Abstract
Intracerebral hemorrhage (ICH) is a major public health problem characterized by cerebral bleeding. Despite recent advances in preclinical studies, there is no effective treatment for ICH making it the deadliest subtype of stroke. The lack of effective treatment options partly attributes to the complexity as well as poorly defined pathophysiology of ICH. The emerging evidence indicates the potential of targeting secondary brain damage and hematoma resolution for improving neurological outcomes after ICH. Herein, we provide an overview of our understanding of the functional roles of activated microglia and brain-infiltrating macrophages in brain injury and repair after ICH. The clinical and preclinical aspects that we discuss in this manuscript are related to ICH that occurs in adults, but not in infants. Also, we attempt to identify the knowledge gap in the field for future functional studies given the potential of targeting microglia and brain-infiltrating macrophages for therapeutic intervention after ICH.
Keywords: Intracerebral hemorrhage, microglia, macrophages, brain injury, repair
1. Introduction
Intracerebral hemorrhage (ICH), defined as spontaneous extravasation of blood into the brain parenchyma, is the second most common form of stroke, accounting for 10–20% of stroke cases worldwide [1, 2]. ICH is the deadliest subtype of stroke, with a very high mortality rate of approximately 40% at one month post the initial ictus. The mortality rate of ICH has not changed over the past two decades [3], despite recent advances in preclinical research. Also, only around 26% of ICH survivors are able to achieve functional independence at one year after symptom onset [4] further indicating the devastating nature of the disease. Currently, the worldwide incidence of ICH is 2 million cases per year [3], with approximately 120,000 cases per year in the United States [5–7]. Importantly, the worldwide incidence and hospital admissions of ICH have increased by ~47% over the last 20 years [1] and 18% in the past ten years [8], respectively. Also, the incidence is estimated to double by 2050 [9] due to increasing life expectancy, population aging and the widespread use of anticoagulants [10].
Based on the pathogenesis of the disease, ICH is classified as primary or secondary ICH. Primary ICH results from the spontaneous rupture of blood vessels at the bifurcation of small arteries mostly due to hypertension or amyloid angiopathy [2] whereas cerebral bleeding that occurs as a consequence of brain tumors, and ischemic stroke is regarded as secondary ICH. The risk factors of ICH include, but are not limited to, hypertension, amyloid angiopathy and advanced age. Of note, around 80% of ICH patients present with hypertension. Cerebral amyloid angiopathy is observed in approximately 20% of ICH patients and is found mostly in the elderly [1, 11]. Other risk factors are smoking, diabetes, alcohol abuse, drug abuse, ethnicity, genetic factors, decreased LDL cholesterol, and anticoagulant and thrombolytic agents [11].
ICH results in severe brain damage which is, in general, categorized into primary brain injury and secondary brain injury [12]. The primary injury is the direct insult to the brain due to the blood vessel rupture and the subsequent hematoma formation [2] and is mainly attributed to the mass effect of the hematoma. Primary injury often leads to the mechanical disruption of brain tissue surrounding the hematoma and subsequent elevation of intracranial pressure (ICP) resulting in brain herniation, based on the size and location of the hematoma [1]. The hematoma is not a static entity due to continued bleeding and in ~30% of ICH patients, significant hematoma expansion is observed which causes an exacerbation of ICP [13] and subsequent decline of neurological functions. The continued cerebral bleeding occurs in 14–20% of ICH patients and the brain regions that are highly vulnerable to hypertensive ICH include the basal ganglia, cerebellum, brainstem, and cortex.
Secondary brain injury (SBI) includes a cascade of cellular and molecular changes in the brain that are initiated by primary brain injury, the release of blood components from the hematoma, and innate immune response characterized by glial activation and infiltration of blood-borne immune cells into the brain [12]. The extravasated blood components such as hemoglobin, hemin, thrombin and iron (Fe) contribute to secondary brain damage [14–16]. Of note, secondary brain injury contributes to further destruction of brain tissue apart from the initial damage due to the cerebral bleeding, and can result in severe neurological deficits and sometimes delayed fatality [17, 18]. ICH patients often exhibit progressive deterioration suggesting a critical role of secondary damage in the pathophysiology of ICH [16, 19]. The key mechanisms of secondary brain injury include neuroinflammation [20], oxidative stress [21] and neurotoxicity induced by blood components [22], which often results in blood- brain barrier break down [23], development of cerebral edema [13], hematoma expansion [13] and severe neurological deterioration or death.
Currently, there is no effective treatment for ICH and the clinical management of ICH is largely restricted to supportive care and surgery for selected patients [4]. Of note, surgical intervention to remove the hematoma has not been shown to provide a significant increase in patient survival, or improve neurological recovery in surviving patients [19]. Therefore, it is pivotal to find other avenues that can improve the prognosis of ICH patients. Along these lines, delineating the molecular mechanisms of secondary brain injury, which is responsible for both acute as well as long term neurological outcomes after ICH is critical. Further, secondary brain injury persists for a prolonged period and hence, it could provide a wider window for therapeutic intervention after ICH. While blood components being the key molecular mediators of secondary brain damage, the major cellular contributor of secondary brain injury after ICH is microglia, the inflammatory cells of the CNS (central nervous system). This review focuses on the role of microglia and brain-infiltrating monocyte-derived macrophages in secondary brain injury and brain repair after ICH. Also, the review discusses the potential of targeting microglia and brain-infiltrating macrophages for therapeutic intervention after ICH. The clinical and preclinical aspects that we discuss throughout this manuscript are related to ICH that occurs in adults, but not in infants and neonatal ICH differs in epidemiology, pathophysiology and clinical management from that in adults. Hence, the concepts described in this article may not fit for ICH in infants.
2. Microglia/macrophage activation and ICH
In a non-injured adult brain, microglia is critical for maintaining brain homeostasis, remodeling synapses and constantly surveying the brain for damaged or dead neurons and foreign bodies [24]. Microglia are the resident immune cells of the CNS and the first-responders of brain injury. Neuroinflammation, characterized by microglial activation, infiltration of leukocytes and subsequent release of cytokines, chemokines and various neurotoxic molecules, is a key determinant of secondary brain injury after ICH [25].
Hemorrhagic injury to brain results in prominent microglial activation in preclinical models of ICH characterized by enlarged cell body with thick and short cell processes and increased expression of microglia or macrophage-specific molecular markers such as Iba1 or cd11b in comparison to non-activated or resting microglia observed in an un-injured or healthy adult brain [26]. Microglial activation was mostly observed in the brain region that encircles the hematoma, called the perihematomal brain region and upon hemorrhagic brain injury microglial/macrophage activation was observed at 1 hour and 4 hours post-ICH in the collagenase injection model [27] and the blood injection model of ICH [28], respectively. In the collagenase injection model, peak microglia/ macrophage activation was observed at day 3 and day 5 post-ICH and there was an 8-fold increase in the number of activated microglia or macrophages on day 3 and day 5 post-ICH in comparison to experimental control [4]. The increase in the number of microglia or macrophages after ICH could be due to the injury-induced proliferation of microglia or migration of microglia to the injury site or brain infiltration of blood-borne macrophages (macrophages). Macrophages have been found to migrate into the injury site within 12 hours in a preclinical model of ICH [29]. Also, activated microglia/macrophages in different brain regions have different morphologic characteristics, indicating different functions [30]. Microglial/macrophage activation persists for 4 weeks in preclinical models. Notably, microglial activation also occurs after ICH in human patients, with brain invasion of leukocytes (neutrophils, macrophages) [31]. Activated microglia and macrophages share several immune cell markers in common [32]. Therefore, it is difficult to differentiate activated microglia from macrophages by commonly used techniques like immunohistochemistry and hence, they are often grouped as microglia/macrophages when discussing their functional roles following a brain injury [33].
Although the precise functional role of activated microglia is yet to be defined, activated microglia are regarded as the critical cellular regulators of neuroinflammation after ICH based on their ability to release cytokines, chemokines, prostaglandins, proteases, ferrous iron, glutamate, and reactive oxygen species [34–36]. The cytokines, in turn, cause further activation of microglia, resulting in a self-propagating inflammatory cascade leading to neuronal dysfunction and cell death [37, 38]. Furthermore, activated microglia also modulate neuroinflammation by recruiting and activating blood-derived macrophages [39–42]. And several preclinical studies focused on assessing the microglia or macrophage activation in the ipsilateral brain region as a measure of neuroinflammatory response post-ICH. Post-injury administration of a tripeptide MIF (macrophage/microglial inhibitory factor, tuftsin fragment 1–3, Thr-Lys-Pro) in a collagenaseinjection mouse model of ICH resulted in decreased microglial/macrophage activation, hemorrhagic lesion size, neurodegeneration, cerebral edema, and acute neurological deficits post-ICH [43]. Further, tuftsin attenuated thrombin-induced release of IL-1-β and TNF-α from rat microglia and intracerebral infusion of tuftsin attenuated cerebral edema in a blood-injection model of ICH [44]. Also, atorvastatin-mediated attenuation of cerebral edema and neuronal death on day 3 post-ICH in a pre-clinical model was associated with the reduction in microglia or macrophage activation [45]. Furthermore, minocycline, a tetracycline derivative and an inhibitor of microglia/macrophage activation attenuated cerebral edema and improved neurological outcomes after ICH [46]. Apart from cytokines, chemokines are also critical regulators of neuroinflammation and CXCR4 (C-X-C chemokine receptor type 4) antagonist, CX807-mediated neuroprotection in a preclinical model of ICH was associated with a profound reduction in microglia/macrophage activation [47]. Altogether, microglial/macrophage activation correlates with blood brain-barrier damage, brain swelling/edema, hematoma expansion, peri-hematomal cell death, neurological deterioration, and poor functional recovery after ICH [45, 48–55] and reduction in microglia/macrophage activation in the acute phase of ICH is associated with improved functional recovery in preclinical models.
Microglia/macrophages are activated by various inflammatory stimuli following ICH that include blood components such as thrombin, hemoglobin, heme, iron and plasma proteins such as fibrinogen [4]. Necrotic neurons release many cellular contents, such as ATP, neurotransmitters, heat shock proteins and high-mobility group protein B1 (HMGB1), that also activate microglia following ICH [33]. The inflammatory stimuli hemin (a hemoglobin degradation product that accumulates in the brain after hemorrhage) and fibrinogen act through Toll-like receptors that are found predominantly on the surface of microglia and macrophages [56–59] and potentiate microglia and macrophage activation [57]. Exogenous hemin treatment increased the expression of TLR4, and proinflammatory cytokines, such as TNF-α, IL-1β and IL-6 in cultured microglia [57]. Consistently, TLR4 expression is found to be up-regulated after ICH [60] and contributes to an inflammatory response through the nuclear factor-κB (NFκB) signaling pathway [33]. TLR4 knockout animals exhibited decreased microglial activation, brain infiltration of neutrophils and monocytes, and improved functional recovery at 3 days post-ICH [60]. Furthermore, the administration of a TLR4 antagonist and anti-TLR4 antibody reduced brain water content, neurological deficit scores, proinflammatory cytokine levels, and brain infiltration of monocytes/macrophages after ICH [57, 61] suggesting a major role of TLR-4 in acute neuroinflammation following ICH. Also, recent clinical studies have shown that increased expression of TLR4 in monocytes is associated with poor functional outcome and enhanced residual lesion volume in ICH patients [62]. Apart from TLR-4, recent studies document NLRP3 inflammasome as a potent regulator of neuroinflammation, and proinflammatory cytokines secretion after ICH [63, 64]. TLR4-NFkB signaling can cause augmented expression of NLRP3 and pro-IL-1 β [64]. Activation of NLRP3 by inflammatory stimuli results in the oligomerization of NLRP3 with an adaptor protein ASC and the inactive form of the enzyme caspase-1 leading to caspase-1 activation and subsequent generation of biologically active proinflammatory cytokines, IL-1β and IL-18 from its precursors. Notably, expression of NLRP3, caspase-1, and IL-1β is up regulated in microglia after collagenase-induced ICH and heme activates NLRP3 inflammasome [63]. In addition, recent studies report increased levels of complement 3 (C3) in the ipsilateral brain area in a preclinical model of ICH and genetic ablation of C3 attenuated ICH-induced IL-1β production suggesting a role of NLRP3 signaling pathway in complement-induced neuroinflammation after ICH [65]. Altogether, NLRP3 activation and NFκB upregulation play a critical role in ICH-induced inflammatory response. In addition, the inhibition of microglia/macrophage inflammatory pathways in preclinical models of ICH has resulted in a reduction in the brain infiltration of leukocytes (neutrophils, macrophages) [61, 66] implicating that microglia recruits other immune cell types to the injury site. To this end, microglia release CXCL2 a chemokine that attracts neutrophils. It has been shown that neutrophils also contribute to inflammation following ICH and its brain recruitment exacerbated the secondary brain injury [33]. Neutrophils may damage the brain by producing reactive oxygen species, releasing pro-inflammatory proteases and thereby modulating blood–brain-barrier permeability[67].
ICH results in profound neuronal death in the peri-hematomal and hematomal brain region contributing to patient morbidity and mortality. Consistent with the role of microglial/macrophage activation in inducing cell death [68–70], neuronal death in the ipsilateral brain region after ICH spatially and temporarily correlated with microglia/macrophage activation [71]. Types of cell death observed after ICH are necrosis, apoptosis, pyroptosis, and autophagy [72–76]. NFκB, a transcription factor is a key regulator of proinflammatory cytokines, such as TNF-α and IL-1-β in various pathological conditions including ICH [57] that can cause direct or indirect neurotoxicity after a brain injury. NFκB transcriptional activity correlated with perilesional cell death after ICH in rats [48] and IL-1β and TNF-α antagonists attenuated apoptotic cell death in experimental ICH [77]. Also, activation of NFκB is positively related to the progression of apoptotic cell death in patients with ICH [78]. In addition, NLRP3-induced pyroptosis, documented in experimental models of ICH, may play a significant role in brain damage after ICH [79] because apart from resulting in brain cell loss, pyroptosis is associated with the release of proinflammatory mediators that can further damage the brain. Given the role of cell death in neurological deficits after ICH, further studies are warranted determining the occurrence of pyroptosis in human ICH and delineating further the precise molecular mechanisms of inflammation-induced cell death in preclinical models.
2a. M1/M2 microglia/macrophages and ICH
Microglia/macrophages have been found to play roles in both brain tissue damage, and recovery after ICH [80]. These opposing roles following a brain injury are largely attributed to the capability of microglia/macrophages to undergo polarization [80]. We and others have demonstrated that microglia/macrophages can assume a pro-inflammatory (M1) or an anti-inflammatory (M2) phenotype after ICH [4]. The molecular markers such as CD16/32, CD86, CD68, and iNOS are being used, in general, to identify the M1 microglia/macrophages. ICH results in profound activation of M1 microglia/macrophages within the first week [4]. Consistently, the kinetics of cytokine expression within the first week after experimental ICH is dynamic, and important clinical sequelae begin to appear within the first 7-days post-ICH [9]. M1 or classically activated microglia/macrophages produce pro-inflammatory cytokines, such as TNF-α, IL-6, and IL1-β, and nitric oxide (NO) and contributes to both inflammatory as well as oxidative brain damage [81]. Proinflammatory cytokines can cause neutrophil recruitment in the region surrounding the hematoma by modulating the blood brain barrier permeability. TNF- α, IL-1-β, reactive oxygen species, and matrix metallo proteinase-9 (MMP-9) released by activated microglia can augment blood-brain-barrier permeability. The intactness of the blood-brain-barrier is essential for brain homeostasis and an increase in blood-brain-barrier permeability leads to the accumulation of blood components in the brain and develops a life-threatening neuropathological condition called cerebral edema, which is often associated with ICH patients. Consistently, plasma TNF-α levels correlated with the severity of brain edema [82] and poor outcomes in ICH patients [83]. The post-injury administration of TNF-α antibody attenuated cerebral edema and improved neurobehavioral outcomes in a murine model of ICH [84]. Importantly, there is a positive correlation between the severity of peri-hematomal edema and a poor outcome in patients with ICH [85–87] and hence, strategies to attenuate brain edema are recommended as part of neurointensive care for patients with ICH [88]. Thrombin is a key regulator of proinflammatory microglial/macrophage activation and inflammatory cytokine release after ICH [89, 90]. Thrombin-induced microglial activation involves PAR (protease-activated receptor) subtypes, PAR-1, and PAR-4 [89, 90]. Genetic knockdown of PAR-1 inhibited M1 phenotypic microglia/macrophage activation and reduced the expression of proinflammatory cytokines, neuronal death, and brain injury after ICH in mice [66]. Furthermore, edaravone (an FDA approved drug for amyotrophic lateral sclerosis) - and pinocembrin (a natural product)-mediated neuroprotection after ICH in pre-clinical models were associated with the reduction of M1 microglia/macrophage phenotype [91, 92]. Importantly, a recent clinical study reported an independent correlation between a reduction in M1- like macrophages in the hematoma and favorable outcome on day 90 post-ICH [93]. Altogether, recent studies have shown that attenuation of M1 microglia/macrophage activation correlates with improved neurological outcomes after ICH.
M2 or alternatively activated microglia/macrophages express scavenger receptors and release anti-inflammatory cytokines, such as TGF-β, IL-4, IL-10, IL-13, and growth factors such as VEGF, BDNF, platelet-derived growth factor (PDGF), and progranulin, facilitating brain recovery [80]. Alternative microglia activation occurs in the absence of strong inflammatory signals such as Toll-like receptors signals and is induced by anti-inflammatory cytokines such as IL-10, IL-4 and IL-13. The molecular markers like CD206, CD163, arginase-1, CD36, YM1 and FIZZ1, are currently in use for the identification and characterization of M2 microglia/macrophages after ICH. In preclinical models of ICH, CD206-positive M2 microglia/ macrophages are observed on day 1 to day 3 post-ICH [4, 66]. However, in contrast to M2, M1 microglial or macrophage activation after ICH persists for a longer period of time implicating a possible role of proinflammatory response in long-term neurological deficits after ICH [4]. M2 microglia/macrophages are further classified into M2a, M2b and M2c, which vary in their anti-inflammatory potential and tissue remodeling capabilities [80, 94–97].
Both preclinical and clinical studies document hematoma volume, as an independent determinant of neurological outcomes after ICH [3] and inflammatory markers in the acute phase of ICH are associated with hematoma expansion after ICH, a predictor of poor prognosis in ICH patients [98]. The timely removal of the components of hematoma, the ongoing source of brain inflammation, is critical for functional recovery after ICH. Recent preclinical findings indicated that M2 microglia/ macrophage play roles in the phagocytosis of cellular debris and blood components in the brain tissue following ICH [17], a process that could enhance hematoma clearance or hematoma resolution [91, 99] and facilitate brain repair and recovery. Consistently, selective depletion of M2 macrophages resulted in reduced hematoma clearance and enhanced brain edema [100]. Furthermore, administration of anti-inflammatory cytokine IL-10 polarized microglia/macrophages toward alternatively activated phenotype and reduced hematoma volume at day 3 post-ICH in mice [42] and neutralization of IL-10 receptor prevented microglial phagocytosis in vitro [42]. Also, the adoptive transfer of T regulatory cells ameliorated ICH-induced inflammatory injury by modulating microglia/macrophage polarization toward the M2 phenotype through the IL-10/GSK3β/PTEN axis [101], implicating the role of anti-inflammatory signaling in the clearance of hematoma and functional recovery after ICH. Consistently, intracerebral injection of an anti-inflammatory cytokine, IL-4 improved neurological outcomes after ICH in rat with concomitant inhibition of M1 activation and induction of M2 activation [102]. Moreover, improved hematoma resolution was observed in TLR-4 knockout mice in comparison to wild type after ICH [103]. Apart from the phagocytic potential, M2 microglia/macrophages could play roles in the catabolism of hemoglobin [104] and hemin that contribute to ICH-induced brain damage. Recent studies demonstrate that induction of HO-1 (heme oxygenase-1), a rate-limiting enzyme in heme catabolism, in microglia is necessary for the clearance of cerebral blood burden and attenuation of neuronal cell death after subarachnoid hemorrhage [105]. However, though HO-1 is induced mainly in microglia/macrophages after ICH [106], the precise functional role of HO-1 in microglial/macrophage-mediated phagocytosis after ICH remains largely unknown, warranting investigation. Further, consistent with the role of iron in ICH- induced inflammatory and oxidative brain damage, administration of VK-28, a brain-permeable iron chelator, augmented CD206-positive M2 macrophages, reduced brain edema and improved neurobehavioral outcomes after ICH in mice [107]. Overall, the strategies that augment M2 microglia/macrophage phenotypes after ICH are found to attenuate secondary brain damage and improve neurological outcomes by augmenting anti-inflammatory response and hematoma clearance.
Though the classification of activated microglia/macrophages into pro-inflammatory “M1”, and anti-inflammatory “M2” phenotypes are widely accepted, recent literature questions the effectiveness of this nomenclature since microglia/macrophage response to a brain injury is too dynamic and complex to be defined by the concept of polarization [108, 109]. To this end, M1-like macrophages, in response to certain stimuli, can be switched towards an M2-like phenotype and vice versa [110]. Also, in a preclinical model of TBI (traumatic brain injury) microglia/macrophages were found to express both M1 and M2 markers concurrently across multiple time points [111]. Another study on TBI found that monocyte-derived macrophages do not differentiate into defined subsets of phenotypes, but exhibit both inflammatory and wound healing features at the same time [112]. It is also postulated that M1-polarized microglia/macrophages could play roles in early repair processes and prolonged M2 activation can result in abnormal repair [113]. Altogether, given the complexity of microglia/macrophage functions coupled with its dynamic nature, it is imperative to conduct further studies characterizing the functional roles associated with microglia or macrophages in a context-and time-dependent manner and its consequences on brain damage and recovery after ICH.
2b. Cell-cell communication and microglial activation after ICH
Astrocytes are the most abundant cell type in the central nervous system, [114] that play roles in axonal guidance, neuronal survival and functioning, maintenance of blood-brain-barrier, regulation of cerebral blood flow, and secretion of neuroprotective factors [114, 115]. Despite a profound induction of astrogliosis after ICH, the precise functional role of reactive or activated astrocytes after ICH is largely unknown. It is reported that reactive astrocytes contribute to innate immune response and play both beneficial [115] as well as detrimental roles after ICH [116]. To this end, reactive astrocytes after a brain injury release numerous pro-inflammatory as well as anti-inflammatory cytokines, which in turn can regulate microglial activation [116]. Of note, a recent study reported interleukin-15 (IL-15) as a key mediator of cross-talk between astrocytes and microglia following ICH [117]. To this end, astrocyte-targeted expression of IL-15 was able to recruit microglia to the peri-hematomal area, and differentiate microglia towards pro-inflammatory phenotype, resulting in worse neurological outcomes after ICH in mice [117]. Also, astrocytes secrete many chemokines such as CCL2 which can differentiate microglia [80] and cause the brain recruitment of peripheral macrophages [118, 119] after a brain injury. To this end, the treatment of microglia culture with the conditioned media derived from TNF-α stimulated-astrocytes activated and differentiated microglia into the M1 phenotype in a CCl2-dependent manner [118]. Another chemokine shown to be secreted by astrocytes that could play roles in the astrocyte-microglia cross-talk is Cxcl2 [4] which exerts its effects by binding to its receptor, Cxcr2 expressed on microglia [120]. The upregulation of Cxcl2 mRNA has been reported in the brains of human ICH patients [121]. Cxcr1/2 antagonist reduced neurological deficits in a preclinical model of ICH [122] implicating a detrimental role of Cxcl2 and astrocyte-microglia interaction. On the contrary, a recent study showed that astrocytes are able to influence microglia to attain a neuroprotective phenotype. To this end, astrocyte-derived peptide, humanin augmented hematoma resolution and attenuated neurological deficits following ICH in mice by up-regulating microglial phagocytic activity [123]. Furthermore, astrocyte-derived chemokines such as Cxcl1, Cxcl3, cxcl10, and CCr1 are found to be upregulated after ICH [80, 116], however, their functional role in astrocyte-microglia communication and the pathophysiology of ICH is yet to established. Also, inhibition of astrocytic synthesis of calcium-binding protein, S100B by arundic acid improved neurobehavioral outcomes in a collagenase-induced rodent model of ICH and that was accompanied by a reduction in microglial/macrophage activation [124]. Notably, activated microglia are able to induce astrocytic proinflammatory response [125], however, the functional consequences of microglia-astrocyte cross- talk remain least explored in ICH-mediated brain injury, requiring investigation.
As discussed earlier, activated microglia play a significant role in the death or survival of the neurons. Also, dying or damaged neurons can modulate glial functions. Along these lines, neuronal death is associated with the release of many Danger-Associated Molecular Patterns (DAMPs) such as ATP, neurotransmitters, heat shock proteins, and high mobility group box 1 (HMGB1) which activate microglia [33]. Among these, HMGB1 exhibits differential roles following ICH. HMGB1 plays a pro-inflammatory role in the acute phase of ICH [126] and high serum levels of HMGB1 in patients with acute ICH correlate with disease severity [127]. In a rat ICH model that pharmacological inhibition of HMGB1 improved acute neurological outcomes [126, 128] possibly by modulating TLR-4/NfκB inflammatory pathway [129]. In contrast, HMGB1 contributes to neurogenesis and brain recovery by modulating the expression of brain-derived neurotrophic factor and matrix metalloproteinase-9 in the delayed phase of ICH [129]. HMGB1 is fairly ubiquitous and is expressed by many different cell types and hence further studies are warranted exploring its therapeutic potential after ICH.
2c. Macrophage activation and ICH
After ICH, monocyte-derived macrophages infiltrate into the brain. Though phagocytosis of apoptotic cells or efferocytosis is one of the primary functions of macrophages and the functional role of infiltrating macrophages after ICH is largely controversial. Based on preclinical studies, macrophages exert both neuroprotective and neurotoxic roles after ICH. Along these lines, the infiltration of leukocytes from the periphery further increases the inflammatory brain damage [130–132]. Also, acute neuroprotection after ICH is often associated with the reduction in macrophage infiltration in preclinical models[133]. In contrast, as per recent reports, infiltrating macrophages also play critical roles in hematoma resolution and thereby functional recovery after ICH [119, 134] and infiltrated macrophages exhibited changes in the expression profile of genes related to pro inflammatory response, anti-inflammatory response and hematoma clearance in a time dependent-manner after ICH [119]. Consistent with this observation, depletion of CCR2+ inflammatory monocytes reduced neurological deficits after ICH in the acute phase but delayed hematoma clearance [131] implicating the dual and opposing roles of macrophages after ICH.
3. Conclusions and future directions
Overall, microglial/macrophage activation plays a detrimental role in the acute phase of ICH. However, preclinical studies have mostly employed male young subjects and hence further studies are warranted testing the efficacy of inhibiting microglia/macrophage activation in female subjects. Moreover, preclinical studies involving aged animals are also required, given the prevalence of ICH in the elderly population. To date, a few studies have documented altered microglial/ macrophage activation in aged animals after ICH implicating the need to further explore the therapeutic efficacy of targeting microglial or macrophage activation in improving neurological outcomes. Also, the development of novel transgenic mouse models using Cre-lox technology could help in functionally differentiating microglia from macrophages and that may result in the identification of microglia or macrophage- specific molecular targets for therapeutic intervention after ICH. In addition, how acute inhibition of microglia/macrophage activation modulates long- term neurobehavioral outcomes after ICH requires investigation. Further, activated microglia and macrophage are less susceptible to blood-induced toxicity in comparison to neurons making them an efficient target for therapeutic intervention after ICH warranting further studies.
Highlights.
Microglial/macrophage activation plays a detrimental role in the acute phase of ICH.
Functional characterization of microglia and macrophages as distinct entities may better define ICH pathophysiology.
Funding
This work was supported by grants from the National Institutes of Health (R01NS107853) and American Heart Association (14SDG18730034) to SSR.
Abbreviations
- ICH
Intracerebral hemorrhage
- LDL
Low density lipoprotein
- ICP
Intracranial pressure
- SBI
Secondary brain injury
- Fe
Iron
- CNS
Central nervous system
- Iba1
Ionized calcium binding adaptor molecule 1
- MIF
Macrophage/microglial inhibitory factor
- IL-1β
Interleukin-1β
- TNF-α
Tumor necrosis factor-α
- CXCR4
C-X-C chemokine receptor type 4
- ATP
Adenosine triphosphate
- HMGB1
High mobility group box 1
- TLR4
Toll-like receptor4
- IL-6
Interleukin-6
- NF-κB
Nuclear factor-κB
- NLRP3
NLR Family Pyrin Domain Containing 3
- IL-18
Interleukin-18
- C3
Complement 3
- Cxcl2
Chemokine (C-X-C motif) ligand 2
- NO
Nitric oxide
- MMP-9
Matrix metalloproteinase-9
- PAR
Protease activated receptor
- PAR-1
Protease activated receptor-1
- PAR-4
Protease activated receptor-4
- FDA
Food and drug administration
- IL-4
Interleukin-4
- IL-10
Interleukin-10
- IL-13
Interleukin-13
- VEGF
Vascular endothelial growth factor
- BDNF
Brain-derived neurotrophic factor
- PDGF
Platelet-derived growth factor
- HO-1
Heme oxygenase-1
- TBI
Traumatic brain injury
- IL-15
Interleukin-15
- CCL2
The chemokine (C-C motif) ligand 2
- Cxcr2
C-X-C motif chemokine receptor 2
- mRNA
messenger RNA
- Cxcl1
Chemokine (C-X-C motif) ligand 1
- Cxcl3
Chemokine (C-X-C motif) ligand 3
- Cxcl10
Chemokine (C-X-C motif) ligand 10
- CCr1
C-C chemokine receptor type 1
- DAMPs
Danger-Associated Molecular Patterns
- CCR2+
C-C chemokine receptor type 2+
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Declarations of interest
None
References
- 1.An SJ, Kim TJ, and Yoon BW, Epidemiology, Risk Factors, and Clinical Features of Intracerebral Hemorrhage: An Update. J Stroke, 2017. 19(1): p. 3–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Qureshi AI, Mendelow AD, and Hanley DF, Intracerebral haemorrhage. Lancet, 2009. 373(9675): p. 1632–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.van Asch CJ, et al. , Incidence, case fatality, and functional outcome of intracerebral haemorrhage over time, according to age, sex, and ethnic origin: a systematic review and meta-analysis. Lancet Neurol, 2010. 9(2): p. 167–76. [DOI] [PubMed] [Google Scholar]
- 4.Bonsack F.t., Alleyne CH Jr., and Sukumari-Ramesh S, Augmented expression of TSPO after intracerebral hemorrhage: a role in inflammation? J Neuroinflammation, 2016. 13(1): p. 151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Aguilar MI and Freeman WD, Spontaneous intracerebral hemorrhage. Semin Neurol, 2010. 30(5): p. 555–64. [DOI] [PubMed] [Google Scholar]
- 6.Broderick J, et al. , Guidelines for the management of spontaneous intracerebral hemorrhage in adults: 2007 update: a guideline from the American Heart Association/American Stroke Association Stroke Council, High Blood Pressure Research Council, and the Quality of Care and Outcomes in Research Interdisciplinary Working Group. Circulation, 2007. 116(16): p. e391–413. [DOI] [PubMed] [Google Scholar]
- 7.Ribo M and Grotta JC, Latest advances in intracerebral hemorrhage. Curr Neurol Neurosci Rep, 2006. 6(1): p. 17–22. [DOI] [PubMed] [Google Scholar]
- 8.Qureshi AI, et al. , Changes in cost and outcome among US patients with stroke hospitalized in 1990 to 1991 and those hospitalized in 2000 to 2001. Stroke, 2007. 38(7): p. 2180–4. [DOI] [PubMed] [Google Scholar]
- 9.Qureshi AI, et al. , Spontaneous intracerebral hemorrhage. N Engl J Med, 2001. 344(19): p. 1450–60. [DOI] [PubMed] [Google Scholar]
- 10.Wang J, Preclinical and clinical research on inflammation after intracerebral hemorrhage. Prog Neurobiol, 2010. 92(4): p. 463–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Morotti A and Goldstein JN, Diagnosis and Management of Acute Intracerebral Hemorrhage. Emerg Med Clin North Am, 2016. 34(4): p. 883–899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Lok J, et al. , Intracranial hemorrhage: mechanisms of secondary brain injury. Acta Neurochir Suppl, 2011. 111: p. 63–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Babi MA and James ML, Peri-Hemorrhagic Edema and Secondary Hematoma Expansion after Intracerebral Hemorrhage: From Benchwork to Practical Aspects. Front Neurol, 2017. 8: p. 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Yang GY, et al. , Experimental intracerebral hemorrhage: relationship between brain edema, blood flow, and blood-brain barrier permeability in rats. J Neurosurg, 1994. 81(1): p. 93–102. [DOI] [PubMed] [Google Scholar]
- 15.Wagner KR, et al. , Lobar intracerebral hemorrhage model in pigs: rapid edema development in perihematomal white matter. Stroke, 1996. 27(3): p. 490–7. [DOI] [PubMed] [Google Scholar]
- 16.Babu R, et al. , Thrombin and hemin as central factors in the mechanisms of intracerebral hemorrhage-induced secondary brain injury and as potential targets for intervention. Neurosurg Focus, 2012. 32(4): p. E8. [DOI] [PubMed] [Google Scholar]
- 17.Aronowski J and Zhao X, Molecular pathophysiology of cerebral hemorrhage: secondary brain injury. Stroke, 2011. 42(6): p. 1781–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Xi G, Keep RF, and Hoff JT, Mechanisms of brain injury after intracerebral haemorrhage. Lancet Neurol, 2006. 5(1): p. 53–63. [DOI] [PubMed] [Google Scholar]
- 19.Elliott J and Smith M, The acute management of intracerebral hemorrhage: a clinical review. Anesth Analg, 2010. 110(5): p. 1419–27. [DOI] [PubMed] [Google Scholar]
- 20.Zhang Z, et al. , Microglial Polarization and Inflammatory Mediators After Intracerebral Hemorrhage. Mol Neurobiol, 2017. 54(3): p. 1874–1886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Duan X, et al. , Intracerebral Hemorrhage, Oxidative Stress, and Antioxidant Therapy. Oxid Med Cell Longev, 2016. 2016: p. 1203285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Garton T, et al. , Brain iron overload following intracranial haemorrhage. Stroke Vasc Neurol, 2016. 1(4): p. 172–184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Aguilar MI and Brott TG, Update in intracerebral hemorrhage. Neurohospitalist, 2011. 1(3): p. 148–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Prinz M and Priller J, Microglia and brain macrophages in the molecular age: from origin to neuropsychiatric disease. Nat Rev Neurosci, 2014. 15(5): p. 300–12. [DOI] [PubMed] [Google Scholar]
- 25.Emsley HC and Tyrrell PJ, Inflammation and infection in clinical stroke. J Cereb Blood Flow Metab, 2002. 22(12): p. 1399–419. [DOI] [PubMed] [Google Scholar]
- 26.Kreutzberg GW, Microglia: a sensor for pathological events in the CNS. Trends Neurosci, 1996. 19(8): p. 312–8. [DOI] [PubMed] [Google Scholar]
- 27.Wang J and Dore S, Heme oxygenase-1 exacerbates early brain injury after intracerebral haemorrhage. Brain, 2007. 130(Pt 6): p. 1643–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Xue M and Del Bigio MR, Intracerebral injection of autologous whole blood in rats: time course of inflammation and cell death. Neurosci Lett, 2000. 283(3): p. 230–2. [DOI] [PubMed] [Google Scholar]
- 29.Hammond MD, Ai Y, and Sansing LH, Gr1+ Macrophages and Dendritic Cells Dominate the Inflammatory Infiltrate 12 Hours After Experimental Intracerebral Hemorrhage. Transl Stroke Res, 2012. 3(1): p. s125–s131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Yang SS, et al. , High Morphologic Plasticity of Microglia/Macrophages Following Experimental Intracerebral Hemorrhage in Rats. Int J Mol Sci, 2016. 17(7). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Shtaya A, et al. , Rapid neuroinflammatory changes in human acute intracerebral hemorrhage. Ann Clin Transl Neurol, 2019. 6(8): p. 1465–1479. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Perry VH and Gordon S, Macrophages and microglia in the nervous system. Trends Neurosci, 1988. 11(6): p. 273–7. [DOI] [PubMed] [Google Scholar]
- 33.Mracsko E and Veltkamp R, Neuroinflammation after intracerebral hemorrhage. Front Cell Neurosci, 2014. 8: p. 388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Aronowski J and Hall CE, New horizons for primary intracerebral hemorrhage treatment: experience from preclinical studies. Neurol Res, 2005. 27(3): p. 268–79. [DOI] [PubMed] [Google Scholar]
- 35.Wang J and Dore S, Inflammation after intracerebral hemorrhage. J Cereb Blood Flow Metab, 2007. 27(5): p. 894–908. [DOI] [PubMed] [Google Scholar]
- 36.Zhang D, et al. , Prostaglandin E2 released from activated microglia enhances astrocyte proliferation in vitro. Toxicol Appl Pharmacol, 2009. 238(1): p. 64–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Melton LM, et al. , Chronic glial activation, neurodegeneration, and APP immunoreactive deposits following acute administration of double-stranded RNA. Glia, 2003. 44(1): p. 1–12. [DOI] [PubMed] [Google Scholar]
- 38.Nakanishi H, Microglial functions and proteases. Mol Neurobiol, 2003. 27(2): p. 163–76. [DOI] [PubMed] [Google Scholar]
- 39.Shiratori M, et al. , P2X7 receptor activation induces CXCL2 production in microglia through NFAT and PKC/MAPK pathways. J Neurochem, 2010. 114(3): p. 810–9. [DOI] [PubMed] [Google Scholar]
- 40.Tessier PA, et al. , Chemokine networks in vivo: involvement of C-X-C and C-C chemokines in neutrophil extravasation in vivo in response to TNF-alpha. J Immunol, 1997. 159(7): p. 3595–602. [PubMed] [Google Scholar]
- 41.Starossom SC, et al. , Galectin-1 deactivates classically activated microglia and protects from inflammation-induced neurodegeneration. Immunity, 2012. 37(2): p. 249–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Chang CF, et al. , Alternative activation-skewed microglia/macrophages promote hematoma resolution in experimental intracerebral hemorrhage. Neurobiol Dis, 2017. 103: p. 54–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Wang J and Tsirka SE, Tuftsin fragment 1–3 is beneficial when delivered after the induction of intracerebral hemorrhage. Stroke, 2005. 36(3): p. 613–8. [DOI] [PubMed] [Google Scholar]
- 44.Wu J, et al. , Microglial activation and brain injury after intracerebral hemorrhage. Acta Neurochir Suppl, 2008. 105: p. 59–65. [DOI] [PubMed] [Google Scholar]
- 45.Jung KH, et al. , HMG-CoA reductase inhibitor, atorvastatin, promotes sensorimotor recovery, suppressing acute inflammatory reaction after experimental intracerebral hemorrhage. Stroke, 2004. 35(7): p. 1744–9. [DOI] [PubMed] [Google Scholar]
- 46.Wu J, et al. , Minocycline attenuates brain edema, brain atrophy and neurological deficits after intracerebral hemorrhage. Acta Neurochir Suppl, 2010. 106: p. 147–50. [DOI] [PubMed] [Google Scholar]
- 47.Yu SJ, et al. , Protective Effect of CXCR4 Antagonist CX807 in a Rat Model of Hemorrhagic Stroke. Int J Mol Sci, 2020. 21(19). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Hickenbottom SL, et al. , Nuclear factor-kappaB and cell death after experimental intracerebral hemorrhage in rats. Stroke, 1999. 30(11): p. 2472–7; discussion 2477–8. [DOI] [PubMed] [Google Scholar]
- 49.Leira R, et al. , Early neurologic deterioration in intracerebral hemorrhage: predictors and associated factors. Neurology, 2004. 63(3): p. 461–7. [DOI] [PubMed] [Google Scholar]
- 50.Platt N, da Silva RP, and Gordon S, Recognizing death: the phagocytosis of apoptotic cells. Trends Cell Biol, 1998. 8(9): p. 365–72. [DOI] [PubMed] [Google Scholar]
- 51.Zhao X, et al. , Distinct patterns of intracerebral hemorrhage-induced alterations in NF-kappaB subunit, iNOS, and COX-2 expression. J Neurochem, 2007. 101(3): p. 652–63. [DOI] [PubMed] [Google Scholar]
- 52.Mayne M, et al. , Adenosine A2A receptor activation reduces proinflammatory events and decreases cell death following intracerebral hemorrhage. Ann Neurol, 2001. 49(6): p. 727–35. [DOI] [PubMed] [Google Scholar]
- 53.Gong C, Hoff JT, and Keep RF, Acute inflammatory reaction following experimental intracerebral hemorrhage in rat. Brain Res, 2000. 871(1): p. 57–65. [DOI] [PubMed] [Google Scholar]
- 54.Chu K, et al. , Celecoxib induces functional recovery after intracerebral hemorrhage with reduction of brain edema and perihematomal cell death. J Cereb Blood Flow Metab, 2004. 24(8): p. 926–33. [DOI] [PubMed] [Google Scholar]
- 55.Li M, et al. , Colony stimulating factor 1 receptor inhibition eliminates microglia and attenuates brain injury after intracerebral hemorrhage. J Cereb Blood Flow Metab, 2017. 37(7): p. 2383–2395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Taylor RA and Sansing LH, Microglial responses after ischemic stroke and intracerebral hemorrhage. Clin Dev Immunol, 2013. 2013: p. 746068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Lin S, et al. , Heme activates TLR4-mediated inflammatory injury via MyD88/TRIF signaling pathway in intracerebral hemorrhage. J Neuroinflammation, 2012. 9: p. 46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Figueiredo RT, et al. , Characterization of heme as activator of Toll-like receptor 4. J Biol Chem, 2007. 282(28): p. 20221–9. [DOI] [PubMed] [Google Scholar]
- 59.Smiley ST, King JA, and Hancock WW, Fibrinogen stimulates macrophage chemokine secretion through toll-like receptor 4. J Immunol, 2001. 167(5): p. 2887–94. [DOI] [PubMed] [Google Scholar]
- 60.Sansing LH, et al. , Toll-like receptor 4 contributes to poor outcome after intracerebral hemorrhage. Ann Neurol, 2011. 70(4): p. 646–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Foss CA, et al. , Noninvasive molecular imaging of tuberculosis-associated inflammation with radioiodinated DPA-713. J Infect Dis, 2013. 208(12): p. 2067–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Rodriguez-Yanez M, et al. , Increased expression of Toll-like receptors 2 and 4 is associated with poor outcome in intracerebral hemorrhage. J Neuroimmunol, 2012. 247(1–2): p. 75–80. [DOI] [PubMed] [Google Scholar]
- 63.Dutra FF, et al. , Hemolysis-induced lethality involves inflammasome activation by heme. Proc Natl Acad Sci U S A, 2014. 111(39): p. E4110–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Xiao L, et al. , Neuroinflammation Mediated by NLRP3 Inflammasome After Intracerebral Hemorrhage and Potential Therapeutic Targets. Mol Neurobiol, 2020. 57(12): p. 5130–5149. [DOI] [PubMed] [Google Scholar]
- 65.Yao ST, et al. , NLRP3 is Required for Complement-Mediated Caspase-1 and IL-1beta Activation in ICH. J Mol Neurosci, 2017. 61(3): p. 385–395. [DOI] [PubMed] [Google Scholar]
- 66.Wan S, et al. , Microglia Activation and Polarization After Intracerebral Hemorrhage in Mice: the Role of Protease-Activated Receptor-1. Transl Stroke Res, 2016. 7(6): p. 478–487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Joice SL, et al. , Modulation of blood-brain barrier permeability by neutrophils: in vitro and in vivo studies. Brain Res, 2009. 1298: p. 13–23. [DOI] [PubMed] [Google Scholar]
- 68.Boje KM and Arora PK, Microglial-produced nitric oxide and reactive nitrogen oxides mediate neuronal cell death. Brain Res, 1992. 587(2): p. 250–6. [DOI] [PubMed] [Google Scholar]
- 69.Colton CA and Gilbert DL, Production of superoxide anions by a CNS macrophage, the microglia. FEBS Lett, 1987. 223(2): p. 284–8. [DOI] [PubMed] [Google Scholar]
- 70.Giulian D, Reactive glia as rivals in regulating neuronal survival. Glia, 1993. 7(1): p. 102–10. [DOI] [PubMed] [Google Scholar]
- 71.Wasserman JK and Schlichter LC, Neuron death and inflammation in a rat model of intracerebral hemorrhage: effects of delayed minocycline treatment. Brain Res, 2007. 1136(1): p. 208–18. [DOI] [PubMed] [Google Scholar]
- 72.Qureshi AI, et al. , Quantitative analysis of injured, necrotic, and apoptotic cells in a new experimental model of intracerebral hemorrhage. Crit Care Med, 2001. 29(1): p. 152–7. [DOI] [PubMed] [Google Scholar]
- 73.Qureshi AI, et al. , Apoptosis as a form of cell death in intracerebral hemorrhage. Neurosurgery, 2003. 52(5): p. 1041–7; discussion 1047–8. [PubMed] [Google Scholar]
- 74.Nakashima K, et al. , Temporal and spatial profile of apoptotic cell death in transient intracerebral mass lesion of the rat. J Neurotrauma, 1999. 16(2): p. 143–51. [DOI] [PubMed] [Google Scholar]
- 75.Felberg RA, et al. , Cell death in experimental intracerebral hemorrhage: the “black hole” model of hemorrhagic damage. Ann Neurol, 2002. 51(4): p. 517–24. [DOI] [PubMed] [Google Scholar]
- 76.Bobinger T, et al. , Programmed Cell Death after Intracerebral Hemorrhage. Curr Neuropharmacol, 2018. 16(9): p. 1267–1281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Fei X, et al. , The role of Toll-like receptor 4 in apoptosis of brain tissue after induction of intracerebral hemorrhage. J Neuroinflammation, 2019. 16(1): p. 234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Wang YX, et al. , Nuclear factor-kappaB and apoptosis in patients with intracerebral hemorrhage. J Clin Neurosci, 2011. 18(10): p. 1392–5. [DOI] [PubMed] [Google Scholar]
- 79.Wu B, et al. , Ac-YVAD-CMK Decreases Blood-Brain Barrier Degradation by Inhibiting Caspase-1 Activation of Interleukin-1beta in Intracerebral Hemorrhage Mouse Model. Transl Stroke Res, 2010. 1(1): p. 57–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Lan X, et al. , Modulators of microglial activation and polarization after intracerebral haemorrhage. Nat Rev Neurol, 2017. 13(7): p. 420–433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Kobayashi K, et al. , Minocycline selectively inhibits M1 polarization of microglia. Cell Death Dis, 2013. 4: p. e525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Castillo J, et al. , Molecular signatures of brain injury after intracerebral hemorrhage. Neurology, 2002. 58(4): p. 624–9. [DOI] [PubMed] [Google Scholar]
- 83.Fang HY, Ko WJ, and Lin CY, Inducible heat shock protein 70, interleukin-18, and tumor necrosis factor alpha correlate with outcomes in spontaneous intracerebral hemorrhage. J Clin Neurosci, 2007. 14(5): p. 435–41. [DOI] [PubMed] [Google Scholar]
- 84.Lei B, et al. , Tumor necrosis factor alpha antagonism improves neurological recovery in murine intracerebral hemorrhage. J Neuroinflammation, 2013. 10: p. 103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Zazulia AR, et al. , Progression of mass effect after intracerebral hemorrhage. Stroke, 1999. 30(6): p. 1167–73. [DOI] [PubMed] [Google Scholar]
- 86.Appelboom G, et al. , Volume-dependent effect of perihaematomal oedema on outcome for spontaneous intracerebral haemorrhages. J Neurol Neurosurg Psychiatry, 2013. 84(5): p. 488–93. [DOI] [PubMed] [Google Scholar]
- 87.Yang J, et al. , Prognostic significance of perihematomal edema in acute intracerebral hemorrhage: pooled analysis from the intensive blood pressure reduction in acute cerebral hemorrhage trial studies. Stroke, 2015. 46(4): p. 1009–13. [DOI] [PubMed] [Google Scholar]
- 88.Balami JS and Buchan AM, Complications of intracerebral haemorrhage. Lancet Neurol, 2012. 11(1): p. 101–18. [DOI] [PubMed] [Google Scholar]
- 89.Wang H, Ubl JJ, and Reiser G, Four subtypes of protease-activated receptors, co-expressed in rat astrocytes, evoke different physiological signaling. Glia, 2002. 37(1): p. 53–63. [DOI] [PubMed] [Google Scholar]
- 90.Suo Z, et al. , Participation of protease-activated receptor-1 in thrombin-induced microglial activation. J Neurochem, 2002. 80(4): p. 655–66. [DOI] [PubMed] [Google Scholar]
- 91.Zhang Y, et al. , Stereotactic Administration of Edaravone Ameliorates Collagenase-Induced Intracerebral Hemorrhage in Rat. CNS Neurosci Ther, 2016. 22(10): p. 824–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Lan X, et al. , Pinocembrin protects hemorrhagic brain primarily by inhibiting toll-like receptor 4 and reducing M1 phenotype microglia. Brain Behav Immun, 2017. 61: p. 326–339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Jiang C, et al. , Immune changes in peripheral blood and hematoma of patients with intracerebral hemorrhage. FASEB J, 2020. 34(2): p. 2774–2791. [DOI] [PubMed] [Google Scholar]
- 94.Ohnishi M, et al. , Gadolinium causes M1 and M2 microglial apoptosis after intracerebral haemorrhage and exerts acute neuroprotective effects. J Pharm Pharmacol, 2020. 72(5): p. 709–718. [DOI] [PubMed] [Google Scholar]
- 95.Ma Y, et al. , Matrix metalloproteinase-28 deletion exacerbates cardiac dysfunction and rupture after myocardial infarction in mice by inhibiting M2 macrophage activation. Circ Res, 2013. 112(4): p. 675–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Philipp D, et al. , Preconditioning of bone marrow-derived mesenchymal stem cells highly strengthens their potential to promote IL-6-dependent M2b polarization. Stem Cell Res Ther, 2018. 9(1): p. 286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Zheng J, et al. , AdipoRon Attenuates Neuroinflammation After Intracerebral Hemorrhage Through AdipoR1-AMPK Pathway. Neuroscience, 2019. 412: p. 116–130. [DOI] [PubMed] [Google Scholar]
- 98.Li Z, et al. , Hematoma Expansion in Intracerebral Hemorrhage: An Update on Prediction and Treatment. Front Neurol, 2020. 11: p. 702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Flores JJ, et al. , PPARgamma-induced upregulation of CD36 enhances hematoma resolution and attenuates long-term neurological deficits after germinal matrix hemorrhage in neonatal rats. Neurobiol Dis, 2016. 87: p. 124–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Ni W, et al. , Role of Erythrocyte CD47 in Intracerebral Hematoma Clearance. Stroke, 2016. 47(2): p. 505–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Zhou K, et al. , Regulatory T cells ameliorate intracerebral hemorrhage-induced inflammatory injury by modulating microglia/macrophage polarization through the IL-10/GSK3beta/PTEN axis. J Cereb Blood Flow Metab, 2017. 37(3): p. 967–979. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 102.Yang J, et al. , Interleukin-4 Ameliorates the Functional Recovery of Intracerebral Hemorrhage Through the Alternative Activation of Microglia/Macrophage. Front Neurosci, 2016. 10: p. 61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Fang H, et al. , Toll-like receptor 4 signaling in intracerebral hemorrhage-induced inflammation and injury. J Neuroinflammation, 2013. 10: p. 27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Wei J, et al. , Multinucleated Giant Cells in Experimental Intracerebral Hemorrhage. Transl Stroke Res, 2020. 11(5): p. 1095–1102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Schallner N, et al. , Microglia regulate blood clearance in subarachnoid hemorrhage by heme oxygenase-1. J Clin Invest, 2015. 125(7): p. 2609–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Koeppen AH, Dickson AC, and Smith J, Heme oxygenase in experimental intracerebral hemorrhage: the benefit of tin-mesoporphyrin. J Neuropathol Exp Neurol, 2004. 63(6): p. 587–97. [DOI] [PubMed] [Google Scholar]
- 107.Li Q, et al. , Neuroprotection of brain-permeable iron chelator VK-28 against intracerebral hemorrhage in mice. J Cereb Blood Flow Metab, 2017. 37(9): p. 3110–3123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Ransohoff RM, A polarizing question: do M1 and M2 microglia exist? Nat Neurosci, 2016. 19(8): p. 987–91. [DOI] [PubMed] [Google Scholar]
- 109.Martinez FO and Gordon S, The M1 and M2 paradigm of macrophage activation: time for reassessment. F1000Prime Rep, 2014. 6: p. 13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Kanazawa M, et al. , Microglia and Monocytes/Macrophages Polarization Reveal Novel Therapeutic Mechanism against Stroke. Int J Mol Sci, 2017. 18(10). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Morganti JM, Riparip LK, and Rosi S, Call Off the Dog(ma): M1/M2 Polarization Is Concurrent following Traumatic Brain Injury. PLoS One, 2016. 11(1): p. e0148001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Kim CC, Nakamura MC, and Hsieh CL, Brain trauma elicits non-canonical macrophage activation states. J Neuroinflammation, 2016. 13(1): p. 117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Zhao H, et al. , Microglia/Macrophage Polarization After Experimental Intracerebral Hemorrhage. Transl Stroke Res, 2015. 6(6): p. 407–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Blackburn D, et al. , Astrocyte function and role in motor neuron disease: a future therapeutic target? Glia, 2009. 57(12): p. 1251–64. [DOI] [PubMed] [Google Scholar]
- 115.Sukumari-Ramesh S, Alleyne CH Jr., and Dhandapani KM, Astrocyte-specific expression of survivin after intracerebral hemorrhage in mice: a possible role in reactive gliosis? J Neurotrauma, 2012. 29(18): p. 2798–804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Scimemi A, Astrocytes and the Warning Signs of Intracerebral Hemorrhagic Stroke. Neural Plast, 2018. 2018: p. 7301623. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Shi SX, et al. , IL (Interleukin)-15 Bridges Astrocyte-Microglia Crosstalk and Exacerbates Brain Injury Following Intracerebral Hemorrhage. Stroke, 2020. 51(3): p. 967–974. [DOI] [PubMed] [Google Scholar]
- 118.He M, et al. , Astrocyte-Derived CCL2 is Associated with M1 Activation and Recruitment of Cultured Microglial Cells. Cell Physiol Biochem, 2016. 38(3): p. 859–70. [DOI] [PubMed] [Google Scholar]
- 119.Chang CF, et al. , Erythrocyte efferocytosis modulates macrophages towards recovery after intracerebral hemorrhage. J Clin Invest, 2018. 128(2): p. 607–624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.De Paola M, et al. , Chemokine MIP-2/CXCL2, acting on CXCR2, induces motor neuron death in primary cultures. Neuroimmunomodulation, 2007. 14(6): p. 310–6. [DOI] [PubMed] [Google Scholar]
- 121.Carmichael ST, et al. , Genomic profiles of damage and protection in human intracerebral hemorrhage. J Cereb Blood Flow Metab, 2008. 28(11): p. 1860–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Matsushita H, et al. , Suppression of CXCL2 upregulation underlies the therapeutic effect of the retinoid Am80 on intracerebral hemorrhage in mice. J Neurosci Res, 2014. 92(8): p. 1024–34. [DOI] [PubMed] [Google Scholar]
- 123.Jung JE, et al. , The Mitochondria-Derived Peptide Humanin Improves Recovery from Intracerebral Hemorrhage: Implication of Mitochondria Transfer and Microglia Phenotype Change. J Neurosci, 2020. 40(10): p. 2154–2165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Cordeiro JL, et al. , Arundic Acid (ONO-2506) Attenuates Neuroinflammation and Prevents Motor Impairment in Rats with Intracerebral Hemorrhage. Cell Mol Neurobiol, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Liddelow SA, et al. , Neurotoxic reactive astrocytes are induced by activated microglia. Nature, 2017. 541(7638): p. 481–487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Lei C, et al. , High-mobility group box1 protein promotes neuroinflammation after intracerebral hemorrhage in rats. Neuroscience, 2013. 228: p. 190–9. [DOI] [PubMed] [Google Scholar]
- 127.Zhou Y, et al. , Elevation of high-mobility group protein box-1 in serum correlates with severity of acute intracerebral hemorrhage. Mediators Inflamm, 2010. 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Ohnishi M, et al. , HMGB1 inhibitor glycyrrhizin attenuates intracerebral hemorrhage-induced injury in rats. Neuropharmacology, 2011. 61(5–6): p. 975–80. [DOI] [PubMed] [Google Scholar]
- 129.Lei C, et al. , Activation of the high-mobility group box 1 protein-receptor for advanced glycation end-products signaling pathway in rats during neurogenesis after intracerebral hemorrhage. Stroke, 2015. 46(2): p. 500–6. [DOI] [PubMed] [Google Scholar]
- 130.Loftspring MC, et al. , Intracerebral hemorrhage leads to infiltration of several leukocyte populations with concomitant pathophysiological changes. J Cereb Blood Flow Metab, 2009. 29(1): p. 137–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Hammond MD, et al. , CCR2+ Ly6C(hi) inflammatory monocyte recruitment exacerbates acute disability following intracerebral hemorrhage. J Neurosci, 2014. 34(11): p. 3901–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Zhou Y, et al. , Inflammation in intracerebral hemorrhage: from mechanisms to clinical translation. Prog Neurobiol, 2014. 115: p. 25–44. [DOI] [PubMed] [Google Scholar]
- 133.Zhong Q, et al. , Interleukin-23 Secreted by Activated Macrophages Drives gammadeltaT Cell Production of Interleukin-17 to Aggravate Secondary Injury After Intracerebral Hemorrhage. J Am Heart Assoc, 2016. 5(10). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Min H, et al. , Alternatively activated brain-infiltrating macrophages facilitate recovery from collagenase-induced intracerebral hemorrhage. Mol Brain, 2016. 9: p. 42. [DOI] [PMC free article] [PubMed] [Google Scholar]