone to the injured region following focal cerebral ischaemia and ICH. 143 , 144 OPN also reportedly enhances the sensitivity of adult corticospinal neurons to insulin‐like growth factor 1, 145 ameliorates cerebral vasospasm 109 , 146 , 147 , 148 and stabilizes smooth muscle cell phenotype 57 following acute brain injury.

5. OSTEOPONTIN AS A BIOMARKER OF ACUTE BRAIN INJURY

Considering the upregulation and multifarious effects of OPN following acute brain injury, it may reflect the occurrence and severity of neurological damage. 149 And the OPN induced in the brain strongly parallels increased OPN protein in the blood, which may owe to the leakage of OPN produced in the brain or at the interface between brain and blood into the circulation. 150 , 151 , 152 Thus, the plasma/serum OPN can be used as a novel biomarker of the susceptibility, severity and outcome of acute brain injury. 149 , 153 Nakatsuka et al 124 demonstrated that plasma OPN levels ≥ 915.9 pmol/L at days 10‐12 was the most useful predictor of poor outcome and that plasma OPN levels ≥ 955.1 pmol/L at days 1‐3 was an independent predictor of poor outcome after SAH. Li et al 151 suggested that plasma OPN levels at 48 h after hypoxic‐ischaemic encephalopathy were accordant with the severity of brain damage at day 7 recovery. Carbone et al 154 showed serum OPN levels (cut‐off value, 30.53 ng/mL) were the best predictor of poor outcome at day 90 after ischaemic stroke, which is analysed by receiver operating characteristic curve, while Jing et al showed the thrombin‐cleaved OPN levels were significantly correlated with the clinical outcome 12 months after hospital discharge and could discriminate ischaemic stroke patients from healthy individuals at a cut‐off of 166.8 ng/mL. 155 Plasma OPN levels also predict the atherosclerotic plaque destabilization. 140 , 156 , 157 Ozaki et al 40 revealed that plasma thrombin‐cleaved OPN N‐terminal levels of >5.47 pmol/L were independent predictors of atherothrombosis even within 3 h from stroke onset, thus exhibiting the early diagnostic value. The sensitivity and specificity of these cut‐off values are shown in Table 2.

Table 2.

Cut‐off values of osteopontin as a biomarker of acute brain injuries

Study object Osteopontin Cut‐off values Time‐point after stroke Clinical value Sensitivity (%) Specificity (%) References
Aneurysmal subarachnoid haemorrhage, 109 patients Blood full‐length OPN 915.9 pmol/L Days 10‐12 The most useful predictor of poor outcome 69.4 84.5 Nakatsuka et al 124
955.1 pmol/L Days 1‐3 An independent predictor of poor outcome 65.8 70.4
Ischaemic stroke, 90 patients Blood full‐length OPN 30.53 ng/mL Day 90 The best predictor of modified rankin score > 2 92 46 Carbone et al 154
Ischaemic stroke, 377 patients; 511 healthy individuals Blood thrombin‐cleaved OPN 166.8 ng/mL 12 mo Discriminate ischaemic stroke patients from healthy individuals 86.3 57.7 Jing et al 155
Ischaemic stroke, 60 patients; atherothrombotic: cardioembolic: lacunar = 28:19:13 Blood thrombin‐cleaved OPN 5.47 pmol/L Within 24 h Independent predictors of atherothrombosis 54 91 Ozaki et al 40

Abbreviation: OPN, osteopontin.

6. CONCLUSIONS AND FUTURE PROSPECTS

In the present review, we mainly focused on the roles and therapeutic potential of OPN in acute brain injury including ICH, SAH, cerebral ischaemia, TBI and hypoxia‐ischaemia brain injury. OPN plays a bidirectional role in neuroinflammation. Following acute brain injury, OPN participates in the initiation of inflammation, meanwhile maintaining inflammation homeostasis with a negative feedback mechanism. In addition, BBB maintenance and anti‐apoptotic actions are involved in the OPN‐induced neuroprotection. Moreover, OPN exerts positive roles in the chemotaxis, proliferation, survival and differentiation of various cells, including cerebrovascular smooth muscle cells, neural progenitor cells, neural stem cells and neuroblasts. Collectively, OPN is a pleiotropic extracellular matrix glycoprotein that is mainly believed to be neuroprotective during acute brain injury. These beneficial effects of OPN are induced through its interaction with integrins and CD44. Therefore, OPN represents a potential therapeutic target for acute brain injury. Furthermore, the blood OPN strongly parallels the OPN induced in the brain and can be used as a novel biomarker of the susceptibility, severity and outcome of acute brain injury.

Notably, although endogenous OPN is believed to be neuroprotective, studies regarding TBI and neonatal hypoxia‐ischaemia brain injury suggest that exogenous OPN treatment may be ineffective or even harmful. Thus far, the reason for this discrepancy remains unclear. The efficacy of OPN may depend on the region, injury pattern and stage of brain lesions, and the age and species of victims. Additional experiments based on these variables need to be conducted to determine whether exogenous OPN can indeed activate neuroprotective signalling pathways and consequently improve neurological outcome. Furthermore, the diversity of receptors, various isoforms, post‐translational modifications and different proteolytic fragments contribute to the multiplicity of functions ascribed to OPN, which makes things more difficult to target this molecule or its putative receptors. On the contrary, precise regulation of these different types of OPN and receptors may represent the potential to control its function. Therefore, in future studies, forms of OPN and corresponding receptors should also be specified, which is not satisfactory in previous studies.

Currently, no OPN‐relevant clinical trials are being conducted. Numerous intracerebroventricular or intranasal administrations of recombinant OPN or OPN peptide in experimental models have been carried out. These administration routes, however, limit the clinical application value of OPN‐related agents, although intranasal administration of vitamin D, oral administration of Ephedra sinica extract and hyperbaric oxygen preconditioning have been shown to confer their neuroprotection through endogenous OPN signalling pathways. Besides, there is no agreement about the time window and the appropriate dose for administration hitherto. Studies concerning the protocols that are more applicable to the clinical application of OPN are warranted.

CONFLICTS OF INTEREST

The authors declare that they have no competing interests.

AUTHOR CONTRIBUTIONS

Yunxiang Zhou: Data curation (equal); Writing‐original draft (equal); Writing‐review & editing (equal). Yihan Yao: Writing‐review & editing (equal). Lesang Shen: Validation (equal); Writing‐review & editing (equal). Jianmin Zhang: Writing‐review & editing (equal). John H. Zhang: Conceptualization (equal); Writing‐review & editing (equal). Anwen Shao: Conceptualization (equal); Funding acquisition (equal); Writing‐review & editing (equal).

ACKNOWLEDGEMENTS

This work was supported by grants from China Postdoctoral Science Foundation (2017M612010) and National Natural Science Foundation of China (81701144).

Zhou Y, Yao Y, Sheng L, Zhang J, Zhang JH, Shao A. Osteopontin as a candidate of therapeutic application for the acute brain injury. J Cell Mol Med. 2020;24:8918–8929. 10.1111/jcmm.15641

Yunxiang Zhou, Yihan Yao and Lesang Shen contributed equally to this manuscript.

Contributor Information

Yunxiang Zhou, Email: yxzhou@zju.edu.cn.

Anwen Shao, Email: anwenshao@sina.com.

DATA AVAILABILITY STATEMENT

No data, models or code were generated or used during the study.

REFERENCES

  • 1. Feigin VL, Nichols E, Alam T, et al. Global, regional, and national burden of traumatic brain injury and spinal cord injury, 1990‐2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 2019;18:56‐87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Maas AIR, Menon DK, Adelson PD, et al. Traumatic brain injury: integrated approaches to improve prevention, clinical care, and research. Lancet Neurol. 2017;16:987‐1048. [DOI] [PubMed] [Google Scholar]
  • 3. Wang W, Jiang B, Sun H, et al. Prevalence, incidence, and mortality of stroke in china: results from a nationwide population‐based survey of 480 687 adults. Circulation. 2017;135:759‐771. [DOI] [PubMed] [Google Scholar]
  • 4. Thrift AG, Thayabaranathan T, Howard G, et al. Global stroke statistics. Int J Stroke. 2017;12:13‐32. [DOI] [PubMed] [Google Scholar]
  • 5. Sorby‐Adams AJ, Marcoionni AM, Dempsey ER, et al. The role of neurogenic inflammation in blood‐brain barrier disruption and development of cerebral oedema following acute. Central Nervous System (CNS) Injury. Int J Mol Sci. 2017;18:1788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Hademenos GJ, Massoud TF. Biophysical mechanisms of stroke. Stroke. 1997;28:2067‐2077. [DOI] [PubMed] [Google Scholar]
  • 7. Rothwell PM, Coull AJ, Giles MF, et al. Change in stroke incidence, mortality, case‐fatality, severity, and risk factors in Oxfordshire, UK from 1981 to 2004 (Oxford Vascular Study). Lancet. 2004;363:1925‐1933. [DOI] [PubMed] [Google Scholar]
  • 8. Polivka J, Polivka J, Pesta M, et al. Risks associated with the stroke predisposition at young age: facts and hypotheses in light of individualized predictive and preventive approach. EPMA J. 2019;10:81‐99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Shao A, Zhou Y, Yao Y, et al. The role and therapeutic potential of heat shock proteins in haemorrhagic stroke. J Cell Mol Med. 2019;23:5846‐5858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Sudlow CL, Warlow CP. Comparable studies of the incidence of stroke and its pathological types: results from an international collaboration. International Stroke Incidence Collaboration. Stroke. 1997;28:491‐499. [DOI] [PubMed] [Google Scholar]
  • 11. Gonzalez‐Perez A, Gaist D, Wallander M‐A, et al. Mortality after hemorrhagic stroke: data from general practice (The Health Improvement Network). Neurology. 2013;81:559‐565. [DOI] [PubMed] [Google Scholar]
  • 12. Benjamin EJ, Blaha MJ, Chiuve SE, et al. Heart disease and stroke statistics‐2017 update: a report from the American Heart Association. Circulation. 2017;135:e146‐e603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Benjamin EJ, Virani SS, Callaway CW, et al. Heart disease and stroke statistics‐2018 update: a report from the American Heart Association. Circulation. 2018;137:e67‐e492. [DOI] [PubMed] [Google Scholar]
  • 14. Kardos J, Héja L, Jemnitz K, et al. The nature of early astroglial protection‐Fast activation and signaling. Prog Neurogibol. 2017;153:86‐99. [DOI] [PubMed] [Google Scholar]
  • 15. An C, Jiang X, Pu H, et al. Severity‐dependent long‐term spatial learning‐memory impairment in a mouse model of traumatic brain injury. Transl Stroke Res. 2016;7:512‐520. [DOI] [PubMed] [Google Scholar]
  • 16. Zhou Y, Shao A, Xu W, et al. Advance of stem cell treatment for traumatic brain injury. Front Cell Neurosci. 2019;13:301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Mann AP, Scodeller P, Hussain S, et al. Identification of a peptide recognizing cerebrovascular changes in mouse models of Alzheimer's disease. Nat Commun. 2017;8:1403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Huang L, Zhang L. Neural stem cell therapies and hypoxic‐ischemic brain injury. Prog Neurogibol. 2019;173:1‐17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Hagberg H, Mallard C, Ferriero DM, et al. The role of inflammation in perinatal brain injury. Nat Rev Neurol. 2015;11:192‐208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Thelin EP, Tajsic T, Zeiler FA, et al. Monitoring the neuroinflammatory response following acute brain injury. Front Neurol. 2017;8:351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Yi BR, Kim SU, Choi KC. Development and application of neural stem cells for treating various human neurological diseases in animal models. Lab Anim Res. 2013;29:131‐137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Yokosaki Y, Tanaka K, Higashikawa F, et al. Distinct structural requirements for binding of the integrins alphavbeta6, alphavbeta3, alphavbeta5, alpha5beta1 and alpha9beta1 to osteopontin. Matrix Biol. 2005;24:418‐427. [DOI] [PubMed] [Google Scholar]
  • 23. Mazzali M, Kipari T, Ophascharoensuk V, et al. Osteopontin–a molecule for all seasons. QJM. 2002;95:3‐13. [DOI] [PubMed] [Google Scholar]
  • 24. Icer MA, Gezmen‐Karadag M. The multiple functions and mechanisms of osteopontin. Clin Biochem. 2018;59:17‐24. [DOI] [PubMed] [Google Scholar]
  • 25. Clemente N, Comi C, Raineri D, et al. Role of anti‐osteopontin antibodies in multiple sclerosis and experimental autoimmune encephalomyelitis. Front Immunol. 2017;8:321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Wung JK, Perry G, Kowalski A, et al. Increased expression of the remodeling‐ and tumorigenic‐associated factor osteopontin in pyramidal neurons of the Alzheimer's disease brain. Curr Alzheimer Res. 2007;4:67‐72. [DOI] [PubMed] [Google Scholar]
  • 27. Choi J‐S, Kim H‐Y, Cha J‐H, et al. Transient microglial and prolonged astroglial upregulation of osteopontin following transient forebrain ischemia in rats. Brain Res. 2007;1151:195‐202. [DOI] [PubMed] [Google Scholar]
  • 28. Iczkiewicz J, Rose S, Jenner P. Osteopontin expression in activated glial cells following mechanical‐ or toxin‐induced nigral dopaminergic cell loss. Exp Neurol. 2007;207:95‐106. [DOI] [PubMed] [Google Scholar]
  • 29. Günther M, Plantman S, Davidsson J, et al. COX‐2 regulation and TUNEL‐positive cell death differ between genders in the secondary inflammatory response following experimental penetrating focal brain injury in rats. Acta Neurochir. 2015;157:649‐659. [DOI] [PubMed] [Google Scholar]
  • 30. Gao N, Zhang‐Brotzge X, Wali B, et al. Plasma osteopontin may predict neuroinflammation and the severity of pediatric traumatic brain injury. J Cerebral Blood Flow Metab. 2020;40(1):35‐43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Clemente N, Raineri D, Cappellano G, et al. Osteopontin bridging innate and adaptive immunity in autoimmune diseases. J Immunol Res. 2016;2016:7675437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Lund SA, Giachelli CM, Scatena M. The role of osteopontin in inflammatory processes. J Cell Commun Signal. 2009;3:311‐322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Franzen A, Heinegard D. Isolation and characterization of two sialoproteins present only in bone calcified matrix. Biochem J. 1985;232:715‐724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Senger DR, Wirth DF, Hynes RO. Transformed mammalian cells secrete specific proteins and phosphoproteins. Cell. 1979;16:885‐893. [DOI] [PubMed] [Google Scholar]
  • 35. Ashkar S, Weber GF, Panoutsakopoulou V, et al. Eta‐1 (osteopontin): an early component of type‐1 (cell‐mediated) immunity. Science (New York, NY). 2000;287:860‐864. [DOI] [PubMed] [Google Scholar]
  • 36. Christensen B, Petersen TE, Sorensen ES. Post‐translational modification and proteolytic processing of urinary osteopontin. Biochem J. 2008;411:53‐61. [DOI] [PubMed] [Google Scholar]
  • 37. Sodek J, Ganss B, McKee MD. Osteopontin. Crit Rev Oral Biol Med. 2000;11:279‐303. [DOI] [PubMed] [Google Scholar]
  • 38. Gimba ER, Tilli TM. Human osteopontin splicing isoforms: known roles, potential clinical applications and activated signaling pathways. Cancer Lett. 2013;331:11‐17. [DOI] [PubMed] [Google Scholar]
  • 39. Briones‐Orta MA, Avendaño‐Vázquez SE, Aparicio‐Bautista DI, et al. Osteopontin splice variants and polymorphisms in cancer progression and prognosis. Biochim Biophys Acta Rev Cancer. 2017;1868:93‐108.a. [DOI] [PubMed] [Google Scholar]
  • 40. Ozaki S, Kurata M, Kumon Y, et al. Plasma thrombin‐cleaved osteopontin as a potential biomarker of acute atherothrombotic ischemic stroke. Hypertens Res. 2017;40:61‐66. [DOI] [PubMed] [Google Scholar]
  • 41. Kon S, Nakayama Y, Matsumoto N, et al. A novel cryptic binding motif, LRSKSRSFQVSDEQY, in the C‐terminal fragment of MMP‐3/7‐cleaved osteopontin as a novel ligand for alpha9beta1 integrin is involved in the anti‐type II collagen antibody‐induced arthritis. PLoS One. 2014;9:e116210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Grassinger J, Haylock DN, Storan MJ, et al. Thrombin‐cleaved osteopontin regulates hemopoietic stem and progenitor cell functions through interactions with alpha9beta1 and alpha4beta1 integrins. Blood. 2009;114:49‐59. [DOI] [PubMed] [Google Scholar]
  • 43. Weber GF, Ashkar S, Glimcher MJ, Cantor H. Receptor‐ligand interaction between CD44 and osteopontin (Eta‐1). Science (New York, NY). 1996;271:509‐512. [DOI] [PubMed] [Google Scholar]
  • 44. Wolak T, Sion‐Vardi N, Novack V, et al. N‐terminal rather than full‐length osteopontin or its C‐terminal fragment is associated with carotid‐plaque inflammation in hypertensive patients. Am J Hypertens. 2013;26:326‐333. [DOI] [PubMed] [Google Scholar]
  • 45. Denhardt DT, Noda M. Osteopontin expression and function: Role in bone remodeling. J Cell Biochem. 1998;72(Suppl 30–31):92‐102. [DOI] [PubMed] [Google Scholar]
  • 46. Chackalaparampil I, Peri A, Nemir M, et al. Cells in vivo and in vitro from osteopetrotic mice homozygous for c‐src disruption show suppression of synthesis of osteopontin, a multifunctional extracellular matrix protein. Oncogene. 1996;12:1457‐1467. [PubMed] [Google Scholar]
  • 47. Wang KX, Denhardt DT. Osteopontin: role in immune regulation and stress responses. Cytokine Growth Factor Rev. 2008;19:333‐345. [DOI] [PubMed] [Google Scholar]
  • 48. Chiocchetti A, Comi C, Indelicato M, et al. Osteopontin gene haplotypes correlate with multiple sclerosis development and progression. J Neuroimmunol. 2005;163:172‐178. [DOI] [PubMed] [Google Scholar]
  • 49. Chan JL, Reeves TM, Phillips LL. Osteopontin expression in acute immune response mediates hippocampal synaptogenesis and adaptive outcome following cortical brain injury. Exp Neurol. 2014;261:757‐771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Jin YC, Lee H, Kim SW, et al. Intranasal delivery of RGD motif‐containing osteopontin icosamer confers neuroprotection in the postischemic brain via alphavbeta3 integrin binding. Mol Neurobiol. 2016;53:5652‐5663. [DOI] [PubMed] [Google Scholar]
  • 51. Sun C, Enkhjargal B, Reis C, et al. Osteopontin‐enhanced autophagy attenuates early brain injury via FAK‐ERK pathway and improves long‐term outcome after subarachnoid hemorrhage in rats. Cells. 2019; 8:980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Ellison JA, Velier JJ, Spera P, et al. Osteopontin and its integrin receptor alpha(v)beta3 are upregulated during formation of the glial scar after focal stroke. Stroke. 1998;29:1698‐1706; discussion 707. [DOI] [PubMed] [Google Scholar]
  • 53. Meller R, Stevens SL, Minami M, et al. Neuroprotection by osteopontin in stroke. J Cereb Blood Flow Metab. 2005;25:217‐225. [DOI] [PubMed] [Google Scholar]
  • 54. Doyle KP, Yang T, Lessov NS, et al. Nasal administration of osteopontin peptide mimetics confers neuroprotection in stroke. J Cereb Blood Flow Metab. 2008;28:1235‐1248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Topkoru BC, Altay O, Duris K, et al. Nasal administration of recombinant osteopontin attenuates early brain injury after subarachnoid hemorrhage. Stroke. 2013;44:3189‐3194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Suzuki H, Ayer R, Sugawara T, et al. Protective effects of recombinant osteopontin on early brain injury after subarachnoid hemorrhage in rats. Crit Care Med. 2010;38:612‐618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Wu J, Zhang Y, Yang P, et al. Recombinant osteopontin stabilizes smooth muscle cell phenotype via integrin receptor/integrin‐linked kinase/Rac‐1 pathway after subarachnoid hemorrhage in rats. Stroke. 2016;47:1319‐1327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Jin Y, Kim I‐Y, Kim I‐D, et al. Biodegradable gelatin microspheres enhance the neuroprotective potency of osteopontin via quick and sustained release in the post‐ischemic brain. Acta Biomater. 2014;10:3126‐3135. [DOI] [PubMed] [Google Scholar]
  • 59. Powell MA, Black RT, Smith TL, et al. Matrix metalloproteinase 9 and osteopontin interact to support synaptogenesis in the olfactory bulb after mild traumatic brain injury. J Neurotrauma. 2019;36:1615‐1631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Dixon B, Malaguit J, Casel D, et al. Osteopontin‐Rac1 on blood‐brain barrier stability following rodent neonatal hypoxia‐ischemia. Acta neurochirurgica Supplement. 2016;121:263‐267. [DOI] [PubMed] [Google Scholar]
  • 61. Chen W, Ma Q, Suzuki H, et al. Osteopontin reduced hypoxia‐ischemia neonatal brain injury by suppression of apoptosis in a rat pup model. Stroke. 2011;42:764‐769. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Jullienne A, Hamer M, Haddad E, et al. Acute intranasal osteopontin treatment in male rats following TBI increases the number of activated microglia but does not alter lesion characteristics. J Neurosci Res. 2020;98(1):141–154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Bonestroo HJC, Nijboer CH, van Velthoven CTJ, et al. The neonatal brain is not protected by osteopontin peptide treatment after hypoxia‐ischemia. Dev Neurosci. 2015;37:142‐152. [DOI] [PubMed] [Google Scholar]
  • 64. Albertsson A‐M, Zhang X, Leavenworth J, et al. The effect of osteopontin and osteopontin‐derived peptides on preterm brain injury. J Neuroinflammation. 2014;11:197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Schroeter M, Zickler P, Denhardt DT, et al. Increased thalamic neurodegeneration following ischaemic cortical stroke in osteopontin‐deficient mice. Brain 2006;129:1426‐1437. [DOI] [PubMed] [Google Scholar]
  • 66. Chiocchetti A, Indelicato M, Bensi T, et al. High levels of osteopontin associated with polymorphisms in its gene are a risk factor for development of autoimmunity/lymphoproliferation. Blood. 2004;103:1376–82. [DOI] [PubMed] [Google Scholar]
  • 67. Murugaiyan G, Mittal A, Weiner HL. Increased osteopontin expression in dendritic cells amplifies IL‐17 production by CD4+ T cells in experimental autoimmune encephalomyelitis and in multiple sclerosis. J Immunol. 2008;181:7480‐7488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Tanaka F, Ozawa Y, Inage Y, et al. Association of osteopontin with ischemic axonal death in periventricular leukomalacia. Acta Neuropathol. 2000;100:69‐74. [DOI] [PubMed] [Google Scholar]
  • 69. Plantman S. Osteopontin is upregulated after mechanical brain injury and stimulates neurite growth from hippocampal neurons through beta1 integrin and CD44. NeuroReport. 2012;23:647‐652. [DOI] [PubMed] [Google Scholar]
  • 70. Shin T. Osteopontin as a two‐sided mediator in acute neuroinflammation in rat models. Acta Histochem. 2012;114:749‐754. [DOI] [PubMed] [Google Scholar]
  • 71. Chung AG, Frye JB, Zbesko JC, et al. Liquefaction of the brain following stroke shares a similar molecular and morphological profile with atherosclerosis and mediates secondary neurodegeneration in an osteopontin‐dependent mechanism. eNeuro. 2018;5:ENEURO.0076‐18.2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Jin R, Yang G, Li G. Inflammatory mechanisms in ischemic stroke: role of inflammatory cells. J Leukoc Biol. 2010;87:779‐789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Pang J, Peng J, Matei N, et al. Apolipoprotein E exerts a whole‐brain protective property by promoting M1? Microglia quiescence after experimental subarachnoid hemorrhage in mice. Transl Stroke Res. 2018;9:654‐668. [DOI] [PubMed] [Google Scholar]
  • 74. Jin R, Liu L, Zhang S, et al. Role of inflammation and its mediators in acute ischemic stroke. Cardiovasc Transl Res. 2013;6:834‐851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Wu L, Walas S, Leung W, et al. Neuregulin1‐beta decreases IL‐1beta‐induced neutrophil adhesion to human brain microvascular endothelial cells. Transl Stroke Res. 2015;6:116‐124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Steinman L. A molecular trio in relapse and remission in multiple sclerosis. Nat Rev Immunol. 2009;9:440‐447. [DOI] [PubMed] [Google Scholar]
  • 77. Zhu Q, Luo XU, Zhang J, et al. Osteopontin as a potential therapeutic target for ischemic stroke. Curr Drug Deliv. 2017;14:766‐772. [DOI] [PubMed] [Google Scholar]
  • 78. Castello LM, Raineri D, Salmi L, et al. Osteopontin at the crossroads of inflammation and tumor progression. Mediators Inflamm. 2017;2017:4049098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Kang WS, Choi JS, Shin YJ, et al. Differential regulation of osteopontin receptors, CD44 and the alpha(v) and beta(3) integrin subunits, in the rat hippocampus following transient forebrain ischemia. Brain Res. 2008;1228:208‐216. [DOI] [PubMed] [Google Scholar]
  • 80. Wang X, Louden C, Yue T‐L, et al. Delayed expression of osteopontin after focal stroke in the rat. J Neurosci. 1998;18:2075‐2083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Ladwig A, Walter HL, Hucklenbroich J, et al. Osteopontin augments M2 microglia response and separates M1‐ and M2‐polarized microglial activation in permanent focal cerebral ischemia. Mediators Inflamm. 2017;2017:7189421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Marcondes MCG, Poling M, Watry DD, et al. In vivo osteopontin‐induced macrophage accumulation is dependent on CD44 expression. Cell Immunol. 2008;254:56‐62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Lund SA, Wilson CL, Raines EW, et al. Osteopontin mediates macrophage chemotaxis via alpha4 and alpha9 integrins and survival via the alpha4 integrin. J Cell Biochem. 2013;114:1194‐1202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Okada Y, Copeland BR, Hamann GF, et al. Integrin alphavbeta3 is expressed in selected microvessels after focal cerebral ischemia. Am J Pathol. 1996;149:37‐44. [PMC free article] [PubMed] [Google Scholar]
  • 85. Wan S, Cheng Y, Jin H, et al. Microglia activation and polarization after intracerebral hemorrhage in mice: the role of protease‐activated receptor‐1. Transl Stroke Res. 2016;7:478‐487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Wu B, Ma Q, Suzuki H, et al. Recombinant osteopontin attenuates brain injury after intracerebral hemorrhage in mice. Neurocrit Care. 2011;14:109‐117. [DOI] [PubMed] [Google Scholar]
  • 87. Chen H, Guan B, Chen XI, et al. Baicalin attenuates blood‐brain barrier disruption and hemorrhagic transformation and improves neurological outcome in ischemic stroke rats with delayed t‐PA treatment: involvement of ONOO(‐)‐MMP‐9 pathway. Transl Stroke Res. 2018;9:515‐529. [DOI] [PubMed] [Google Scholar]
  • 88. Ji B, Zhou F, Han L, et al. Sodium tanshinone IIA sulfonate enhances effectiveness Rt‐PA treatment in acute ischemic stroke patients associated with ameliorating blood‐brain barrier damage. Transl Stroke Res. 2017;8:334‐340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Al Dera DH. Neuroprotective effect of resveratrol against late cerebral ischemia reperfusion induced oxidative stress damage involves upregulation of osteopontin and inhibition of interleukin‐1beta. J Physiol Pharmacol. 2017;68:47‐56. [PubMed] [Google Scholar]
  • 90. Baliga SS, Merrill GF, Shinohara ML, Denhardt DT. Osteopontin expression during early cerebral ischemia‐reperfusion in rats: enhanced expression in the right cortex is suppressed by acetaminophen. PLoS One. 2011;6:e14568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Gong L, Manaenko A, Fan R, et al. Osteopontin attenuates inflammation via JAK2/STAT1 pathway in hyperglycemic rats after intracerebral hemorrhage. Neuropharmacology. 2018;138:160‐169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Wu CY, Hsieh HL, Jou MJ, Yang CM. Involvement of p42/p44 MAPK, p38 MAPK, JNK and nuclear factor‐kappa B in interleukin‐1beta‐induced matrix metalloproteinase‐9 expression in rat brain astrocytes. J Neurochem. 2004;90:1477‐1488. [DOI] [PubMed] [Google Scholar]
  • 93. Hu S‐L, Huang Y‐X, Hu R, et al. Osteopontin mediates hyperbaric oxygen preconditioning‐induced neuroprotection against ischemic stroke. Mol Neurobiol. 2015;52:236‐243. [DOI] [PubMed] [Google Scholar]
  • 94. Elmore S. Apoptosis: a review of programmed cell death. Toxicol Pathol. 2007;35:495‐516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Nijhawan D, Honarpour N, Wang X. Apoptosis in neural development and disease. Annu Rev Neurosci. 2000;23:73‐87. [DOI] [PubMed] [Google Scholar]
  • 96. Broughton BR, Reutens DC, Sobey CG. Apoptotic mechanisms after cerebral ischemia. Stroke. 2009;40:e331‐e339. [DOI] [PubMed] [Google Scholar]
  • 97. Gary DS, Mattson MP. Integrin signaling via the PI3‐kinase‐Akt pathway increases neuronal resistance to glutamate‐induced apoptosis. J Neurochem. 2001;76:1485‐1496. [DOI] [PubMed] [Google Scholar]
  • 98. Shi L, Al‐Baadani A, Zhou K, et al. PCMT1 Ameliorates Neuronal Apoptosis by Inhibiting the Activation of MST1 after Subarachnoid Hemorrhage in Rats. Transl Stroke Res. 2017;8:474‐483. [DOI] [PubMed] [Google Scholar]
  • 99. Zhang W, Cui Y, Gao J, et al. Recombinant osteopontin improves neurological functional recovery and protects against apoptosis via PI3K/Akt/GSK‐3beta pathway following intracerebral hemorrhage. Med Sci Monit. 2018;24:1588‐1596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Wu H, Niu H, Shao A, et al. Astaxanthin as a potential neuroprotective agent for neurological diseases. Marine Drugs. 2015;13:5750‐5766. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Denhardt DT, Noda M, O’Regan AW, et al. Osteopontin as a means to cope with environmental insults: regulation of inflammation, tissue remodeling, and cell survival. J Clin Investig. 2001;107:1055‐1061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Khan SA, Lopez‐Chua CA, Zhang J, et al. Soluble osteopontin inhibits apoptosis of adherent endothelial cells deprived of growth factors. J Cell Biochem. 2002;85:728‐736. [DOI] [PubMed] [Google Scholar]
  • 103. Cryns V, Yuan J. Proteases to die for. Genes Dev. 1998;12:1551‐1570. [DOI] [PubMed] [Google Scholar]
  • 104. Nicholson DW, Thornberry NA. Caspases: killer proteases. Trends Biochem Sci. 1997;22:299‐306. [DOI] [PubMed] [Google Scholar]
  • 105. Thornberry NA, Lazebnik Y. Caspases: enemies within. Science (New York, NY). 1998;281:1312‐1316. [DOI] [PubMed] [Google Scholar]
  • 106. Porter AG, Janicke RU. Emerging roles of caspase‐3 in apoptosis. Cell Death Differ. 1999;6:99‐104. [DOI] [PubMed] [Google Scholar]
  • 107. Tulsulkar J, Glueck B, Hinds TD Jr, Shah ZA. Ginkgo biloba extract prevents female mice from ischemic brain damage and the mechanism is independent of the HO1/Wnt pathway. Transl Stroke Res. 2016;7:120‐131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Manabat C, Han BH, Wendland M, et al. Reperfusion differentially induces caspase‐3 activation in ischemic core and penumbra after stroke in immature brain. Stroke. 2003;34:207‐213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. He J, Liu M, Liu Z, Luo L. Recombinant osteopontin attenuates experimental cerebral vasospasm following subarachnoid hemorrhage in rats through an anti‐apoptotic mechanism. Brain Res. 2015;1611:74‐83. [DOI] [PubMed] [Google Scholar]
  • 110. Wang J, Lu Z, Fu X, et al. Alpha‐7 nicotinic receptor signaling pathway Participates in the neurogenesis induced by ChAT‐positive neurons in the subventricular zone. Transl Stroke Res. 2017;8(5):484‐493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Lin YH, Yang‐Yen HF. The osteopontin‐CD44 survival signal involves activation of the phosphatidylinositol 3‐kinase/Akt signaling pathway. J Biol Chem. 2001;276:46024‐46030. [DOI] [PubMed] [Google Scholar]
  • 112. Hasegawa Y, Suzuki H, Sozen T, et al. Apoptotic mechanisms for neuronal cells in early brain injury after subarachnoid hemorrhage. Vienna: Springer Vienna; 2011:43‐48. [DOI] [PubMed] [Google Scholar]
  • 113. Shapira M, Licht A, Milman A, et al. Role of glycogen synthase kinase‐3beta in early depressive behavior induced by mild traumatic brain injury. Mol Cell Neurosci. 2007;34:571‐577. [DOI] [PubMed] [Google Scholar]
  • 114. Krafft PR, Altay O, Rolland WB, et al. alpha7 nicotinic acetylcholine receptor agonism confers neuroprotection through GSK‐3beta inhibition in a mouse model of intracerebral hemorrhage. Stroke. 2012;43:844‐850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Valerio A, Bertolotti P, Delbarba A, et al. Glycogen synthase kinase‐3 inhibition reduces ischemic cerebral damage, restores impaired mitochondrial biogenesis and prevents ROS production. J Neurochem. 2011;116:1148‐1159. [DOI] [PubMed] [Google Scholar]
  • 116. Gabryel B, Kost A, Kasprowska D. Neuronal autophagy in cerebral ischemia–a potential target for neuroprotective strategies? Pharmacol Rep. 2012;64:1‐15. [DOI] [PubMed] [Google Scholar]
  • 117. Chen W, Sun Y, Liu K, Sun X. Autophagy: a double‐edged sword for neuronal survival after cerebral ischemia. Neural Regen Res. 2014;9:1210‐1216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Keaney J, Campbell M. The dynamic blood‐brain barrier. FEBS J. 2015;282:4067‐4079. [DOI] [PubMed] [Google Scholar]
  • 119. Kalluri HS, Dempsey RJ. Osteopontin increases the proliferation of neural progenitor cells. Int J Develop Neurosci. 2012;30:359‐362. [DOI] [PubMed] [Google Scholar]
  • 120. Claassen J, Carhuapoma JR, Kreiter KT, et al. Global cerebral edema after subarachnoid hemorrhage: frequency, predictors, and impact on outcome. Stroke. 2002;33:1225‐1232. [DOI] [PubMed] [Google Scholar]
  • 121. Pang J, Chen Y, Kuai LI, et al. Inhibition of blood‐brain barrier disruption by an apolipoprotein e‐mimetic peptide ameliorates early brain injury in experimental subarachnoid hemorrhage. Transl Stroke Res. 2017;8:257‐272. [DOI] [PubMed] [Google Scholar]
  • 122. Hom S, Egleton RD, Huber JD, Davis TP. Effect of reduced flow on blood‐brain barrier transport systems. Brain Res. 2001;890:38‐48. [DOI] [PubMed] [Google Scholar]
  • 123. Garbuzova‐Davis S, Rodrigues MCO, Hernandez‐Ontiveros DG, et al. Blood‐brain barrier alterations provide evidence of subacute diaschisis in an ischemic stroke rat model. PLoS One. 2013;8:e63553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Nakatsuka Y, Shiba M, Nishikawa H, et al. Acute‐phase plasma osteopontin as an independent predictor for poor outcome after aneurysmal subarachnoid hemorrhage. Mol Neurobiol. 2018;55:6841‐6849. [DOI] [PubMed] [Google Scholar]
  • 125. Suzuki H, Hasegawa Y, Kanamaru K, Zhang JH. Mechanisms of osteopontin‐induced stabilization of blood‐brain barrier disruption after subarachnoid hemorrhage in rats. Stroke. 2010;41:1783‐1790. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Iwanaga Y, Ueno M, Ueki M, et al. The expression of osteopontin is increased in vessels with blood‐brain barrier impairment. Neuropathol Appl Neurobiol. 2008;34:145‐154. [DOI] [PubMed] [Google Scholar]
  • 127. Enkhjargal B, McBride DW, Manaenko A, et al. Intranasal administration of vitamin D attenuates blood‐brain barrier disruption through endogenous upregulation of osteopontin and activation of CD44/P‐gp glycosylation signaling after subarachnoid hemorrhage in rats. J Cereb Blood Flow Metab 2017;37:2555‐2566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Fujimoto M, Takagi Y, Aoki T, et al. Tissue inhibitor of metalloproteinases protect blood‐brain barrier disruption in focal cerebral ischemia. J Cereb Blood Flow Metab. 2008;28:1674‐1685. [DOI] [PubMed] [Google Scholar]
  • 129. Petty MA, Lo EH. Junctional complexes of the blood‐brain barrier: permeability changes in neuroinflammation. Prog Neurogibol. 2002;68:311‐323. [DOI] [PubMed] [Google Scholar]
  • 130. Yatsushige H, Ostrowski RP, Tsubokawa T, et al. Role of c‐Jun N‐terminal kinase in early brain injury after subarachnoid hemorrhage. J Neurosci Res. 2007;85:1436‐1448. [DOI] [PubMed] [Google Scholar]
  • 131. Guo Z, Sun X, He Z, et al. Matrix metalloproteinase‐9 potentiates early brain injury after subarachnoid hemorrhage. Neurol Res. 2010;32:715‐720. [DOI] [PubMed] [Google Scholar]
  • 132. Zuo S, Li W, Li Q, et al. Protective effects of Ephedra sinica extract on blood‐brain barrier integrity and neurological function correlate with complement C3 reduction after subarachnoid hemorrhage in rats. Neurosci Lett. 2015;609:216‐222. [DOI] [PubMed] [Google Scholar]
  • 133. Mahajan SD, Aalinkeel R, Sykes DE, et al. Tight junction regulation by morphine and HIV‐1 tat modulates blood‐brain barrier permeability. J Clin Immunol. 2008;28:528‐541. [DOI] [PubMed] [Google Scholar]
  • 134. Huang B, Krafft PR, Ma Q, et al. Fibroblast growth factors preserve blood‐brain barrier integrity through RhoA inhibition after intracerebral hemorrhage in mice. Neurobiol Dis. 2012;46:204‐214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Ferrara N, Gerber HP, LeCouter J. The biology of VEGF and its receptors. Nat Med. 2003;9:669‐676. [DOI] [PubMed] [Google Scholar]
  • 136. Kusaka G, Ishikawa M, Nanda A, et al. Signaling pathways for early brain injury after subarachnoid hemorrhage. J Cereb Blood Flow Metab. 2004;24:916‐925. [DOI] [PubMed] [Google Scholar]
  • 137. Gavard J, Patel V, Gutkind JS. Angiopoietin‐1 prevents VEGF‐induced endothelial permeability by sequestering Src through mDia. Dev Cell. 2008;14:25‐36. [DOI] [PubMed] [Google Scholar]
  • 138. Gliem M, Krammes K, Liaw L, et al. Macrophage‐derived osteopontin induces reactive astrocyte polarization and promotes re‐establishment of the blood brain barrier after ischemic stroke. Glia. 2015;63:2198‐2207. [DOI] [PubMed] [Google Scholar]
  • 139. Lee H, Jin Y‐C, Kim S‐W, et al. Proangiogenic functions of an RGD‐SLAY‐containing osteopontin icosamer peptide in HUVECs and in the postischemic brain. Exp Mol Med. 2018;50:e430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Brenner D, Labreuche J, Touboul P‐J, et al. Cytokine polymorphisms associated with carotid intima‐media thickness in stroke patients. Stroke. 2006;37:1691‐1696. [DOI] [PubMed] [Google Scholar]
  • 141. van Velthoven CT, Heijnen CJ, van Bel F, Kavelaars A. Osteopontin enhances endogenous repair after neonatal hypoxic‐ischemic brain injury. Stroke. 2011;42:2294‐2301. [DOI] [PubMed] [Google Scholar]
  • 142. Rabenstein M, Hucklenbroich J, Willuweit A, et al. Osteopontin mediates survival, proliferation and migration of neural stem cells through the chemokine receptor CXCR4. Stem Cell Res Ther. 2015;6:99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Yan YP, Lang BT, Vemuganti R, Dempsey RJ. Persistent migration of neuroblasts from the subventricular zone to the injured striatum mediated by osteopontin following intracerebral hemorrhage. J Neurochem. 2009;109:1624‐1635. [DOI] [PubMed] [Google Scholar]
  • 144. Yan YP, Lang BT, Vemuganti R, Dempsey RJ. Osteopontin is a mediator of the lateral migration of neuroblasts from the subventricular zone after focal cerebral ischemia. Neurochem Int. 2009;55:826‐832. [DOI] [PubMed] [Google Scholar]
  • 145. Tao XG, Shi JH, Hao SY, et al. Protective Effects of Calpain Inhibition on Neurovascular Unit Injury through Downregulating Nuclear Factor‐kappaB‐related Inflammation during Traumatic Brain Injury in Mice. Chinese medical journal. 2017;130:187‐198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Suzuki H, Shiba M, Fujimoto M, et al. Matricellular protein: a new player in cerebral vasospasm following subarachnoid hemorrhage. Acta neurochir Suppl. 2013;115:213‐218. [DOI] [PubMed] [Google Scholar]
  • 147. Suzuki H, Hasegawa Y, Kanamaru K, Zhang JH. Effect of recombinant osteopontin on cerebral vasospasm after subarachnoid hemorrhage in rats. Acta Neurochir Suppl. 2011;110:29‐32. [DOI] [PubMed] [Google Scholar]
  • 148. Suzuki H, Hasegawa YU, Chen W, et al. Recombinant osteopontin in cerebral vasospasm after subarachnoid hemorrhage. Ann Neurol. 2010;68:650‐660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149. Mathold K, Wanby P, Brudin L, et al. Alterations in bone turnover markers in patients with noncardio‐embolic ischemic stroke. PLoS One. 2018;13:e0207348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. Ozaki T, Muramatsu R, Sasai M, et al. The P2X4 receptor is required for neuroprotection via ischemic preconditioning. Sci Rep. 2016;6:25893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Li Y, Dammer EB, Zhang‐Brotzge X, et al. Osteopontin is a blood biomarker for microglial activation and brain injury in experimental hypoxic‐ischemic encephalopathy. eNeuro. 2017;4:ENEURO.0253‐16.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152. Abate MG, Moretto L, Licari I, et al. Osteopontin in the cerebrospinal fluid of patients with severe aneurysmal subarachnoid hemorrhage. Cells. 2019;8:695. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153. Carbone F, Busto G, Padroni M, et al. Radiologic cerebral reperfusion at 24 h predicts good clinical outcome. Transl Stroke Res. 2019;10:178‐188. [DOI] [PubMed] [Google Scholar]
  • 154. Carbone F, Vuilleumier N, Burger F, et al. Serum osteopontin levels are upregulated and predict disability after an ischaemic stroke. Eur J Clin Invest. 2015;45:579‐586. [DOI] [PubMed] [Google Scholar]
  • 155. Jing M, Li B, Hou X, et al. OPN gene polymorphism and the serum OPN levels confer the susceptibility and prognosis of ischemic stroke in Chinese patients. Cell Physiol Biochem. 2013;32:1798‐1807. [DOI] [PubMed] [Google Scholar]
  • 156. Kurata M, Okura T, Kumon Y, et al. Plasma thrombin‐cleaved osteopontin elevation after carotid artery stenting in symptomatic ischemic stroke patients. Hypertens Res. 2012;35:207‐212. [DOI] [PubMed] [Google Scholar]
  • 157. Kadoglou NPE, Gerasimidis T, Golemati S, et al. The relationship between serum levels of vascular calcification inhibitors and carotid plaque vulnerability. J Vasc Surg. 2008;47:55‐62. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

No data, models or code were generated or used during the study.


Articles from Journal of Cellular and Molecular Medicine are provided here courtesy of Blackwell Publishing

RESOURCES