Open access

Plasma Membrane Channels Formed by Connexins or Pannexins in Microglia: Possible Role in the Inflamed Brain

Written By

Juan A. Orellana

Submitted: 20 April 2012 Published: 15 May 2013

DOI: 10.5772/54306

From the Edited Volume

Neurodegenerative Diseases

Edited by Uday Kishore

Chapter metrics overview

2,361 Chapter Downloads

View Full Metrics

1. Introduction

In a healthy brain, microglia exhibit a resting surveillance state associated with active exploration of their environment for exogenous or endogenous signals representing a threat to the homeostasis [1-5]. When physiological balance is impaired in the central nervous system (CNS), resting phenotype of microglia shift to a reactive phenotype with different degrees of activation according to the nature of the stimuli and the context. During intense CNS inflammation, rather than show a repair-orientated activity profile, reactive microglia constitute a source of toxic factors and participate in the recruitment of non-resident brain cells involved in the innate immune response, which worsen brain damage. The brain performs exceptionally complex and dynamic tasks that depend on the coordinated interaction of glial cells, therefore it is conceivable that impairment of intercellular signaling and coordination among microglia could play an important role on several CNS disorders. In vertebrate cells, this synchronization is in part mediated by gap junctions [6-10]. They are aggregates of intercellular channels termed gap junction channels that allow direct, but selective, cytoplasmic continuity between contacting cells, promoting the exchange of ions (allowing eletrical coupling), metabolites (e.g., ADP, glucose, glutamate and glutathione) and second messengers (e.g., cAMP and IP3)[11-16]. Whereas a gap junction channel is formed by the serial docking of two hemichannels each one contributed by one of two adjacent cells, each hemichannel is composed by six protein subunits termed connexins (Fig. 1). The latter belong to a highly conserved protein family encoded by 21 genes in human and 20 in mouse with orthologs in other vertebrate species [17-19]. Connexins are abundantly expressed in cells of the CNS, and they are named after their predicted molecular mass expressed in kDa, so that connexin43 (Cx43) has a molecular mass of ~43 kDa.

Figure 1.

Diagram illustrating basic structures of gap junction channels and hemichannels formed by connexins or pannexins in microglia. Connexins and pannexins share similar membrane topology, with four α-helical transmembrane domains (M1-M4) connected by two extracellular loops (E1 and E2), one cytoplasmic loop (CL) where both amino (NH2)- and caboxy (COOH)-termini are intracellular. Top and bottom center show hemichannels formed by six connexin or pannexin subunits each, respectively. The middle center shows a connexin gap junction channel, at a close contact between two microglia. A hemichannel is formed by six connexins or pannexins that oligomerize laterally leaving a central pore in the activated state (open). Under resting conditions hemichannels remain preferentially closed, but they can be activated by diverse physiological and pathological conditions, offering a diffusional transmembrane route between the intra and extracellular milieu. In addition, it is depicted the types of hemichannels and gap junction channels expressed by microglia. This figure includes only the available information obtained under in vivo and/or in vitro studies using more than one experimental approach.

For a long time the main function attributed to connexin hemichannels was the formation of gap junction channels. Nevertheless, in the last decade, the presence of functional connexin hemichannels in nonjunctional membranes has been demonstrated by several experimental approaches [20-24]. These channels serve like aqueous pores permeable to ions and small molecules that permit diffusional exchange between the intra and extracellular compartments, allowing cellular release of relevant quantities of autocrine/paracrine signaling molecules (e.g., ATP, glutamate, NAD+ and PGE2) to the extracellular milieu [25-30], as well as uptake of small molecules (e.g., glucose) [31]. One decade ago, a new gene family of gap junction proteins composed by three members was discovered in chordates [32, 33]. These proteins are the chordate homologs of innexins (the gap junction proteins of non chordates), and were denominated pannexins (panx1, 2 and 3) because apparently they are present in all eumetazoans except echinoderms [34] (Fig. 1). It has been suggested that gap junctional intercellular communication occur via Panx3 in osteoblasts [35], whereas other studies have shown that overexpression of exogenous Panx1 could form gap junctions in vitro [33, 36, 37]. Nevertheless, the absence of ultrastructural evidences for gap junction formation and demonstration of functional communication mediated by other endogenously expressed pannexins indicate that they apparently act mainly as hemichannels [38].

Current knowledge regarding brain hemichannels state that, under physiological conditions, they have a low activity, but enough to ensure the release of paracrine substances necessary for diverse functions of the CNS, including ischemic tolerance [39, 40], establishment of adhesive interactions [41]; fear memory consolidation [42], glucosensing [30], chemoreception [43], blood-brain barrier permeability [44], neuronal migration [45, 46] and metabolic autocrine regulation [47]. Nevertheless, under acute or chronic neurodegeneration dysregulation of hemichannel properties could be critical on the beginning and maintenance of homeostatic imbalances observed in diverse brain diseases [48-50]. Pioneering findings from Paul and colleagues showed that Xenopus oocytes transfected with Cx46 mRNA exhibited non-selective cation currents associated to depolarization and cell lysis within 24 h [51]. From then on, several studies supported the idea that dysregulated opening of hemichannels is incompatible with normal cell life. In the CNS, the first convincing evidence of hemichannel opening was provided by Contreras and colleagues, whose work showed that opening of Cx43 hemichannels accelerate astroglial cell death induced by ischemia-like conditions [52]. Such increased hemichannel activity induced by ischemia-like conditions has been observed in neurons [40, 53-55], oligodendrocytes [56], and also in brain cells subjected to other pro-inflammatory conditions [48]. Up to now, it is believed that sustained hemichannel opening contributes to increased intracellular Ca2+ concentration ([Ca2+]i), which in turn may favor even more the hemichannel activity (De Vuyst et al., 2007, Schalper et al., 2008), inducing Ca2+ and Na+ intracellular overload (Fig. 2).

Under these conditions, ionic (or electrolyte) imbalance leads to an osmotic imbalance that results in cell swelling and plasma membrane breakdown. Calcium overload induced in part by hemichannel opening may also activate phospholipase A2, with the subsequent generation of arachidonic acid and activation of cyclooxygenase/lipoxygenase pathways leading to increased free radicals, lipid peroxidation and further plasma membrane damage. Possibly, exacerbated or uncontrolled hemichannel opening could lead to cellular damage by several ways: 1) High increase of [Ca2+]i by Ca2+ entry through hemichannels, 2) cellular swelling by increased entry of Na2+ and Cl- through hemichannels, 3) release of metabolic products essential to cell viability as glucose, NAD+ or glutathione via hemichannels and 4) alternatively, spread of toxic molecules released by hemichannels (e.g., glutamate) could affect the viability of healthy neighboring cells.

Figure 2.

Dysregulated opening of hemichannels induces cell damage by different mechanisms. Under normal conditions, hemichannels (yellow channels) exhibit a low activity. However, upon exposure to inflammatory conditions, hemichannels undergo a dysregulation proccess leading to an uncontrolled opening which further results in cellular damage by various mechanisms. (A) Ca2+ entry through hemichannels activate phospholipase A2, with the subsequent generation of arachidonic acid and activation of cyclooxygenase/lipoxygenase pathways leading to increased free radicals, lipid peroxidation and further plasma membrane damage. Note that increased levels of [Ca2+]i may activate even more hemichannel opening as demonstrated previously [57, 58]. (B) Na2+ and Cl- entry through hemichannels could produce cellular swelling by increased influx of H2O via aquoporins (green channels). (C) Release of essencial metabolic products via hemichannels (eg., glucose, NAD+ or glutathione) could increase cell vulnerability. (D) Release via hemichannels of molecules that in high amounts are toxic (e.g., ATP and glutamate) could affect the viability of healthy neighboring cells and spread damage.

Taking into account that hemichannels participate in the paracrine signaling among brain cells, the current chapter attempts to review and discuss the role of gap junction channels and hemichannels in microglia on normal and inflamed brain.

Advertisement

2. Gap junction channels in microglia

In a resting surveillance state, microglia express almost undetectable levels of Cx43 and Cx36 [59-65]. Nevertheless, when microglia are subjected to pro-inflammatory conditions, they exhibit expression of Cx43 and are able to form gap junction channels among them, as evaluated by dye-coupling experiments. In fact, Cx43 expression and gap junctional communication is induced in microglia by LPS, TNF-α plus IFN-γ [61], calcium ionophore plus PMA [66], or Staphylococcus aureus-derived peptidoglycan [64]. Despite the above, cultured human or mouse microglia treated with LPS, granulocyte-macrophage colony-stimulating factor, INF-γ or TNF-α do not exhibit modifications in connexin expression [60, 63]. Recently has been showed that resting microglia exhibit detectable levels of surface and total Cx43, whereas upon treatment with amyloid-β peptide (Aβ) a high increase in Cx43 expression is observed (Orellana 2011a). The discrepancy in the above mentioned studies may be related to different types of animal used to obtain brain tissue, dissimilar methods to take out cells and different culture conditions.

The ability to establish gap junctional communication among microglia, requires a rise in [Ca2+]i [66], while cAMP, cGMP or activation of PKC have been ruled out as possible inductors gap junction-mediated coupling [66]. In this regard, different degrees of microglial activation may trigger intracellular pathways that further result in a specific pattern of expression of gap juntion proteins. Communication via gap junctions may allow to activated microglia to recruit resting microglia at the site of injury, resulting in more damage or repair depending on the circumstances. Interestingly, microglia stimulated with cytokines or LPS exhibit reduced levels of Cx43 expression and gap junctional communication in astrocytes when both cell types are in co-culture or when conditioned media from activated microglia is used [31, 55, 59, 60, 67, 68]. Interestingly, gap junctions among dendritic cells ensure sharing of antigenic peptides [69-74], suggesting the possibility that these channels in microglia also could coordinate the CNS immune response. Importantly, recently it has been shown that the release of TNF-α and IL-1β by microglia depend on the activity of gap junction channels, because secretion of those cytokines was partially blocked by a gap junction blocker, α-glicirretinic acid [75]. Thus, it was proposed that gap junction channels play a key role into coordinate the microglial mediated inflammation.

Advertisement

3. Hemichannels in microglia

Up to now only few studies have documented the expression of functional hemichannels in microglia. Contrary to the expectations regarding as Cx43 the most possible protein to form hemichannels in microglia, TNF-α treatment was shown to induce release of glutamate through a pathway inhibited by a Cx32 (32Gap27), but not Cx43 (43Gap27) mimetic peptide [76]. Moreover, surface levels of Cx32 were increased in microglia treated with TNF-α. Noteworthy, the increased neuronal death associated with the release of glutamate was inhibited completely with the 32Gap27 mimetic peptide [76]. Later, the same group of authors proposed that glutamate released via Cx32 hemichannels play a key role in neuronal damage originated by brain ischemia [77] and experimental autoimmune encephalomyelitis [78]. Accordingly, microglial cells from Mecp2 null mice, a model of a neurodevelopmental disorder known as Rett syndrome, promote neuronal death through glutamate release via a cell membrane pathway inhibited by 32Gap27 and 32Gap24, two Cx32 hemichannel mimetic peptides [79]. It is relevant to kept in mind that these and other mimetic peptides are homologous to extracellular domains of the respective connexin sequences, but their effects on hemichannel activity have not been documented, thereby some studies have questioned their specificity [80-82]. The use of cell cultures derived from connexin null mice and/or performing knockdown of the respective connexin, along the appropriate use of mimetic peptides could ensure the involvement of Cx32 hemichannels in these studies.

Almost two years ago, the opening of Cx43 and Panx1 hemichannels, evaluated by dye uptake and macroscopic cell membrane currents, were shown to be increased in microglia by Aβ25-35 exposure (Orellana et al. 2012a). These observations were confirmed by using microglial cultures from Cx43 KO mice and Panx1 mimetic peptides. These currents were recorded at negative holding potential (-60 mV) in the presence of external divalent cations, suggesting that opening of microglial hemichannels may occur in Alzheimer's disease (AD). Importantly, ATP and glutamate released from microglia treated with Aβ25-35 trigger hemichannel opening in neurons causing deleterious effects on them [83]. Supporting the idea of hemichannels as possible regulators in damage observed in AD, a novel putative hemichannel blocker (INI-0602) that crosses the blood brain barrier was recently shown to inhibit in vivo the LPS-induced glutamate release from microglia and to improve memory deficits in APP/PS1 mice [84]. Due the pharmacological pattern of this response," it was proposed the involvement of Cx32 hemichannels. However, the possible implication of other hemichannel forming proteins or even other channels was not ruled out and studies on the specificity of INI-0602 require further demonstration using, for example, in vivo experiments with Cx32-/- microglia or knockdown of Cx32. To demonstrate the participation of hemichannels in this disease it is necessary to analyze the functional state of microglial hemichannels in brain slices from AD model mice (APP/PS1) by using patch-clamp and membrane permeability assays.

Figure 3.

Role of microglial cell hemichannels and gap junction channels during neuroinflammation. Chronic or acute inflammation increases hemichannel (HC) activity in microglia allowing the influx of Ca2+ (1) and its spread to neighbor cells through gap junctions (GJCs) (2) raising the intracellular free Ca2+ concentration ([Ca2+]i). HC opening induced by inflammation in microglia leads to ATP release (3), which diffuses through the extracellular space and activates membrane purinergic (P2) receptors (4). High levels of [Ca2+]i (5) allow the release of glutamate through microglial cell HCs (6) and further activation of neuronal NMDA receptors (7). P2 and NMDA receptor activation in neurons increase the activity of neuronal Panx1 and Cx36 HCs, affecting electrochemical and Ca2+ imbalance in neurons, which leads to cell death (8).

Advertisement

4. Conclusions

Microglial cells are known to play a relevant role in neuronal survival [3]. In pathological situations, dysregulation of connexin- and pannexin-based channels expressed by microglia, contribute importantly to determine the neuronal fate [48, 50]. Microgliosis and brain inflammation are associated with most, if not all, brain injuries and pathologies. Hemichannel activation in microglia could play a crucial role in the reinforcement of the neuronal death, due to their capacity to release glutamate and ATP (Fig. 3) [55, 76, 83, 85]. Opening of Cx43, Cx32 and Panx1 hemichannels could increase [Ca2+]i in microglia, which further propagate Ca2+ waves via gap junction channels to neighbor cells (Fig. 3). Moreover, in distant microglia, Ca2+ waves can activate hemichannels, as demonstrated previously [57, 58, 86]. Then, opening of neuronal Panx1 hemichannels could be triggered by the rise in [Ca2+]i via activation of NMDA and P2X receptors by glutamate and ATP, respectively. Panx1 hemichannels are likely to contribute to the intracellular Ca2+ overload that activates neurotoxic intracellular cascades during excitotoxicity [87] (Fig. 3). Thus, the prevention of hemichannel activation under pro-inflammatory conditions may represent an unexplored strategy to prevent neuronal damage and death. Altogether these observations strengthen the emerging concept that unregulated membrane permeability through enhanced hemichannel permeability and dysfunctional gap junction channels may contribute to the development of CNS pathologies and connexins as well as pannexins might represent potential and alternative targets for therapeutic intervention in neuroinflammatory diseases.

Advertisement

Acknowledgments

This work was partially supported by CONICYT 79090028 and FONDECYT 11121133 (to JAO) grants.

References

  1. 1. KettenmannHet alPhysiology of microglia. Physiol Rev, 2011461553
  2. 2. BlockM. Land J. SHongMicroglia and inflammation-mediated neurodegeneration: multiple triggers with a common mechanism. Prog Neurobiol, 20057798
  3. 3. BlockM. LLZeccaand J. SHongMicroglia-mediated neurotoxicity: uncovering the molecular mechanisms. Nat Rev Neurosci, 20075769
  4. 4. HanischU. KMicroglia as a source and target of cytokines. Glia, 2002140155
  5. 5. StreitW. JMicroglia as neuroprotective, immunocompetent cells of the CNS. Glia, 2002133139
  6. 6. SaezJ. Cet alPlasma membrane channels formed by connexins: their regulation and functions. Physiol Rev, 200313591400
  7. 7. SohlGand KWilleckeGap junctions and the connexin protein family. Cardiovasc Res, 2004228232
  8. 8. LairdD. WThe gap junction proteome and its relationship to disease. Trends Cell Biol, 201092101
  9. 9. GoodenoughD. Aand D. LPaulGap junctions. Cold Spring Harb Perspect Biol, 2009a002576
  10. 10. EvansW. HEDe Vuystand LLeybaertThe gap junction cellular internet: connexin hemichannels enter the signalling limelight. Biochem J, 2006114
  11. 11. SaezJ. Cet alcAMP delays disappearance of gap junctions between pairs of rat hepatocytes in primary culture. Am J Physiol, 1989Pt 1): C111
  12. 12. ConnorsB. Wand M. ALongElectrical synapses in the mammalian brain. Annu Rev Neurosci, 2004393418
  13. 13. GoldbergG. SP. DLampeand B. JNicholsonSelective transfer of endogenous metabolites through gap junctions composed of different connexins. Nat Cell Biol, 1999457459
  14. 14. KamYet alTransfer of second messengers through gap junction connexin 43 channels reconstituted in liposomes. Biochim Biophys Acta, 1998384388
  15. 15. LawrenceT. SW. HBeersand N. BGilulaTransmission of hormonal stimulation by cell-to-cell communication. Nature, 1978501506
  16. 16. NiessenHet alSelective permeability of different connexin channels to the second messenger inositol 1,4,5-trisphosphate. J Cell Sci, 2000Pt 8): 13651372
  17. 17. CrucianiVand S. OMikalsenThe connexin gene family in mammals. Biol Chem, 2005325332
  18. 18. AbascalFand RZardoyaEvolutionary analyses of gap junction protein families. Biochim Biophys Acta, 2012
  19. 19. WilleckeKet alStructural and functional diversity of connexin genes in the mouse and human genome. Biol Chem, 2002725737
  20. 20. GoodenoughD. Aand D. LPaulBeyond the gap: functions of unpaired connexon channels. Nat Rev Mol Cell Biol, 2003285294
  21. 21. StoutCD. AGoodenoughand D. LPaulConnexins: functions without junctions. Curr Opin Cell Biol, 2004507512
  22. 22. SaezJ. Cet alConnexin-based gap junction hemichannels: gating mechanisms. Biochim Biophys Acta, 2005215224
  23. 23. SchalperK. Aet alCurrently used methods for identification and characterization of hemichannels. Cell Commun Adhes, 2008207218
  24. 24. SaezJ. Cet alCell membrane permeabilization via connexin hemichannels in living and dying cells. Exp Cell Res, 201023772389
  25. 25. BruzzoneSet alConnexin 43 hemi channels mediate Ca2+-regulated transmembrane NAD+ fluxes in intact cells. FASEB J, 20011012
  26. 26. YeZ. Cet alFunctional hemichannels in astrocytes: a novel mechanism of glutamate release. J Neurosci, 200335883596
  27. 27. CherianP. Pet alMechanical strain opens connexin 43 hemichannels in osteocytes: a novel mechanism for the release of prostaglandin. Mol Biol Cell, 200531003106
  28. 28. StoutC. Eet alIntercellular calcium signaling in astrocytes via ATP release through connexin hemichannels. J Biol Chem, 20021048210488
  29. 29. BraetKet alPharmacological sensitivity of ATP release triggered by photoliberation of inositol-1,4,5-trisphosphate and zero extracellular calcium in brain endothelial cells. J Cell Physiol, 2003205213
  30. 30. OrellanaJ. Aet alGlucose increases intracellular free Ca(2+) in tanycytes via ATP released through connexin 43 hemichannels. Glia, 20125368
  31. 31. RetamalM. Aet alCx43 hemichannels and gap junction channels in astrocytes are regulated oppositely by proinflammatory cytokines released from activated microglia. J Neurosci, 20071378113792
  32. 32. PanchinYet alA ubiquitous family of putative gap junction molecules. Curr Biol, 2000R473R474
  33. 33. BruzzoneRet alPannexins, a family of gap junction proteins expressed in brain. Proc Natl Acad Sci U S A, 20031364413649
  34. 34. ShestopalovV. Iand YPanchinPannexins and gap junction protein diversity. Cell Mol Life Sci, 2008376394
  35. 35. IshikawaMet alPannexin 3 functions as an ER Ca(2+) channel, hemichannel, and gap junction to promote osteoblast differentiation. J Cell Biol, 201112571274
  36. 36. LaiC. Pet alTumor-suppressive effects of pannexin 1 in C6 glioma cells. Cancer Res, 200715451554
  37. 37. Vanden AbeeleF., et al., Functional implications of calcium permeability of the channel formed by pannexin 1. J Cell Biol, 2006535546
  38. 38. MacVicarB.A. and R.J. Thompson, Non-junction functions of pannexin-1 channels. Trends Neurosci, 201093102
  39. 39. LinJ. Het alA central role of connexin 43 in hypoxic preconditioning. J Neurosci, 2008681695
  40. 40. SchockS. Cet alATP release by way of connexin 36 hemichannels mediates ischemic tolerance in vitro. Biochem Biophys Res Commun, 2008138144
  41. 41. CotrinaM. LJ. HLinand MNedergaardAdhesive properties of connexin hemichannels. Glia, 200817911798
  42. 42. StehbergJet alRelease of gliotransmitters through astroglial connexin 43 hemichannels is necessary for fear memory consolidation in the basolateral amygdala. FASEB J, 2012
  43. 43. WenkerI. Cet alRegulation of ventral surface CO2/H+-sensitive neurons by purinergic signalling. J Physiol, 2012Pt 9): 21372150
  44. 44. De BockMet alConnexin channels provide a target to manipulate brain endothelial calcium dynamics and blood-brain barrier permeability. J Cereb Blood Flow Metab, 201119421957
  45. 45. LiuXet alGap junctions/hemichannels modulate interkinetic nuclear migration in the forebrain precursors. J Neurosci, 201041974209
  46. 46. LiuXet alConnexin 43 controls the multipolar phase of neuronal migration to the cerebral cortex. Proc Natl Acad Sci U S A, 201282808285
  47. 47. KawamuraMJrD. NRuskinand S. AMasino, Metabolic autocrine regulation of neurons involves cooperation among pannexin hemichannels, adenosine receptors, and KATP channels. J Neurosci, 201038863895
  48. 48. OrellanaJ. Aet alModulation of brain hemichannels and gap junction channels by pro-inflammatory agents and their possible role in neurodegeneration. Antioxid Redox Signal, 2009369399
  49. 49. OrellanaJ. Aet alHemichannels in the neurovascular unit and white matter under normal and inflamed conditions. CNS Neurol Disord Drug Targets, 2011404414
  50. 50. OrellanaJ. Aet alGlial hemichannels and their involvement in aging and neurodegenerative diseases. Rev Neurosci, 2012163177
  51. 51. PaulD. Let alConnexin46, a novel lens gap junction protein, induces voltage-gated currents in nonjunctional plasma membrane of Xenopus oocytes. J Cell Biol, 199110771089
  52. 52. ContrerasJ. Eet alMetabolic inhibition induces opening of unapposed connexin 43 gap junction hemichannels and reduces gap junctional communication in cortical astrocytes in culture. Proc Natl Acad Sci U S A, 2002495500
  53. 53. ThompsonR. JNZhouand B.A. MacVicar, Ischemia opens neuronal gap junction hemichannels. Science, 2006924927
  54. 54. ThompsonR. Jet alActivation of pannexin-1 hemichannels augments aberrant bursting in the hippocampus. Science, 200815551559
  55. 55. OrellanaJ. Aet alATP and glutamate released via astroglial connexin 43 hemichannels mediate neuronal death through activation of pannexin 1 hemichannels. J Neurochem, 2011826840
  56. 56. DomercqMet al2X7receptors mediate ischemic damage to oligodendrocytes. Glia, 2010p. 730-40.
  57. 57. SchalperK. Aet alConnexin 43 hemichannels mediate the Ca2+ influx induced by extracellular alkalinization. Am J Physiol Cell Physiol, 2010C1504C1515
  58. 58. De VuystEet alConnexin hemichannels and gap junction channels are differentially influenced by lipopolysaccharide and basic fibroblast growth factor. Mol Biol Cell, 20073446
  59. 59. RouachNet alBrain macrophages inhibit gap junctional communication and downregulate connexin 43 expression in cultured astrocytes. Eur J Neurosci, 2002403407
  60. 60. MemeWet alProinflammatory cytokines released from microglia inhibit gap junctions in astrocytes: potentiation by beta-amyloid. FASEB J, 2006494496
  61. 61. EugeninE. Aet alMicroglia at brain stab wounds express connexin 43 and in vitro form functional gap junctions after treatment with interferon-gamma and tumor necrosis factor-alpha. Proc Natl Acad Sci U S A, 200141904195
  62. 62. ParentiRet alImmunocytochemical and RT-PCR analysis of connexin36 in cultures of mammalian glial cells. Arch Ital Biol, 2002101108
  63. 63. DobrenisKet alHuman and mouse microglia express connexin36, and functional gap junctions are formed between rodent microglia and neurons. J Neurosci Res, 2005306315
  64. 64. GargSMMdSyed, and T. Kielian, Staphylococcus aureus-derived peptidoglycan induces Cx43 expression and functional gap junction intercellular communication in microglia. J Neurochem, 2005475483
  65. 65. LeeI. Het alGlial and neuronal connexin expression patterns in the rat spinal cord during development and following injury. J Comp Neurol, 2005110
  66. 66. MartinezA. Det alIdentification of second messengers that induce expression of functional gap junctions in microglia cultured from newborn rats. Brain Res, 2002191201
  67. 67. FaustmannP. Met alMicroglia activation influences dye coupling and Cx43 expression of the astrocytic network. Glia, 2003101108
  68. 68. HinkeroheDet alEffects of cytokines on microglial phenotypes and astroglial coupling in an inflammatory coculture model. Glia, 20058597
  69. 69. NeijssenJet alCross-presentation by intercellular peptide transfer through gap junctions. Nature, 20058388
  70. 70. MatsueHet alGap junction-mediated intercellular communication between dendritic cells (DCs) is required for effective activation of DCs. J Immunol, 2006181190
  71. 71. CorvalanL. Aet alInjury of skeletal muscle and specific cytokines induce the expression of gap junction channels in mouse dendritic cells. J Cell Physiol, 2007649660
  72. 72. HandelAet alGap junction-mediated antigen transport in immune responses. Trends Immunol, 2007463466
  73. 73. Mendoza-naranjoAet alFunctional gap junctions facilitate melanoma antigen ransfer and cross-presentation between human dendritic cells. J Immunol, 200769496957
  74. 74. PangBet alDirect antigen presentation and gap junction mediated cross-presentation during apoptosis. J Immunol, 200910831090
  75. 75. EugeninE. Aet alThe Role of Gap Junction Channels During Physiologic and Pathologic Conditions of the Human Central Nervous System. J Neuroimmune Pharmacol, 2012
  76. 76. TakeuchiHet alTumor necrosis factor-alpha induces neurotoxicity via glutamate release from hemichannels of activated microglia in an autocrine manner. J Biol Chem, 20062136221368
  77. 77. TakeuchiHet alBlockade of microglial glutamate release protects against ischemic brain injury. Exp Neurol, 2008144146
  78. 78. ShijieJet alBlockade of glutamate release from microglia attenuates experimental autoimmune encephalomyelitis in mice. Tohoku J Exp Med, 20098792
  79. 79. MaezawaIet alRett syndrome astrocytes are abnormal and spread MeCP2 deficiency through gap junctions. J Neurosci, 200950515061
  80. 80. WangJet alModulation of membrane channel currents by gap junction protein mimetic peptides: size matters. Am J Physiol Cell Physiol, 2007C1112C1119
  81. 81. DahlGGap junction-mimetic peptides do work, but in unexpected ways. Cell Commun Adhes, 2007259264
  82. 82. EvansW. Hand LLeybaertMimetic peptides as blockers of connexin channel-facilitated intercellular communication. Cell Commun Adhes, 2007265273
  83. 83. OrellanaJ. Aet alAmyloid beta-induced death in neurons involves glial and neuronal hemichannels. J Neurosci, 201149624977
  84. 84. TakeuchiHet alBlockade of gap junction hemichannel suppresses disease progression in mouse models of amyotrophic lateral sclerosis and Alzheimer’s disease. PLoS One, 2011e21108
  85. 85. KangJet alConnexin 43 hemichannels are permeable to ATP. J Neurosci, 200847024711
  86. 86. SanchezH. Aet alMetabolic inhibition increases activity of connexin-32 hemichannels permeable to Ca2+ in transfected HeLa cells. Am J Physiol Cell Physiol, 2009C665C678
  87. 87. SzydlowskaKand MTymianskiCalcium, ischemia and excitotoxicity. Cell Calcium, 2010122129

Written By

Juan A. Orellana

Submitted: 20 April 2012 Published: 15 May 2013