Next Article in Journal
Forensic Tools for Species Identification of Skeletal Remains: Metrics, Statistics, and OsteoID
Next Article in Special Issue
Function and Plasticity of Electrical Synapses in the Mammalian Brain: Role of Non-Junctional Mechanisms
Previous Article in Journal
Small Angle X-ray Scattering Sensing Membrane Composition: The Role of Sphingolipids in Membrane-Amyloid β-Peptide Interaction
Previous Article in Special Issue
First Responders to Hyperosmotic Stress in Murine Astrocytes: Connexin 43 Gap Junctions Are Subject to an Immediate Ultrastructural Reorganization
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Gap Junctions and Hemichannels Composed of Connexins and Pannexins Mediate the Secondary Brain Injury Following Intracerebral Hemorrhage

1
Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450014, China
2
Academy of Medical Science, Zhengzhou University, Zhengzhou 450001, China
3
Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Calgary, AB T3A 4X9, Canada
*
Authors to whom correspondence should be addressed.
Biology 2022, 11(1), 27; https://doi.org/10.3390/biology11010027
Submission received: 5 December 2021 / Revised: 19 December 2021 / Accepted: 24 December 2021 / Published: 25 December 2021

Abstract

:

Simple Summary

Intracerebral hemorrhage (ICH) is a leading medical problem without effective treatment options. The poor prognosis is attributed to the primary brain injury of the mechanical compression caused by hematoma, and secondary brain injury (SBI) that includes inflammation, glutamate excitotoxicity, oxidative stress and disruption of the blood brain barrier (BBB). Evidences suggests that gap junctions and hemichannels composed of connexins and pannexins regulate the inflammation and excitotoxicity insult in the pathological process of central nervous system disease, such as cerebral ischemia and neurodegeneration disease. In this manuscript, we discuss the fact that connexins- and pannexins-based channels could be involved in secondary brain injury of ICH, particularly through mediating inflammation, oxidative stress, BBB disruption and cell death. The details provided in this manuscript may help develop potential targets for therapeutic intervention of ICH.

Abstract

Intracerebral hemorrhage (ICH) is a devastating disease with high mortality and morbidity; the mortality rate ranges from 40% at 1 month to 54% at 1 year; only 12–39% achieve good outcomes and functional independence. ICH affects nearly 2 million patients worldwide annually. In ICH development, the blood leakage from ruptured vessels generates sequelae of secondary brain injury (SBI). This mechanism involves activated astrocytes and microglia, generation of reactive oxygen species (ROS), the release of reactive nitrogen species (RNS), and disrupted blood brain barrier (BBB). In addition, inflammatory cytokines and chemokines, heme compounds, and products of hematoma are accumulated in the extracellular spaces, thereby resulting in the death of brain cells. Recent evidence indicates that connexins regulate microglial activation and their phenotypic transformation. Moreover, communications between neurons and glia via gap junctions have crucial roles in neuroinflammation and cell death. A growing body of evidence suggests that, in addition to gap junctions, hemichannels (composed of connexins and pannexins) play a key role in ICH pathogenesis. However, the precise connection between connexin and pannexin channels and ICH remains to be resolved. This review discusses the pathological roles of gap junctions and hemichannels in SBI following ICH, with the intent of discovering effective therapeutic options of strategies to treat ICH.

1. Introduction

ICH refers to blood entering into the brain parenchyma, ventricle system, or subarachnoid space from a rupturing cerebral vessel. ICH causes a huge burden to patients and to society. The mortality rate in ICH patients is approximately 50%, while most of the survivors lose the capability of living independently [1,2]. Notably, secondary brain injury (SBI), which refers to inflammation [3], oxidative stress [4], cerebral vasospasm [5], and blood brain barrier (BBB) hyperpermeability [6], further drives brain cell death. SBI is a major cause of the high mortality and morbidity in ICH patients. Until now, only limited treatment options are available for ICH, therefore novel therapeutic options are needed to be developed [7,8,9].
Gap junctions and hemichannels composed of connexins (Cx) play a crucial role in the ICH pathology. A hemichannel or connexon comprised of six connexin proteins allows approximately 1kDa sized molecules to pass through the membrane. These molecules include metabolites, ATP, ADP, cAMP, Ca2+, nutrients, second messengers, and adenosine. The connexons on the membrane are docked with a connexon on a neighboring cell resulting in a gap junction channel, which then allows passage of ions into adjacent cells [10] in order to modulate the communications between cells or the extracellular environment and intracellular environment [11]. The gating of hemichannels and gap junctions is regulated by different factors, such as trans-junctional/transmembrane voltage, changes of extracellular and intracellular Ca2+, mechanical stimulation, changes in phosphorylation, reactive oxygen species (ROS), and nitrosylation [12]. Moreover, pannexins are also important channels that can form structurally similar channels to connexins. Pannexins are permeable to molecules below 900 Da, such as nucleotides and ions [13]. At least one of the three known proteins, Panx1, Panx2, and Panx3, has been found in every organ in mammals.
Previous evidence has shown that pannexin and connexin play a critical role in the pathological process of cerebrovascular diseases, such as cerebral ischemia [14] and neurodegeneration disease [15], mainly through regulating inflammatory and excitotoxic insults. The precise role of connexin and pannexin channels in ICH has not been well understood yet. The aim of our current review is to outline the possible roles of connexin and pannexin channels in SBI following ICH.

2. The Expression and Functions of Connexins and Pannexins in the Brain

Connexins and pannexins are abundantly expressed in the CNS. All neurons express Cx36 and Cx45; astrocytes, the most numerous cells in CNS are coupled via gap junctions, which primarily express Cx43 and Cx30; microglia predominantly express Cx46, Cx32, Cx36, and Cx43. Oligodendrocytes (the myelinating cells of the CNS) are known for their ability of extensive connectivity with astrocytes, as well as with each other, and they chiefly express Cx29, Cx32, and Cx47 [16]. Endothelial cells mainly express Cx37 and Cx40 [17]. Until now, three pannexins have been identified, namely Panx1, Panx2, and Panx3, with Panx1 and Panx2 being widely expressed in the CNS [15].

2.1. The Expression and Function of Gap Junction Channels in the Brain

Under physiology conditions, gap junction channels between brain cells exert critical functions in maintaining the brain’s homeostasis. Gap junctions between neurons form electrical synapses, which play an important functional role in brain cell synchronization [18]. On the other hand, carbenoxolone, a general gap junction blocker, inhibits spikelets induced by local stimulation of stratum oriens, thereby decreasing the rate of firing antidromic action potential, which contributes to early events generation in the hippocampus of neonates [19]. Besides, Cx36 (neuronal gap junction protein) increases the junctional conductance by 10-fold or even more within ten minutes after cell break-in with pipettes [20]. Moreover, neuronal gap junctions induce memory and play an important role in learning.
Oligodendrocytes and astrocytes form few gap junctions essential for homeostatic and nutrient support. Specific transporters allow glucose molecules to enter from blood to the astrocytic end-feet. These glucose molecules are then transported from one astrocyte to another through Cx43/Cx30 gap junctions. After their release in the proximity of neurons, uptake occurs. In addition, gap junctions coupled with the astrocyte network are essential for extracellular potassium ion (K+) clearance. After uncoupling of the astrocytes, hampering of K+ clearance takes place, while extracellular K+ accumulation causes hyperexcitability of neurons. Accumulating evidence indicates that the defective control of ion/fluid exchanges of astrocytes may cause brain edema, fluid cysts, and astrocyte/myelin vacuolation [21]. Furthermore, Cx30 and Cx43 of the astrocytes are normally distributed in perivascular end-feet; Cx30 and Cx43 double knockout of astrocytes induce end-feet swelling [22].
While confronting pathological insults, connexin-based gap junction channels may exhibit opposite roles depending on the disease conditions. A substantial body of evidence indicates that gap junction channels may mediate cell death through a mechanism termed ‘bystander death’ or the ‘kiss of death’, which describes the death signal that propagates to neighboring cells through gap junctions [12]. Evidence is accumulating that Ca2+ ions are the most probable cell-killing signals spread through gap junctions [23]. Moreover, except for the large molecules of cytochrome C, apoptosis inducer Ca2+ and IP3 have also been shown to spread via gap junctions to exacerbate cell apoptosis cascades. Earlier research had found that upregulation of the proto-oncogene b-cell leukemia/lymphoma 2 (Bcl2) increases resistance to injury, however, the gap junction formation with vulnerable cells leads to compromised resistance to metabolic inhibition, calcium overload, and oxidative stress [24]. Knockdown Cx36-containing gap junctions prevent neuronal death from ischemia [25], while over-expression of Cx36 results in increased neuronal death [25]. Consistent with this phenomenon, the inhibition of the gap junction (astrocytic) increases the protective effects following cerebral ischemia [26], while the gap junction blockers modulate seizure-linked behavioral parameters [27].
By contrast, several investigations show that gap junctions can attenuate brain cell death by distributing intracellular toxic substances to healthy neighbors. Besides, potential ‘rescue messengers’ such as ATP, energy molecules, and reduced glutathione, and ascorbic acid can also flow through gap junctions in order to induce cell survival [12]. Gap junctional intercellular communication (GJIC) promotes reversal of ischemia-mediated hippocampal apoptosis and cognitive impairment. While GJIC suppression facilitates hippocampal apoptosis and cognitive impairment [28].
These results suggest that the occurrence of two-way traffic between injured cells possibly depends on the different stages of pathological process and signal-receiving cellular environment.

2.2. The Expression and Function of Connexin and Pannexin Hemichannels in the Brain

Connexin and pannexin mediated communications of neurons widely occur during brain development. In late prenatal neurogenesis, gap junctions can control the migration and division of precursor cells [29]. Connexin hemichannels are thought to remain closed, primarily while pannexin channels are active under normal physiological conditions. However, the pannexin channels would contribute to ATP release and the connexin undocked hemichannel may take on a prolonged or more frequently open state following a pathology insult. These functions may further induce cell death through depolarization of the membrane, loss of small metabolites, cytoplasmic Ca2+ elevation, and the ionic gradients’ collapse [30].
It has been proposed that inflammation is associated with the opening of unopposed hemichannels [31]. ATP released from hemichannels leads to autocrine activation of purinergic P2X receptors (P2X). P2X7 then activates pyrin domain-containing-3 (NLRP3) inflammasome, the nucleotide-binding oligomerization domain-like receptor, and enhances the inflammatory effect, which then leads to increased Cx43 expression, opening hemichannels, as well as higher ATP release [32]. Thus, blockade of hemichannel expression or inhibiting P2X7 receptors during neuroinflammation might prevent neuronal damage [33]. Hemichannels also release glutamate, which is known as a paracrine messenger of cell death [34]. Orellana et al. pointed out that glutamate and ATP released by astroglial Cx43 hemichannels activate Panx1 hemichannels, which facilitate neuronal death [35]. Moreover, cell death also was accelerated by the metabolic inhibition of astrocytes through the opening of Cx43 hemichannel [36]. Hence, the abnormal opening of connexin and pannexin hemichannels would predominantly lead to detrimental effects in the brain.

3. Connexin and Pannexin Channels in ICH

The poor prognosis of ICH may attribute to the primary brain injury that, due to the mechanical compression, caused by hematoma and second brain injury, mainly including inflammation and oxidative stress, which is mediated through a series of events induced by primary injury and releasing of clot components [2]. When a blood vessel in the brain ruptures, local cerebral blood flow and cerebral perfusion pressure may drop while intracranial pressure increases, which would induce cerebral vasospasm and cerebral ischemia [37]. While blood leaks into the brain parenchyma, the hematoma and the degradation products of erythrocytes (such as hemoglobin, heme and iron), and complement components (mainly C3a and C5a) activate microglia. The invasion of neutrophils may lead to the release of toxic substances such as thrombin, ROS and matrix metalloproteinases (MMPs). Collectively, the result is neuroinflammation, neuronal and glial cell death, vasogenic edema, and further breakdown of the BBB [38]. Based on the impact of connexin and pannexin channels in brain pathology discussed above, the subsequent paragraphs will explore the effects of connexin and pannexin channels in SBI following ICH.

3.1. Connexin and Pannexin Channels Are Implicated in Neuroinflammation Following ICH

Following ICH or primary injury, a proinflammatory cascade in the peri-hematoma induces neural cell death. This proinflammatory cascade is composed of infiltrated leucocytes, activated astrocytes, and microglia [3,4,5,6,7,8,9,10,11,12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39], and is associated with the activity of connexin channels.
During the development and progression of ICH, hematoma degradation products (hemin and hemoglobin) activate astrocytes [40], which are then accumulated in the surrounding area and release chemokines and cytokines to induce inflammatory responses, neuronal apoptosis and destruction of BBB [41]. Further inflammatory factors are released by the excessive opening of Cx43 hemichannels, which are present on the surface of reactive astrocytes, in order to aggravate the inflammatory response and activate the immune system at the damaged area [42]. In the ICH model inflicted by collagenase IV injection into the brain, Cx43 upregulation and excessive Cx43 hemichannel opening have been observed [43]. Furthermore, downregulated Cx43 and nuclear translocated YAP play a role in hemoglobin-activated astrocytes and are interdependent, such that reducing Cx43 induces YAP nuclear translocation [44].
Microglia are key players in ICH mediated inflammatory responses [45]. Microglia can be activated by hematoma degradation products and fibrinogen in the hematoma [46]. Activated microglia release IL−1β, IL−6 and TNF-α molecules in order to propagate inflammatory responses and promote the recruitment of leukocytes. They also facilitate the release of chemokines such as CCL2, CCL5, CXCL8 in order to attract monocytes, lymphocytes, and neutrophils [38]. The expression levels of Cx43 and Cx36 are comparatively very minimally produced during the resting surveillance state of microglia. However, these expression levels are increased after the activation of microglia by pro-inflammatory conditions. More importantly, the released proinflammatory cytokines from activated microglia impede GJIC and activate Cx43 hemichannels in astrocytes [47] (Figure 1). Extracellular ATP released by the opening of hemichannels leads to the activation of purinergic receptors P2X7R and P2X4R, which can further activate NRLP3 pro-inflammatory inflammasome pathway in microglia to exacerbate inflammation responses [48] (Figure 1). Hence, connexin based channels play an important role in inflammatory injury mediated by activated microglia, and purinergic signaling would be partly involved in determining the activation state of microglia. Besides, Cx43 hemichannels mediated release of gliotransmitters (ATP/glutamate) may also result in the opening of Panx1 channels and Cx36 hemichannels in neurons. Consequent neuronal Ca2+ overload can lead to numerous deleterious consequences, including structural neuronal alterations and increased oxidative stress [30] (Figure 1).
At the same time, gap junction channels can be regulated by pro-inflammatory cytokines. Pro-inflammatory cytokines and ATP (10–100 µM) induce a rapid and concentration-dependent inhibition of GJIC in cultured cortical astrocytes [49]. IL−1β in combination with TNF leads to a robust decrease in astroglial coupling [50]. Moreover, IL−1β treatment also reduces wave propagation of calcium between astrocytes [51]. Pro-inflammatory cytokines mediated GJIC inhibition reduces death signal molecules or the spread of toxic substances, thereby preventing impairment and survival of neurons. Correspondent with this idea, astroglial GJIC contributes to the propagation of death signals partly by activating p38/stress-activated protein kinase 2 following CNS injury [52].

3.2. Connexin Channels Mediate the Permeability of BBB Following ICH

The integrity of BBB is maintained by gap junctions, tight junctions, and adherens junctions, which constitute a unique junction complex. The expression of connexin-based channels takes place throughout the BBB as these connexins are produced by pericytes and endothelial cells [6]. Evidence suggests that connexin-mediated GJIC regulates the integrity and maintains the normal function of the BBB [14]. Non-specific pannexin and connexin blockers inhibit the barrier’s function, suggesting that these molecules induce the progression of ICH, which disrupts the BBB, as well as increase permeability in paracellular and transcellular routes, thereby leading to vascular leakage. Recently, Cx43 upregulation and excessive Cx43 hemichannel opening were observed in mice after ICH injury [43], which may confer detrimental barrier permeability. Moreover, connexin hemichannels may increase BBB permeability by releasing ATP and glutamate after ICH. Allison et al. demonstrated that Cx43 gap junctions play a crucial role in the hyperpermeability of the endothelial barrier by modulating the structure of tight junction [53]. Similar to this finding, another group showed that Cx36 may interact with the PDZ domain-containing protein zonula occludens-1 (ZO-1), ZO-2 and ZO−3 [54]. Further investigations confirmed that increasing p-Cx43 altered the integrity of the BBB through the activation of the PI3K and ERK pathways in recombinant tissue plasminogen activator (rtPA)-associated brain ischemia hemorrhagic transformation [55]. Since propagation of Ca2+ affects the integrity of cytoskeleton or endothelial cell function, therefore elevated GJIC is thought to be pathogenic to the function of BBB [56]. Besides, the excessive opening of Cx43 hemichannels following hypoxia would directly mediate BBB hyperpermeability by resulting endothelial cell death [57].

3.3. Connexin Channels Are Involved in Oxidative Stress Following ICH

Following ICH progression, the accumulation of ROS induces oxidative stress, which contributes to SBI caused by inflammation and BBB disruption [4]. Interestingly, ROS/RNS are involved in regulating the change of connexin and pannexin-based channel properties, both in systemic vasculature and brain cells [58,59]. Moreover, oxidation products were shown to increase hemichannel activity whilst reducing GJIC [60]. Thus, antioxidant therapy may inhibit hemichannel activity [61] and attenuate apoptosis [62]. As mentioned above, Cx43 gap junctions between astrocytes essentially retain homeostasis. The absence of expression or its channels blockage induces ROS-mediated death of astrocytes, and Cx43-mediated gap junctions in astrocytes positively affect oxidative stress resistance [60]. Recent studies have found that astrocyte mediated apoptosis disrupts homeostasis simultaneously, as well as downregulated Cx43, which in turn lead to the conformational changes in Cx43, closing and degradation of the channels. These can further prevent Nrf2 nuclear translocation and protein kinase C alpha (PKCα) phosphorylation to antioxidant stress. Cx43 helps liberate Nrf2 from Kelch-like ECH-associated protein 1, as well as allows the nuclear translocation in order to promote genes encoded for phase II detoxification enzymes, which are involved in processes of anti-apoptosis and antioxidant stress [63] (Figure 2). According to these findings, it seems that hemichannels and gap junctions in astrocytes may exert a beneficial effect on oxidative stress prevention through Nrf2 regulation.

3.4. Connexin Channels Take Part in Regulating Cerebral Vasospasm Following ICH

Cerebral vasospasm, a severe complication of subarachnoid hemorrhage [64], is a well known condition, in which vasomotion requires gap junctions for intercellular communications. Cx 37 and 40 are mainly expressed by endothelial cells in healthy conduit arteries, while Cx43 and Cx45 are expressed by vascular smooth muscle cells [17]. Connexin channels are thought to be involved in the process of cerebral vasospasm, which is associated with subarachnoid hemorrhage. Increased phosphorylation of Cx43 via the p38MAPK and protein kinase C (PKC) pathways was found to mediate the development of cerebral vasospasm in animal models [65,66]. Moreover, experimental cerebral vasospasm is attenuated by gap junction blockers, which also down-regulate overexpressed Cx43 protein in subarachnoid hemorrhage [67]. However, Lan et al. showed a marked decrease in Cx40 after subarachnoid hemorrhage, upregulation of Cx40 mediated by nitric oxide attenuated cerebral vasospasm via the nitric oxide-cyclic guanosine monophosphate-protein kinase G pathway after subarachnoid hemorrhage [17]. Thus, different connexin subtypes may play various roles in vasospasm following subarachnoid hemorrhage.

3.5. Hemichannels Are Involved in Cell Death Following ICH

The local concentration of glutamate is highly elevated following ICH, which subsequently overstimulates N-methyl-D-aspartate receptors (NMDARs) to increase intracellular Ca2+ and neuronal death [68]. Current evidence indicates that Panx1 channels can be activated by NMDARs via Src family kinases to induce neuronal apoptosis [69]. Furthermore, in rat primary spinal neurocytes, Panx1 overexpression enhances signaling of intracellular Ca2+, as well as upregulates Bax (apoptotic protein) levels, and apoptosis pathway proteins, including cleaved caspase-3 and PARP-1; conversely, Panx1 depletion reversed the pro-apoptosis effect [70]. It is important to highlight that the Panx1 is linked with arresting the metabolic activity of apoptotic cells through facilitating intracellular ATP loss [71]. Accordingly, Zhou and colleagues recently demonstrated that the expression of Panx1 significantly increased after ICH, while the peak level was noted at 48h post-ICH [72].

4. Strategies Target Gap Junctions and Hemichannels Following ICH

A growing body of evidence showed that hemichannels blocking by pharmaceutic preparation or genetic measures may improve neurological function following ICH. Gap19, a Cx43 mimetic peptide, significantly alleviated hematoma volume and neurological deficits after ICH injury by downregulating Cx43 and regulating YAP inflammation signaling in astrocytes [43]. Consistent with this result, multiple Cx43 inhibitors (eg, carbenoxolone and dynasore) were demonstrated to decrease hematoma volume and BBB disruption in ICH mice [73]. Most importantly, carbenoxolone, also as a Panx1 inhibitor, remarkably improved cognitive function, reduced brain edema and BBB injury in rats post-ICH. Moreover, it reduced the degenerative Fluoro-Jade B positive cells, inhibited caspase3 activation and attenuated TUNEL positive cells in the proximity of ICH hematoma [72]. In another investigation, carbenoxolone was reported to down-regulate Cx43 protein expression and attenuate the experimental cerebral vasospasm after subarachnoid hemorrhage in rabbits [67].
For a long time, GJIC was deemed to amplify the extent of injury by transmitting the signals of apoptotic and necrotic cells. Thus, blockade of gap junctions has been shown to provide neuroprotection in CNS diseases, such as cerebral ischemia [74]. However, administration of gap junction inhibitors, octanol and carbenoxolone, failed to attenuate the neurological deficits induced by subarachnoid hemorrhage, and they did not reduce neuronal apoptosis. On the contrary, carbenoxolone increased post subarachnoid hemorrhage mortality and exacerbated its apoptosis [75]. Similar results have been found in ICH mice, where a high dose of carbenoxolone aggravated neurological deficits and increased mortality 72 h after the treatment [76]. Moreover, carbenoxolone treatment deteriorated barrier permeability after ICH [76].
These pieces of evidence suggest that gap junction and hemichannel inhibitors may play a diverse role in different models and periods of ICH. In addition, the dosage of the medication is also an important factor affecting its neuroprotective effect.

5. Conclusions

This review summarized evidence for the roles that connexin and pannexin channels appear to play in ICH. Based on the data, we infer that connexin and pannexin-based channels are involved in the process of inflammation, oxidative stress, BBB disruption and cell death in ICH. Notably, the inhibition of both pannexin channel and connexin hemichannels appears beneficial following ICH, whereas the role of gap junction channel function is ambiguous, since gap junction channel uncoupling appears to be detrimental in BBB dysfunction and oxidative stress, but to be beneficial in reducing inflammatory responses. These data suggest that regulating gap junctions and hemichannels may lead to new therapeutic strategies against secondary brain injury induced by ICH. However, the most specific blockers currently available are mimetic peptides with sequences very similar to that of the extracellular loop of connexins. Therefore, specific channel blockers targeting certain connexin or pannexin monomer subtypes are urgently required. It is obvious that studies focusing on the association between connexin channels and ICH are sporadic and far from complete. The precise mechanism of connexin and pannexin channels to different metabolic products of hematoma remains indistinct, and further investigations are required to explore the temporal and spatial characteristics of connexin and pannexin channel responses after ICH. It is worth noting that understanding the precise pathologic roles of connexin and pannexin channels in ICH would offer better therapeutic options to reduce the dismal consequences of ICH.

Author Contributions

Conceptualisation, formal analysis, writing—original draft and writing—review and editing, Y.Z.; supervision, formal analysis and writing—review and editing, S.K.; writing—review and editing, Y.L., R.S. and R.Z.; formal analysis and writing—review and editing, M.X. and supervision and writing—review and editing, V.W.Y. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by operating grant support from National Key Research and Development Program of China (grant no: 2018YFC1312200), the National Natural Science Foundation of China (grants no: 82071331, 81870942, and 81520108011), and from the Canadian Institutes of Health Sciences (V.W.Y).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Please contact the corresponding author to discuss the availability of the data and materials.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Gross, B.A.; Jankowitz, B.T.; Friedlander, R.M. Cerebral Intraparenchymal Hemorrhage: A Review. JAMA 2019, 321, 1295–1303. [Google Scholar] [CrossRef]
  2. Chen, L.; Chen, T.; Mao, G.; Chen, B.; Li, M.; Zhang, H.; Xi, H.; She, X.; Tang, Z.; Zhang, P.; et al. Clinical Neurorestorative Therapeutic Guideline for Brainstem Hemorrhage (2020 China Version). J. Neurorestoratology 2020, 8, 232–240. [Google Scholar] [CrossRef]
  3. Xue, M.; Yong, V.W. Neuroinflammation in Intracerebral Haemorrhage: Immunotherapies with Potential for Translation. Lancet Neurol. 2020, 19, 1023–1032. [Google Scholar] [CrossRef]
  4. Yao, Z.; Bai, Q.; Wang, G. Mechanisms of Oxidative Stress and Therapeutic Targets Following Intracerebral Hemorrhage. Oxid. Med. Cell Longev. 2021, 2021, 8815441. [Google Scholar] [CrossRef] [PubMed]
  5. Athiraman, U.; Zipfel, G.J. Role of Anesthetics and their Adjuvants in Neurovascular Protection in Secondary Brain Injury after Aneurysmal Subarachnoid Hemorrhage. Int. J. Mol. Sci. 2021, 22, 6550. [Google Scholar] [CrossRef]
  6. Keep, R.F.; Andjelkovic, A.V.; Xiang, J.; Stamatovic, S.M.; Antonetti, D.A.; Hua, Y.; Xi, G. Brain Endothelial Cell Junctions After Cerebral Hemorrhage: Changes, Mechanisms and Therapeutic Targets. J. Cereb. Blood Flow Metab. 2018, 38, 1255–1275. [Google Scholar] [CrossRef] [PubMed]
  7. Bai, Q.; Sheng, Z.; Liu, Y.; Zhang, R.; Yong, V.W.; Xue, M. Intracerebral Haemorrhage: From Clinical Settings to Animal Models. Stroke Vasc. Neurol. 2020, 5, 388–395. [Google Scholar] [CrossRef] [Green Version]
  8. Guo, X.; Xue, Q.; Zhao, J.; Yang, Y.; Yu, Y.; Liu, D.; Liu, J.; Yang, W.; Mu, L.; Zhang, P.; et al. Clinical Diagnostic and Therapeutic Guidelines of Stroke Neurorestoration (2020 China Version). J. Neurorestoratology 2020, 8, 241–251. [Google Scholar] [CrossRef]
  9. Zhang, R.; Xue, M.; Yong, V.W. Central Nervous System Tissue Regeneration after Intracerebral Hemorrhage: The Next Frontier. Cells 2021, 10, 2513. [Google Scholar] [CrossRef] [PubMed]
  10. Li, X. Seeing is Believing: Gap Junctions in Motion. Biology 2021, 10, 494. [Google Scholar] [CrossRef]
  11. Orellana, J.A.; Martinez, A.D.; Retamal, M.A. Gap Junction Channels and Hemichannels in the Cns: Regulation by Signaling Molecules. Neuropharmacology 2013, 75, 567–582. [Google Scholar] [CrossRef] [PubMed]
  12. Decrock, E.; Vinken, M.; De Vuyst, E.; Krysko, D.V.; D’Herde, K.; Vanhaecke, T.; Vandenabeele, P.; Rogiers, V.; Ley-baert, L. Connexin-Related Signaling in Cell Death: To Live Or Let Die? Cell Death Differ. 2009, 16, 524–536. [Google Scholar] [CrossRef] [PubMed]
  13. Shestopalov, V.I.; Slepak, V.Z. Molecular Pathways of Pannexin1-Mediated Neurotoxicity. Front Physiol. 2014, 5, 23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Kim, Y.; Davidson, J.O.; Green, C.R.; Nicholson, L.; O’Carroll, S.J.; Zhang, J. Connexins and Pannexins in Cerebral Ischemia. Biochim. Biophys. Acta Biomembr. 2018, 1860, 224–236. [Google Scholar] [CrossRef] [PubMed]
  15. Bargiotas, P.; Krenz, A.; Hormuzdi, S.G.; Ridder, D.A.; Herb, A.; Barakat, W.; Penuela, S.; von Engelhardt, J.; Monyer, H.; Schwaninger, M. Pannexins in Ischemia-Induced Neurodegeneration. Proc. Natl. Acad. Sci. USA 2011, 108, 20772–20777. [Google Scholar] [CrossRef] [Green Version]
  16. Takeuchi, H.; Suzumura, A. Gap Junctions and Hemichannels Composed of Connexins: Potential Therapeutic Targets for Neurodegenerative Diseases. Front. Cell Neurosci. 2014, 8, 189. [Google Scholar] [CrossRef] [Green Version]
  17. Lan, S.H.; Lai, W.T.; Zheng, S.Y.; Yang, L.; Fang, L.C.; Zhou, L.; Tang, B.; Duan, J.; Hong, T. Upregulation of Connexin 40 Mediated by Nitric Oxide Attenuates Cerebral Vasospasm After Subarachnoid Hemorrhage Via the Nitric Oxide-Cyclic Guanosine Monophosphate-Protein Kinase G Pathway. World Neurosurg. 2020, 136, e476–e486. [Google Scholar] [CrossRef]
  18. Lynn, B.D.; Li, X.; Hormuzdi, S.G.; Griffiths, E.K.; McGlade, C.J.; Nagy, J.I. E3 Ubiquitin Ligases Lnx1 and Lnx2 Localize at Neuronal Gap Junctions Formed by Connexin36 in Rodent Brain and Molecularly Interact with Connexin36. Eur. J. Neurosci. 2018, 48, 3062–3081. [Google Scholar] [CrossRef] [Green Version]
  19. Molchanova, S.M.; Huupponen, J.; Lauri, S.E.; Taira, T. Gap Junctions Between Ca3 Pyramidal Cells Contribute to Network Synchronization in Neonatal Hippocampus. Neuropharmacology 2016, 107, 9–17. [Google Scholar] [CrossRef]
  20. Del, C.C.; Iglesias, R.; Zoidl, G.; Dermietzel, R.; Spray, D.C. Calmodulin Dependent Protein Kinase Increases Conductance at Gap Junctions Formed by the Neuronal Gap Junction Protein Connexin36. Brain Res. 2012, 1487, 69–77. [Google Scholar] [CrossRef] [Green Version]
  21. Lanciotti, A.; Brignone, M.S.; Belfiore, M.; Columba-Cabezas, S.; Mallozzi, C.; Vincentini, O.; Molinari, P.; Petrucci, T.C.; Visentin, S.; Ambrosini, E. Megalencephalic Leukoencephalopathy with Subcortical Cysts Disease-Linked Mlc1 Protein Favors Gap-Junction Intercellular Communication by Regulating Connexin 43 Trafficking in Astrocytes. Cells 2020, 9, 1425. [Google Scholar] [CrossRef] [PubMed]
  22. Giaume, C.; Naus, C.C.; Saez, J.C.; Leybaert, L. Glial Connexins and Pannexins in the Healthy and Diseased Brain. Physiol. Rev. 2021, 101, 93–145. [Google Scholar] [CrossRef] [PubMed]
  23. Krutovskikh, V.A.; Piccoli, C.; Yamasaki, H. Gap Junction Intercellular Communication Propagates Cell Death in Cancerous Cells. Oncogene 2002, 21, 1989–1999. [Google Scholar] [CrossRef] [Green Version]
  24. Lin, J.H.; Weigel, H.; Cotrina, M.L.; Liu, S.; Bueno, E.; Hansen, A.J.; Hansen, T.W.; Goldman, S.; Nedergaard, M. Gap-Junction-Mediated Propagation and Amplification of Cell Injury. Nat. Neurosci. 1998, 1, 494–500. [Google Scholar] [CrossRef] [PubMed]
  25. Fontes, J.D.; Ramsey, J.; Polk, J.M.; Koop, A.; Denisova, J.V.; Belousov, A.B. Death of Neurons Following Injury Requires Conductive Neuronal Gap Junction Channels but Not a Specific Connexin. PLoS ONE 2015, 10, e125395. [Google Scholar] [CrossRef]
  26. Ma, D.; Feng, L.; Cheng, Y.; Xin, M.; You, J.; Yin, X.; Hao, Y.; Cui, L.; Feng, J. Astrocytic Gap Junction Inhibition by Carbenoxolone Enhances the Protective Effects of Ischemic Preconditioning Following Cerebral Ischemia. J. Neuroinflammation 2018, 15, 198. [Google Scholar] [CrossRef]
  27. Manjarrez-Marmolejo, J.; Franco-Perez, J. Gap Junction Blockers: An Overview of their Effects On Induced Seizures in Animal Models. Curr. Neuropharmacol. 2016, 14, 759–771. [Google Scholar] [CrossRef]
  28. Zhou, S.; Fang, Z.; Wang, G.; Wu, S. Gap Junctional Intercellular Communication Dysfunction Mediates the Cognitive Impairment Induced by Cerebral Ischemia-Reperfusion Injury: Pi3K/Akt Pathway Involved. Am. J. Transl. Res. 2017, 9, 5442–5451. [Google Scholar]
  29. Bruzzone, R.; Dermietzel, R. Structure and Function of Gap Junctions in the Developing Brain. Cell Tissue Res. 2006, 326, 239–248. [Google Scholar] [CrossRef]
  30. Orellana, J.A.; Saez, P.J.; Shoji, K.F.; Schalper, K.A.; Palacios-Prado, N.; Velarde, V.; Giaume, C.; Bennett, M.V.; Saez, J.C. Modulation of Brain Hemichannels and Gap Junction Channels by Pro-Inflammatory Agents and their Possible Role in Neurodegeneration. Antioxid. Redox Signal 2009, 11, 369–399. [Google Scholar] [CrossRef]
  31. Bennett, M.V.; Garre, J.M.; Orellana, J.A.; Bukauskas, F.F.; Nedergaard, M.; Saez, J.C. Connexin and Pannexin Hemichannels in Inflammatory Responses of Glia and Neurons. Brain Res. 2012, 1487, 3–15. [Google Scholar] [CrossRef] [Green Version]
  32. Acosta, M.L.; Mat, N.M.; Guo, C.X.; Mugisho, O.O.; Coutinho, F.P.; Rupenthal, I.D.; Green, C.R. Connexin Therapeutics: Blocking Connexin Hemichannel Pores Is Distinct from Blocking Pannexin Channels or Gap Junctions. Neural Regen. Res. 2021, 16, 482–488. [Google Scholar] [CrossRef] [PubMed]
  33. Orellana, J.A.; von Bernhardi, R.; Giaume, C.; Saez, J.C. Glial Hemichannels and their Involvement in Aging and Neurodegenerative Diseases. Rev. Neurosci. 2012, 23, 163–177. [Google Scholar] [CrossRef]
  34. Gangoso, E.; Talaveron, R.; Jaraiz-Rodriguez, M.; Dominguez-Prieto, M.; Ezan, P.; Koulakoff, A.; Medina, J.M.; Giaume, C.; Tabernero, A. A C-Src Inhibitor Peptide Based On Connexin43 Exerts Neuroprotective Effects through the Inhibition of Glial Hemichannel Activity. Front. Mol. Neurosci. 2017, 10, 418. [Google Scholar] [CrossRef] [Green Version]
  35. Orellana, J.A.; Froger, N.; Ezan, P.; Jiang, J.X.; Bennett, M.V.; Naus, C.C.; Giaume, C.; Saez, J.C. Atp and Glutamate Released Via Astroglial Connexin 43 Hemichannels Mediate Neuronal Death through Activation of Pannexin 1 Hemichannels. J. Neurochem. 2011, 118, 826–840. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Ge, Z.; Diao, H.; Yu, M.; Ji, X.; Liu, Q.; Chang, X.; Wu, Q. Connexin 43 Mediates Changes in Protein Phosphorylation in Hk-2 Cells During Chronic Cadmium Exposure. Environ. Toxicol. Pharmacol. 2017, 53, 184–190. [Google Scholar] [CrossRef] [PubMed]
  37. Shen, J.; Shen, J.; Zhu, K.; Zhou, H.; Tian, H.; Yu, G. Efficacy of Statins in Cerebral Vasospasm, Mortality, and Delayed Cerebral Ischemia in Patients with Aneurysmal Subarachnoid Hemorrhage: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. World Neurosurg. 2019, 131, e65–e73. [Google Scholar] [CrossRef]
  38. Bai, Q.; Xue, M.; Yong, V.W. Microglia and Macrophage Phenotypes in Intracerebral Haemorrhage Injury: Therapeutic Opportunities. Brain 2020, 143, 1297–1314. [Google Scholar] [CrossRef]
  39. Yong, H.; Rawji, K.S.; Ghorbani, S.; Xue, M.; Yong, V.W. The Benefits of Neuroinflammation for the Repair of the Injured Central Nervous System. Cell Mol. Immunol. 2019, 16, 540–546. [Google Scholar] [CrossRef]
  40. Yang, Y.; Xi, Z.; Xue, Y.; Ren, J.; Sun, Y.; Wang, B.; Zhong, Z.; Yang, G.Y.; Sun, Q.; Bian, L. Hemoglobin Pretreatment Endows Rat Cortical Astrocytes Resistance to Hemin-Induced Toxicity Via Nrf2/Ho-1 Pathway. Exp. Cell Res. 2017, 361, 217–224. [Google Scholar] [CrossRef]
  41. Munakata, M.; Shirakawa, H.; Nagayasu, K.; Miyanohara, J.; Miyake, T.; Nakagawa, T.; Katsuki, H.; Kaneko, S. Transient Receptor Potential Canonical 3 Inhibitor Pyr3 Improves Outcomes and Attenuates Astrogliosis After Intracerebral Hemorrhage in Mice. Stroke 2013, 44, 1981–1987. [Google Scholar] [CrossRef] [Green Version]
  42. Kim, Y.; Davidson, J.O.; Gunn, K.C.; Phillips, A.R.; Green, C.R.; Gunn, A.J. Role of Hemichannels in Cns Inflammation and the Inflammasome Pathway. Adv. Protein Chem. Struct. Biol. 2016, 104, 1–37. [Google Scholar] [CrossRef] [PubMed]
  43. Yu, H.; Cao, X.; Li, W.; Liu, P.; Zhao, Y.; Song, L.; Chen, J.; Chen, B.; Yu, W.; Xu, Y. Targeting Connexin 43 Provides Anti-Inflammatory Effects after Intracerebral Hemorrhage Injury by Regulating Yap Signaling. J. Neuroinflammation 2020, 17, 322. [Google Scholar] [CrossRef]
  44. Yang, Y.; Ren, J.; Sun, Y.; Xue, Y.; Zhang, Z.; Gong, A.; Wang, B.; Zhong, Z.; Cui, Z.; Xi, Z.; et al. A Connexin43/Yap Axis Regulates Astroglial-Mesenchymal Transition in Hemoglobin Induced Astrocyte Activation. Cell Death Differ. 2018, 25, 1870–1884. [Google Scholar] [CrossRef] [Green Version]
  45. Li, Z.; Liu, Y.; Wei, R.; Khan, S.; Xue, M.; Yong, V.W. The Combination of Deferoxamine and Minocycline Strengthens Neuroprotective Effect On Acute Intracerebral Hemorrhage in Rats. Neurol. Res. 2021, 43, 854–864. [Google Scholar] [CrossRef]
  46. Davalos, D.; Ryu, J.K.; Merlini, M.; Baeten, K.M.; Le Moan, N.; Petersen, M.A.; Deerinck, T.J.; Smirnoff, D.S.; Be-dard, C.; Hakozaki, H.; et al. Fibrinogen-Induced Perivascular Microglial Clustering is Required for the Development of Axonal Damage in Neuroinflammation. Nat. Commun. 2012, 3, 1227. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Retamal, M.A.; Froger, N.; Palacios-Prado, N.; Ezan, P.; Saez, P.J.; Saez, J.C.; Giaume, C. Cx43 Hemichannels and Gap Junction Channels in Astrocytes are Regulated Oppositely by Proinflammatory Cytokines Released From Activated Microglia. J. Neurosci. 2007, 27, 13781–13792. [Google Scholar] [CrossRef]
  48. Makarenkova, H.P.; Shestopalov, V.I. The Role of Pannexin Hemichannels in Inflammation and Regeneration. Front Physiol. 2014, 5, 63. [Google Scholar] [CrossRef] [Green Version]
  49. Meme, W.; Ezan, P.; Venance, L.; Glowinski, J.; Giaume, C. Atp-Induced Inhibition of Gap Junctional Communication is Enhanced by Interleukin-1 Beta Treatment in Cultured Astrocytes. Neuroscience 2004, 126, 95–104. [Google Scholar] [CrossRef] [PubMed]
  50. Abudara, V.; Roux, L.; Dallerac, G.; Matias, I.; Dulong, J.; Mothet, J.P.; Rouach, N.; Giaume, C. Activated Microglia Impairs Neuroglial Interaction by Opening Cx43 Hemichannels in Hippocampal Astrocytes. Glia 2015, 63, 795–811. [Google Scholar] [CrossRef]
  51. John, G.R.; Scemes, E.; Suadicani, S.O.; Liu, J.S.; Charles, P.C.; Lee, S.C.; Spray, D.C.; Brosnan, C.F. Il-1Beta Differentially Regulates Calcium Wave Propagation Between Primary Human Fetal Astrocytes Via Pathways Involving P2 Receptors and Gap Junction Channels. Proc. Natl. Acad. Sci. USA 1999, 96, 11613–11618. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Zvalova, D.; Cordier, J.; Mesnil, M.; Junier, M.P.; Chneiweiss, H. P38/Sapk2 Controls Gap Junction Closure in Astrocytes. Glia 2004, 46, 323–333. [Google Scholar] [CrossRef]
  53. Johnson, A.M.; Roach, J.P.; Hu, A.; Stamatovic, S.M.; Zochowski, M.R.; Keep, R.F.; Andjelkovic, A.V. Connexin 43 Gap Junctions Contribute to Brain Endothelial Barrier Hyperpermeability in Familial Cerebral Cavernous Malformations Type Iii by Modulating Tight Junction Structure. FASEB J. 2018, 32, 2615–2629. [Google Scholar] [CrossRef] [Green Version]
  54. Li, X.; Lu, S.; Nagy, J.I. Direct Association of Connexin36 with Zonula Occludens-2 and Zonula Occludens-3. Neurochem. Int. 2009, 54, 393–402. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Yang, X.; Chu, H.; Tang, Y.; Dong, Q. The Role of Connexin43 in Hemorrhagic Transformation After Thrombolysis in Vivo and in Vitro. Neuroscience 2016, 329, 54–65. [Google Scholar] [CrossRef]
  56. De Bock, M.; Culot, M.; Wang, N.; Bol, M.; Decrock, E.; De Vuyst, E.; Da, C.A.; Dauwe, I.; Vinken, M.; Simon, A.M.; et al. Connexin Channels Provide a Target to Manipulate Brain Endothelial Calcium Dynamics and Blood-Brain Barrier Permeability. J. Cereb. Blood Flow Metab. 2011, 31, 1942–1957. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Danesh-Meyer, H.V.; Kerr, N.M.; Zhang, J.; Eady, E.K.; O’Carroll, S.J.; Nicholson, L.F.; Johnson, C.S.; Green, C.R. Connexin43 Mimetic Peptide Reduces Vascular Leak and Retinal Ganglion Cell Death Following Retinal Ischaemia. Brain 2012, 135, 506–520. [Google Scholar] [CrossRef] [Green Version]
  58. Rozas-Villanueva, M.F.; Casanello, P.; Retamal, M.A. Role of Ros/Rns in Preeclampsia: Are Connexins the Missing Piece? Int. J. Mol. Sci. 2020, 21, 4698. [Google Scholar] [CrossRef]
  59. Quintanilla, R.A.; Orellana, J.A.; von Bernhardi, R. Understanding Risk Factors for Alzheimer’s Disease: Interplay of Neuroinflammation, Connexin-Based Communication and Oxidative Stress. Arch. Med. Res. 2012, 43, 632–644. [Google Scholar] [CrossRef]
  60. Le, H.T.; Sin, W.C.; Lozinsky, S.; Bechberger, J.; Vega, J.L.; Guo, X.Q.; Saez, J.C.; Naus, C.C. Gap Junction Intercellular Communication Mediated by Connexin43 in Astrocytes is Essential for their Resistance to Oxidative Stress. J. Biol. Chem. 2014, 289, 1345–1354. [Google Scholar] [CrossRef] [Green Version]
  61. Balboa, E.; Saavedra, F.; Cea, L.A.; Ramirez, V.; Escamilla, R.; Vargas, A.A.; Regueira, T.; Saez, J.C. Vitamin E Blocks Connexin Hemichannels and Prevents Deleterious Effects of Glucocorticoid Treatment On Skeletal Muscles. Int. J. Mol. Sci. 2020, 21, 4094. [Google Scholar] [CrossRef] [PubMed]
  62. Ma, J.W.; Ji, D.D.; Li, Q.Q.; Zhang, T.; Luo, L. Inhibition of Connexin 43 Attenuates Oxidative Stress and Apoptosis in Human Umbilical Vein Endothelial Cells. BMC Pulm. Med. 2020, 20, 19. [Google Scholar] [CrossRef]
  63. Chen, X.; Liang, H.; Xi, Z.; Yang, Y.; Shan, H.; Wang, B.; Zhong, Z.; Xu, C.; Yang, G.Y.; Sun, Q.; et al. Bm-Msc Transplantation Alleviates Intracerebral Hemorrhage-Induced Brain Injury, Promotes Astrocytes Vimentin Expression, and Enhances Astrocytes Antioxidation Via the Cx43/Nrf2/Ho-1 Axis. Front Cell Dev. Biol. 2020, 8, 302. [Google Scholar] [CrossRef]
  64. Smetana, K.S.; Buschur, P.L.; Owusu-Guha, J.; May, C.C. Pharmacologic Management of Cerebral Vasospasm in Aneurysmal Subarachnoid Hemorrhage. Crit. Care Nurs. Q. 2020, 43, 138–156. [Google Scholar] [CrossRef] [PubMed]
  65. Yang, L.; Yan, J.; Zhang, J.A.; Zhou, X.H.; Fang, C.; Zeng, E.M.; Tang, B.; Duan, J.; Lu, G.H.; Hong, T. The Important Role of Connexin 43 in Subarachnoid Hemorrhage-Induced Cerebral Vasospasm. J. Transl. Med. 2019, 17, 433. [Google Scholar] [CrossRef] [Green Version]
  66. Lei, C.; Ruan, Y.; Cai, C.; He, B.; Zhao, D. Role of P38 Mitogen-Activated Protein Kinase On Cx43 Phosphorylation in Cerebral Vasospasm After Subarachnoid Hemorrhage. Int. J. Neurosci. 2019, 129, 461–469. [Google Scholar] [CrossRef]
  67. Hong, T.; Wang, H.; Wang, Y.; Wang, H. Effects of Gap Junctional Blockers On Cerebral Vasospasm After Subarachnoid Hemorrhage in Rabbits. Neurol. Res. 2009, 31, 238–244. [Google Scholar] [CrossRef]
  68. Li, H.; Liu, S.; Sun, X.; Yang, J.; Yang, Z.; Shen, H.; Li, X.; Liu, Y.; Chen, G. Critical Role for Annexin a7 in Secondary Brain Injury Mediated by its Phosphorylation After Experimental Intracerebral Hemorrhage in Rats. Neurobiol. Dis. 2018, 110, 82–92. [Google Scholar] [CrossRef] [PubMed]
  69. Weilinger, N.L.; Lohman, A.W.; Rakai, B.D.; Ma, E.M.; Bialecki, J.; Maslieieva, V.; Rilea, T.; Bandet, M.V.; Ikuta, N.T.; Scott, L.; et al. Metabotropic Nmda Receptor Signaling Couples Src Family Kinases to Pannexin-1 During Excitotoxicity. Nat. Neurosci. 2016, 19, 432–442. [Google Scholar] [CrossRef]
  70. Huang, Y.; Lin, J.; Chen, X.; Lin, J. Pannexin-1 Contributes to the Apoptosis of Spinal Neurocytes in Spinal Cord Injury. Front. Physiol. 2021, 12, 656647. [Google Scholar] [CrossRef]
  71. Imamura, H.; Sakamoto, S.; Yoshida, T.; Matsui, Y.; Penuela, S.; Laird, D.W.; Mizukami, S.; Kikuchi, K.; Kakizuka, A. Single-Cell Dynamics of Pannexin-1-Facilitated Programmed Atp Loss During Apoptosis. Elife 2020, 9, e61960. [Google Scholar] [CrossRef]
  72. Zhou, L.; Liu, C.; Wang, Z.; Shen, H.; Wen, Z.; Chen, D.; Sun, Q.; Chen, G. Pannexin-1 is Involved in Neuronal Apoptosis and Degeneration in Experimental Intracerebral Hemorrhage in Rats. Mol. Med. Rep. 2018, 17, 5684–5691. [Google Scholar] [CrossRef]
  73. Chu, H.; Gao, Z.; Huang, C.; Dong, J.; Tang, Y.; Dong, Q. Relationship Between Hematoma Expansion Induced by Hypertension and Hyperglycemia and Blood-Brain Barrier Disruption in Mice and its Possible Mechanism: Role of Aquaporin-4 and Connexin43. Neurosci. Bull. 2020, 36, 1369–1380. [Google Scholar] [CrossRef] [PubMed]
  74. de Pina-Benabou, M.H.; Szostak, V.; Kyrozis, A.; Rempe, D.; Uziel, D.; Urban-Maldonado, M.; Benabou, S.; Spray, D.C.; Federoff, H.J.; Stanton, P.K.; et al. Blockade of Gap Junctions in Vivo Provides Neuroprotection After Perinatal Global Ischemia. Stroke 2005, 36, 2232–2237. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Ayer, R.; Chen, W.; Sugawara, T.; Suzuki, H.; Zhang, J.H. Role of Gap Junctions in Early Brain Injury Following Subarachnoid Hemorrhage. Brain Res. 2010, 1315, 150–158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Manaenko, A.; Lekic, T.; Sozen, T.; Tsuchiyama, R.; Zhang, J.H.; Tang, J. Effect of Gap Junction Inhibition On Intracerebral Hemorrhage-Induced Brain Injury in Mice. Neurol. Res. 2009, 31, 173–178. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Possible mechanisms by which connexins and pannexins mediates inflammatory responses following ICH. Microglia activated by hematoma degradation products release pro-inflammatory molecules, including IL−1β, IL−6, TNF-α and ATP. These molecules may auto-activate P2X receptors on the membrane of microglia, leading to the activation of NRLP3 pro-inflammatory inflammasome to exacerbate the release of pro-inflammatory molecules. Conversely, they also activate Cx43 hemichannels in astrocytes to aggravate the inflammatory responses by inactivating pro-inflammatory YAP-SOCS1-SOCS3-TLR4-NFκB and JAK2-STAT3 axis pathways. Furthermore, ATP released from Cx43 hemichannels results in the opening of Cx36 hemichannel and Panx1 channels in neurons, eventually causing neuronal damage. ICH, intracerebral hemorrhage; Cx, connexins; Panx, pannexins; P2X, purinergic P2X receptors; NRLP3, nucleotide-binding oligomerization domain-like receptor family, pyrin domain-containing−3; HCs, hemichannels; GJIC, gap junctional intercellular communication.
Figure 1. Possible mechanisms by which connexins and pannexins mediates inflammatory responses following ICH. Microglia activated by hematoma degradation products release pro-inflammatory molecules, including IL−1β, IL−6, TNF-α and ATP. These molecules may auto-activate P2X receptors on the membrane of microglia, leading to the activation of NRLP3 pro-inflammatory inflammasome to exacerbate the release of pro-inflammatory molecules. Conversely, they also activate Cx43 hemichannels in astrocytes to aggravate the inflammatory responses by inactivating pro-inflammatory YAP-SOCS1-SOCS3-TLR4-NFκB and JAK2-STAT3 axis pathways. Furthermore, ATP released from Cx43 hemichannels results in the opening of Cx36 hemichannel and Panx1 channels in neurons, eventually causing neuronal damage. ICH, intracerebral hemorrhage; Cx, connexins; Panx, pannexins; P2X, purinergic P2X receptors; NRLP3, nucleotide-binding oligomerization domain-like receptor family, pyrin domain-containing−3; HCs, hemichannels; GJIC, gap junctional intercellular communication.
Biology 11 00027 g001
Figure 2. Possible mechanism by which astrocyte Cx43 resists oxidative stress following ICH. Cx43 from endoplasmic reticulum helps liberate Nrf2 from Keap1 and allows its nuclear translocation, promoting phase II detoxification enzyme genes involved in antioxidant stress and anti-apoptosis. ICH, intracerebral hemorrhage; Cx43, connexin 43; OS, oxidative stress; Nrf2, nuclear factor erythroid 2-related factor 2; CREB, cAMP-response element-binding protein.
Figure 2. Possible mechanism by which astrocyte Cx43 resists oxidative stress following ICH. Cx43 from endoplasmic reticulum helps liberate Nrf2 from Keap1 and allows its nuclear translocation, promoting phase II detoxification enzyme genes involved in antioxidant stress and anti-apoptosis. ICH, intracerebral hemorrhage; Cx43, connexin 43; OS, oxidative stress; Nrf2, nuclear factor erythroid 2-related factor 2; CREB, cAMP-response element-binding protein.
Biology 11 00027 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Zhang, Y.; Khan, S.; Liu, Y.; Siddique, R.; Zhang, R.; Yong, V.W.; Xue, M. Gap Junctions and Hemichannels Composed of Connexins and Pannexins Mediate the Secondary Brain Injury Following Intracerebral Hemorrhage. Biology 2022, 11, 27. https://doi.org/10.3390/biology11010027

AMA Style

Zhang Y, Khan S, Liu Y, Siddique R, Zhang R, Yong VW, Xue M. Gap Junctions and Hemichannels Composed of Connexins and Pannexins Mediate the Secondary Brain Injury Following Intracerebral Hemorrhage. Biology. 2022; 11(1):27. https://doi.org/10.3390/biology11010027

Chicago/Turabian Style

Zhang, Yan, Suliman Khan, Yang Liu, Rabeea Siddique, Ruiyi Zhang, Voon Wee Yong, and Mengzhou Xue. 2022. "Gap Junctions and Hemichannels Composed of Connexins and Pannexins Mediate the Secondary Brain Injury Following Intracerebral Hemorrhage" Biology 11, no. 1: 27. https://doi.org/10.3390/biology11010027

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop