Connexins in Fibrosis, EMTs, and Wound Healing: Comparison
Please note this is a comparison between Version 2 by Mona Zou and Version 1 by Yuting Li.

Fibrosis initially appears as a normal response to damage, where activated fibroblasts produce large amounts of the extracellular matrix (ECM) during the wound healing process to assist in the repair of injured tissue. However, the excessive accumulation of the ECM, unresolved by remodeling mechanisms, leads to organ dysfunction. Connexins, a family of transmembrane channel proteins, are widely recognized for their major roles in fibrosis, the epithelial–mesenchymal transition (EMT), and wound healing. Efforts have been made in recent years to identify novel mediators and targets for this regulation. Connexins form gap junctions and hemichannels, mediating communications between neighboring cells and inside and outside of cells, respectively. Recent evidence suggests that connexins, beyond forming channels, possess channel-independent functions in fibrosis, the EMT, and wound healing. One crucial channel-independent function is their role as the primary functional component for cell adhesion. Other channel-independent functions of connexins involve their roles in mitochondria and exosomes.

  • connexins
  • gap junctions
  • hemichannels
  • fibrosis
  • EMT
  • wound healing

1. Structure of Connexin

Connexins are transmembrane proteins. Approximately 20 types of connexin genes have been reported in humans and mice [1,2][1][2]. The corresponding genes are abbreviated as “GJ” (for gap junction), while the most commonly used protein nomenclature employs “Cx” (for connexin). Connexin proteins are often identified by the molecular mass of the predicted polypeptide in kilodaltons (e.g., connexin43, Cx43) [3]. Different connexin sequences have been used to recognize connexin subfamilies, with five connexin subfamilies defined (α, β, γ, δ, and ε in mice or GJA, GJB, GJC, GJD, and GJE in humans) [1], each of them containing distinct biophysical properties and expression patterns with various biological functions. All connexins have similar topological structural characteristics, consisting of four transmembrane domains: the amino (NT) and carboxyl termini (CTs) in the cytoplasm, a cytoplasmic loop (CL), and two extracellular loops (EL1 and EL2) [4,5][4][5]. The CTs are considered regulatory regions for multiple post-translational modifications and protein binding sites, exhibiting the greatest heterogeneity in amino acid sequences [6]. Every six connexin monomers form a hexagonal lattice of protein subunits, as seen under electron crystallography, called connexons or hemichannels (HCs) [7]. After trafficking to the membrane docks, two HCs from adjacent cells with a “head-to-head” pattern form an intercellular channel called a GJ [8]. These GJs and HCs allow the passage of small molecules (Mw < 1 kDa), including ions, essential metabolites, and second messengers such as Ca2+, IP3, nicotinamide adenine dinucleotide (NAD), prostaglandin E2 (PGE2), ATP, ADP, cAMP, and cGMP [9]. This type of intercellular communication is known to be essential for various physiological activities, including cell growth, proliferation, and differentiation. It also plays crucial roles in tissue homeostasis, tumorigenicity, fibrosis, and wound healing.

2. Channel-Dependent Functions of Cx in Fibrosis, EMTs, and Wound Healing

2.1. Connexin GJs and HCs in Eye Diseases

2.1.1. Cornea

The eye comprises the cornea, lens, uvea, retina, and several other subcomponents, within which various connexins exist. GJ communication in the human corneal epithelium is mediated by Cx26, Cx30, Cx31.1, and Cx43 [21,22][10][11]. Specifically, Cx43 has been associated with inflammatory responses and wound healing in injured corneas [23,24][12][13]. Cx43 knockdown can accelerate wound healing in the corneal endothelium and promote re-epithelialization by suppressing stromal oedema and inflammatory responses [25,26][14][15]. The interaction between Cx43 and zonula occludens-1 (ZO-1), a scaffolding protein that plays a role in regulating GJ assembly, has been extensively studied [27,28,29,30][16][17][18][19]. The alpha-carboxy terminus 1 (αCT1) peptide, derived from the carboxyl terminus of Cx43, disrupts Cx43 interaction with ZO-1, leading to an enhancement of GJs [31][20]. Treatment with αCT1 may play a role in the early stages of migration during corneal healing, as well as in affecting EMT pathway genes [32,33][21][22]. Cx43 antisense oligodeoxynucleotides (AsODNs), which knockdown Cx43 expression, have been shown to have a definite therapeutic effect in a reproducible model of corneal wound healing [26][15]. Cx43 AsODN-treated corneas show improved corneal clarity, faster re-epithelialization, and reduced inflammation. Additionally, the reduction in Cx43 GJ function with AsODNs in human nonhealing ocular burn wounds could enhance the recovery of limbal reperfusion and reduce inflammation, holding therapeutic potential for the treatment of severe, often unresponsive corneal injuries. This is achieved by targeting the downregulation of Cx43 to enable vascular recovery, making Cx43 a possible key factor in the treatment of wounds [34][23]. Moore et al. also found a downregulation of Cx43 in corneal wound healing. Between the 24- and 72-h time points, as the wound is healing, Cx43 is upregulated as cell–cell adhesion and GJ formation recur. By day 21, there was a downregulation in Cx43 [32][21]. The critical point is the time for treatment. Early use increases GJs, which are closely related to inhibiting inflammation and reducing edema. But in later stages, reducing over-expressed Cx43 can promote repair, speed healing, and prevent excessive scarring. Gap27 is a mimicking sequence on the second external loop of the Cx43 HC [9,35][9][24]. Its use in the early stages of wound healing for superficial corneal epithelial wounds has been proposed for promoting corneal epithelial healing, including persistent corneal ulcers, limbal stem cell deficiency, and dry eye syndrome. However, prolonged and frequent treatment in stromal wounds could elicit an undesirable inflammatory response [36][25].

2.1.2. Lens

The lens epithelial cells express Cx43 and Cx50, while fiber cells express Cx46 and Cx50 [37][26]. Past research primarily focuses on the function and regulation of GJs because these channels play essential roles in maintaining lens cell homeostasis, metabolic coupling, and preventing the accumulation of reactive oxidants. Connexin mutations are being identified and linked to numerous types of human congenital cataracts [38,39][27][28]. Abnormal activities of GJs and/or HCs would greatly compromise these crucial functions. Our group has previously reported that the G143R mutation increases the interaction between the intracellular loop domain and Cx46, which is associated with the reduction in GJs but an increase in HC function [40][29]. Additionally, Cx50 HCs activated by H2O2 mediate glutathione (GSH) transport and protect lens fiber cells from oxidative stress [41][30]. Mechanically activated Cx HCs and GSH transport can reduce H2O2- and UVB-induced intracellular reactive oxygen species (ROS), further mitigating cellular apoptosis and death [42][31]. Our recent study confirmed that protein kinase A (PKA) activation reduces cataracts induced by oxidative stress, increases GJs/HCs in Cx50, Cx46, or Cx50 and Cx46 co-expressing cells, and decreases ROS levels and cell death [43][32].
The researchers proposed a new molecular mechanism of HCs in lens epithelial cells that protects the lens against oxidative stress. The researchers found that Cx43 HCs mediate the exchange of oxidants and antioxidants in lens epithelial cells undergoing oxidative stress. These transporting activities facilitate a reduction in intracellular ROS accumulation and maintain intracellular glutathione levels through the exchange of redox metabolites and changes in anti-oxidative gene expression. Additionally, Cx43 HCs can be regulated by the intracellular redox state, and this regulation is mediated by residue Cys260 located at the Cx43 C-terminus [19][33]. All these findings demonstrate that GJs between lens fiber cells are an important mechanism for maintaining lens transparency by transmitting redox metabolites and that HCs assist in delivering nutrients and anti-oxidants into lens epithelial and fiber cells.
Posterior capsular opacification (PCO), a common complication following cataract surgery, results from the proliferation and degenerative fibrosis of residual lens epithelial cells [44][34]. The type-2 EMT and fibrosis are closely related to lens epithelial cells. Numerous studies have implicated transforming growth factor-β (TGF-β) in the fibrotic formation of PCO, including isoforms of TGFβ1 and TGFβ2 [45,46][35][36]. Additionally, there is a link between the inflammatory response to a lens injury and the mechanisms responsible for TGFβ1/β2 activation and the induction of fibrosis [47,48][37][38]. Moreover, cell adhesion signaling has also been implicated in the regulation of the EMT and fibrosis in the lens [49][39]. However, there is limited knowledge regarding the role of connexin channels in fibrosis. It is not clear whether GJs/HCs participate in the TGF-β-induced EMT of lens epithelial cells in response to an injury, including capsular scarring after cataract lens removal and intraocular lens implantation. Although TGF-β1 at a low concentration of 0.4 ng/mL markedly increases the expression of EMT markers, this amount is not sufficient to enhance GJ intercellular communication (GJIC) in chick embryonic lens epithelial cells. This finding has led to the deduction that the upregulation of GJIC by TGF-β is not an obvious downstream consequence of the TGF-β-induced EMT [50][40].

2.1.3. Retina

Connexins are present in the five neuronal types of the retina, vascular endothelial cells, pericytes, glial cells, and the retinal pigment epithelium (RPE) [51,52][41][42]. The neuronal types of the retina are composed of photoreceptors and horizontal, bipolar, amacrine, and ganglion cells. There is a large array of connexins on these cells in mice and humans, with previous research demonstrating the presence of several Cx subtypes, including Cx35, Cx36, Cx43, Cx45, Cx57, Cx59, among others [52,53,54,55][42][43][44][45]. Furthermore, studies have largely focused on the role connexins play in material transport and signaling transmission. Cxs are reported to play crucial roles in the fibrotic development of several chronic retinal diseases, such as diabetic retinopathy (DR), retinal ischemia, age-related macular degeneration (AMD), and RPE-related diseases [51,56][41][46]. It is also worth noting that all these diseases are associated with astrocytes, Müller cells, microglia, RPE, and vessel endothelial cells in conjunction with inflammatory processes and vascular responses [51][41]. However, the participation of neuronal retinal Cxs in wound healing and fibrosis has not been directly reported.
Cx43, the most ubiquitously expressed isoform in these three types of glial cells, RPE, and endothelial cells, plays an essential role in multiple cellular processes [53,57,58][43][47][48]. As an example, Tien et al. previously reported findings indicating that the high glucose-induced downregulation of Cx43 expression and GJIC may contribute to the breakdown of endothelial barrier tight junctions associated with DR [59][49]. They then showed that Cx43 protein expression and GJIC were significantly reduced in diabetic retinas compared to non-diabetic retinas. These results signify that the reduction in Cx43 is associated with increased vascular cell death in human diabetic retinas [60][50]. In other studies, Toychiev et al. investigated the outcome of an increase in Cx43 expression and GJ coupling during an acute ischemia/reperfusion injury, which exacerbated ganglion cell loss. The inhibition of astrocytic Cx43 channels may represent a useful strategy for promoting RGC survival in pathologic conditions [61][51].
GJs and functional HCs are major focus areas of prior research. Cx HCs have a low probability of being open under normal physiological conditions. However, when open in an undocked form, these HCs allow for the ubiquitous release of paracrine and autocrine signals [62,63][52][53]. Overactivated HCs have been associated with secondary tissue injuries, including edema, compromised vascular integrity, and retinal damage, by allowing the passage of molecules and ions between the cell cytoplasm and the extracellular milieu. They can also affect downstream GJ communication [51,56,64][41][46][54]. During inflammation, HC-mediated ATP release is highly prominent [65][55]. Certain peptides, such as low concentrations of Peptide5 and Gap19, have been used to block Cx43 HCs specifically without affecting GJ coupling. These tools appear to provide viable therapeutic options for treating retinal ischemia and retinal inflammatory diseases associated with Cx43 HC opening [62,66,67][52][56][57]. Meanwhile, the novel benzopyran derivative Tonabersat, a sodium channel blocker that also inhibits Cx43 HCs, is used in some inflammatory-related retinal diseases, including AMD. Previous work has shown that orally administered Tonabersat improved clinical signs in animal models of both DR and dry AMD and was associated with reduced inflammation. It was discovered that Tonabersat significantly preserved the function of the retina, particularly the function of photoreceptors and bipolar cells in the inner retina [68][58]. Tonabersat has also been shown to prevent the formation of NOD-like receptor protein 3 (NLRP3), an ATP-mediated inflammasome, and the cleaved caspase-1 complex assembly, as well as the release of the proinflammatory cytokines IL-1β, VEGF, and IL-6 [69][59]. Non-obese diabetic mice have been used to evaluate the ocular safety and efficacy of Tonabersat [70][60]. Overall, drug treatment significantly reduces macrovascular abnormalities, hyperreflective foci, sub-retinal fluid accumulation, vascular leakage, inflammation, and inflammasome activation. Lyon H et al. have found that Tonabersat also attenuates both TGF-β2 release and RPE EMTs under disease-mimicking conditions, providing a therapeutic target for diseases, such as DR, underlined by the EMT [71][61]. The demonstrated efficacy of Tonabersat could be attributed in part to the inhibition of Cx43 HCs. All these findings suggest that Cx HCs inhibitors may be effective tools and hold potential as therapeutics for multiple eye diseases related to inflammation or fibrosis.

2.2. Connexin GJ/HCs in the Central Nervous System

A disruption of the balance of GJs, HCs, and Cxs affects tissue homeostasis and allows cells to progress toward pathological conditions with varying degrees of severity in the central nervous system (CNS) [72][62]. The most abundant Cx in the brain is Cx43. Cx43 is prominently expressed in astrocytes but is also present in microglial cells [73,74][63][64]. Neuronal survival and physiological functions strictly depend on the maintenance of the blood–brain barrier (BBB). Astrocytes, which establish an elaborate network at the BBB, play critical roles in neuroprotection [75,76][65][66].
Cx HCs form an integrated network of channels in neurons and astrocytes. Most cell types in the CNS exhibit HC activity, while most studies revolve around Cx43 HCs in astrocytes [76][66]. The Cx43-HC-mediated release of gliotransmitters (ATP/glutamate) results in structural neuronal alterations and increased oxidative stress [77][67]. While strategies that promote axonal growth are still limited, other treatment paradigms that limit secondary damage, especially maintaining a balance between pro- and anti-inflammatory factors in incomplete spinal cord injuries (SCIs), can potentially allow patients to retain greater function post-injury [78,79][68][69]. During inflammation, Ca2+ signaling in the astrocyte network is elevated, resulting in increased ATP production and release through the opening of HCs. ATP stimulates purinoceptors, leading to increased Ca2+ release from internal stores [80][70]. This extracellular Ca2+ signaling attenuates intercellular Ca2+ signaling, causing reduced cell communication via GJs [81,82][71][72]. Similar to the treatment of retinal diseases mentioned before, GJ and HC inhibitors strongly stimulate research toward the development of new modulators to be used against CNS disorders [83][73]. On one hand, Cx43 blockers can be used to deal with neuropathic pain after SCIs [84,85][74][75]. On the other hand, channel inhibitors can reduce the spread of damage and improve functional recovery after an injury [86,87][76][77]. A spinal injection of Gap26 and Gap27 effectively reduces neuropathic pain symptoms (mechanical allodynia) by decreasing the release of the chemokines CCL2 and CXCL1 in the late phase (21 days) [85][75]. Peptide 5, by preventing HCs from opening, significantly reduces the degree of swelling and the loss of neurons in a concentration- and time-dependent manner [86][76]. Cx43 AsODN treatment can reduce glial cell activation, neutrophil recruitment, and the functional consequences of a spinal cord injury. Within 24 h of a compression injury, rats treated with Cx43 AsODNs scored higher than sense- and vehicle-treated controls on behavioral tests of locomotion. In rats tested for locomotor ability over the next 28 days, the initial improvement in scores was sustained [87][77]. Our group has developed a novel monoclonal antibody, MHC1, that targets Cx43 and specifically inhibits HC opening but has no effect on GJ coupling in two incomplete SCI mouse models. The researchers have shown that a single antibody administration within 30 min after an SCI significantly decreased secondary injury, improved locomotion function, attenuated gliosis, preserved white and gray matter, and protected neurons [88][78] (Figure 1).
Figure 1. Connexin GJs and HCs in eye diseases. The eye comprises the cornea, lens, retina, and several other subcomponents, within which various connexins exist. (A) Cx26, Cx30, Cx31.1, and Cx43 mediate GJ communication in the human corneal epithelium. ZO-1 is a scaffolding protein that plays a role in regulating GJ assembly. A disruption of Cx43 interaction with ZO-1 leads to the enhancement of GJs; targeting this disruption could be a means of enhancing wound healing. (B) Cx50 and AQP0, located at lens fibers, respectively, mediate cell–cell adhesion, maintaining lens fiber integrity. Deficiency of Cx50 and AQP0 leads to a loss in cell–cell adhesion, resulting in alterations of lens structures. At the posterior part of the lens, newly formed fibers cannot bundle properly, leading to lens posterior extrusion and capsule rupture. Macrophages are recruited to the vitreous cavity adjacent to the ruptured posterior capsule. M1 macrophages mainly mediate the removal of the tissue mass, while M2 macrophages play a crucial role in posterior capsule sealing and fibrosis. (C) The neuronal types of the retina are composed of photoreceptors, horizontal cells, bipolar, amacrine, and ganglion cells, all of which contain connexins. The participation of neuronal retinal Cxs in wound healing and fibrosis has not been directly reported. Tonabersat, a sodium channel blocker inhibiting Cx43 HCs, is used in some inflammatory-related retinal diseases. Tonabersat has also been shown to prevent the formation of NOD-like receptor protein 3 (NLRP3), an ATP-mediated inflammasome, as well as the release of other proinflammatory cytokines. Throughout the lens tissue, targeting Cxs for inflammation or fibrosis may hold therapeutic potential.

References

  1. Beyer, E.C.; Berthoud, V.M. Gap junction gene and protein families: Connexins, innexins, and pannexins. Biochim. Biophys. Acta Biomembr. 2018, 1860, 5–8.
  2. Laird, D.W.; Lampe, P.D. Therapeutic strategies targeting connexins. Nat. Rev. Drug Discov. 2018, 17, 905–921.
  3. Beyer, E.C.; Paul, D.L.; Goodenough, D.A. Connexin43: A protein from rat heart homologous to a gap junction protein from liver. J. Cell Biol. 1987, 105, 2621–2629.
  4. Kumar, N.M.; Gilula, N.B. The gap junction communication channel. Cell 1996, 84, 381–388.
  5. Yeager, M.; Harris, A.L. Gap junction channel structure in the early 21st century: Facts and fantasies. Curr. Opin. Cell Biol. 2007, 19, 521–528.
  6. Saez, J.C.; Berthoud, V.M.; Branes, M.C.; Martinez, A.D.; Beyer, E.C. Plasma membrane channels formed by connexins: Their regulation and functions. Physiol. Rev. 2003, 83, 1359–1400.
  7. Goodenough, D.A. The structure of cell membranes involved in intercellular communication. Am. J. Clin. Pathol. 1975, 63, 636–645.
  8. Delmar, M.; Laird, D.W.; Naus, C.C.; Nielsen, M.S.; Verselis, V.K.; White, T.W. Connexins and Disease. Cold Spring Harb. Perspect. Biol. 2018, 10, a029348.
  9. Goodenough, D.A.; Goliger, J.A.; Paul, D.L. Connexins, connexons, and intercellular communication. Annu. Rev. Biochem. 1996, 65, 475–502.
  10. Shurman, D.L.; Glazewski, L.; Gumpert, A.; Zieske, J.D.; Richard, G. In vivo and in vitro expression of connexins in the human corneal epithelium. Investig. Ophthalmol. Vis. Sci. 2005, 46, 1957–1965.
  11. Williams, K.; Watsky, M. Gap junctional communication in the human corneal endothelium and epithelium. Curr. Eye Res. 2002, 25, 29–36.
  12. Zhang, J.; Green, C.R.; Mugisho, O.O. Cell transdifferentiation in ocular disease: Potential role for connexin channels. Exp. Cell Res. 2021, 407, 112823.
  13. D’Hondt, C.; Iyyathurai, J.; Himpens, B.; Leybaert, L.; Bultynck, G. Cx43-hemichannel function and regulation in physiology and pathophysiology: Insights from the bovine corneal endothelial cell system and beyond. Front. Physiol. 2014, 5, 348.
  14. Nakano, Y.; Oyamada, M.; Dai, P.; Nakagami, T.; Kinoshita, S.; Takamatsu, T. Connexin43 knockdown accelerates wound healing but inhibits mesenchymal transition after corneal endothelial injury in vivo. Investig. Ophthalmol. Vis. Sci. 2008, 49, 93–104.
  15. Grupcheva, C.N.; Laux, W.T.; Rupenthal, I.D.; McGhee, J.; McGhee, C.N.; Green, C.R. Improved corneal wound healing through modulation of gap junction communication using connexin43-specific antisense oligodeoxynucleotides. Investig. Ophthalmol. Vis. Sci. 2012, 53, 1130–1138.
  16. Giepmans, B.N.G.; Verlann, I.; Hengeveld, T.; Janssen, H.; Calafat, J.; Falk, M.M.; Moolenaar, W.H. Gap junction protein connexin-43 interacts directly with microtubules. Curr. Biol. 2001, 11, 1364–1368.
  17. Hunter, A.L.; Choy, J.C.; Granville, D.J. Detection of apoptosis in cardiovascular diseases. Methods Mol. Med. 2005, 112, 277–289.
  18. Rhett, J.M.; Jourdan, J.; Gourdie, R.G. Connexin 43 connexon to gap junction transition is regulated by zonula occludens-1. Mol. Biol. Cell 2011, 22, 1516–1528.
  19. Sorgen, P.L.; Duffy, H.S.; Sahoo, P.; Coombs, W.; Delmar, M.; Spray, D.C. Structural changes in the carboxyl terminus of the gap junction protein connexin43 indicates signaling between binding domains for c-Src and zonula occludens-1. J. Biol. Chem. 2004, 279, 54695–54701.
  20. Hunter, A.W.; Barker, R.J.; Zhu, C.; Gourdie, R.G. Zonula occludens-1 alters connexin43 gap junction size and organization by influencing channel accretion. Mol. Biol. Cell 2005, 16, 5686–5698.
  21. Moore, K.; Bryant, Z.J.; Ghatnekar, G.; Singh, U.P.; Gourdie, R.G.; Potts, J.D. A synthetic connexin 43 mimetic peptide augments corneal wound healing. Exp. Eye Res. 2013, 115, 178–188.
  22. Rhett, J.M.; Ghatnekar, G.S.; Palatinus, J.A.; O’Quinn, M.; Yost, M.J.; Gourdie, R.G. Novel therapies for scar reduction and regenerative healing of skin wounds. Trends Biotechnol. 2008, 26, 173–180.
  23. Ormonde, S.; Chou, C.Y.; Goold, L.; Petsoglou, C.; Al-Taie, R.; Sherwin, T.; McGhee, C.N.; Green, C.R. Regulation of connexin43 gap junction protein triggers vascular recovery and healing in human ocular persistent epithelial defect wounds. J. Membr. Biol. 2012, 245, 381–388.
  24. Evans, W.H.; Boitano, S. Connexin mimetic peptides: Specific inhibitors of gap-junctional intercellular communication. Biochem. Soc. Trans. 2001, 29, 606–612.
  25. Elbadawy, H.M.; Mirabelli, P.; Xeroudaki, M.; Parekh, M.; Bertolin, M.; Breda, C.; Cagini, C.; Ponzin, D.; Lagali, N.; Ferrari, S. Effect of connexin 43 inhibition by the mimetic peptide Gap27 on corneal wound healing, inflammation and neovascularization. Br. J. Pharmacol. 2016, 173, 2880–2893.
  26. Beyer, E.C.; Berthoud, V.M. Connexin hemichannels in the lens. Front. Physiol. 2014, 5, 20.
  27. Berthoud, V.M.; Gao, J.; Minogue, P.J.; Jara, O.; Mathias, R.T.; Beyer, E.C. Connexin Mutants Compromise the Lens Circulation and Cause Cataracts through Biomineralization. Int. J. Mol. Sci. 2020, 21, 5822.
  28. Jiang, J.X. Gap junctions or hemichannel-dependent and independent roles of connexins in cataractogenesis and lens development. Curr. Mol. Med. 2010, 10, 851–863.
  29. Ren, Q.; Riquelme, M.A.; Xu, J.; Yan, X.; Nicholson, B.J.; Gu, S.; Jiang, J.X. Cataract-causing mutation of human connexin 46 impairs gap junction, but increases hemichannel function and cell death. PLoS ONE 2013, 8, e74732.
  30. Shi, W.; Riquelme, M.A.; Gu, S.; Jiang, J.X. Connexin hemichannels mediate glutathione transport and protect lens fiber cells from oxidative stress. J. Cell Sci. 2018, 131, jcs212506.
  31. Liu, J.; Riquelme, M.A.; Li, Z.; Li, Y.; Tong, Y.; Quan, Y.; Pei, C.; Gu, S.; Jiang, J.X. Mechanosensitive collaboration between integrins and connexins allows nutrient and antioxidant transport into the lens. J. Cell Biol. 2020, 219, e202002154.
  32. Du, Y.; Tong, Y.; Quan, Y.; Wang, G.; Cheng, H.; Gu, S.; Jiang, J.X. Protein kinase A activation alleviates cataract formation via increased gap junction intercellular communication. iScience 2023, 26, 106114.
  33. Quan, Y.; Du, Y.; Wu, C.; Gu, S.; Jiang, J.X. Connexin hemichannels regulate redox potential via metabolite exchange and protect lens against cellular oxidative damage. Redox Biol. 2021, 46, 102102.
  34. Apple, D.J.; Solomon, K.D.; Tetz, M.R.; Assia, E.I.; Holland, E.Y.; Legler, U.F.; Tsai, J.C.; Castaneda, V.E.; Hoggatt, J.P.; Kostick, A.M. Posterior capsule opacification. Surv. Ophthalmol. 1992, 37, 73–116.
  35. Wormstone, I.M.; Tamiya, S.; Anderson, I.; Duncan, G. TGF-beta2-induced matrix modification and cell transdifferentiation in the human lens capsular bag. Investig. Ophthalmol. Vis. Sci. 2002, 43, 2301–2308.
  36. Boswell, B.A.; Korol, A.; West-Mays, J.A.; Musil, L.S. Dual function of TGFbeta in lens epithelial cell fate: Implications for secondary cataract. Mol. Biol. Cell 2017, 28, 907–921.
  37. Jiang, J.; Shihan, M.H.; Wang, Y.; Duncan, M.K. Lens Epithelial Cells Initiate an Inflammatory Response Following Cataract Surgery. Investig. Ophthalmol. Vis. Sci. 2018, 59, 4986–4997.
  38. Stepp, M.A.; Menko, A.S. Immune responses to injury and their links to eye disease. Transl. Res. 2021, 236, 52–71.
  39. Taiyab, A.; West-Mays, J. Lens Fibrosis: Understanding the Dynamics of Cell Adhesion Signaling in Lens Epithelial-Mesenchymal Transition. Front. Cell Dev. Biol. 2022, 10, 886053.
  40. Boswell, B.A.; VanSlyke, J.K.; Musil, L.S. Regulation of lens gap junctions by Transforming Growth Factor beta. Mol. Biol. Cell 2010, 21, 1686–1697.
  41. Danesh-Meyer, H.V.; Zhang, J.; Acosta, M.L.; Rupenthal, I.D.; Green, C.R. Connexin43 in retinal injury and disease. Prog. Retin. Eye Res. 2016, 51, 41–68.
  42. Gonzalez-Casanova, J.; Schmachtenberg, O.; Martinez, A.D.; Sanchez, H.A.; Harcha, P.A.; Rojas-Gomez, D. An Update on Connexin Gap Junction and Hemichannels in Diabetic Retinopathy. Int. J. Mol. Sci. 2021, 22, 3194.
  43. Sohl, G.; Joussen, A.; Kociok, N.; Willecke, K. Expression of connexin genes in the human retina. BMC Ophthalmol. 2010, 10, 27.
  44. Harrison, K.R.; Chervenak, A.P.; Resnick, S.M.; Reifler, A.N.; Wong, K.Y. Amacrine Cells Forming Gap Junctions With Intrinsically Photosensitive Retinal Ganglion Cells: ipRGC Types, Neuromodulator Contents, and Connexin Isoform. Investig. Ophthalmol. Vis. Sci. 2021, 62, 10.
  45. Guldenagel, M.; Sohl, G.; Plum, A.; Traub, O.; Teubner, B.; Weiler, R.; Willecke, K. Expression patterns of connexin genes in mouse retina. J. Comp. Neurol. 2000, 425, 193–201.
  46. Roy, S.; Jiang, J.X.; Li, A.F.; Kim, D. Connexin channel and its role in diabetic retinopathy. Prog. Retin. Eye Res. 2017, 61, 35–59.
  47. Kerr, N.M.; Johnson, C.S.; Green, C.R.; Danesh-Meyer, H.V. Gap junction protein connexin43 (GJA1) in the human glaucomatous optic nerve head and retina. J. Clin. Neurosci. 2011, 18, 102–108.
  48. Mendes-Jorge, L.; Llombart, C.; Ramos, D.; Lopez-Luppo, M.; Valenca, A.; Nacher, V.; Navarro, M.; Carretero, A.; Mendez-Ferrer, S.; Rodriguez-Baeza, A.; et al. Intercapillary bridging cells: Immunocytochemical characteristics of cells that connect blood vessels in the retina. Exp. Eye Res. 2012, 98, 79–87.
  49. Tien, T.; Barrette, K.F.; Chronopoulos, A.; Roy, S. Effects of high glucose-induced Cx43 downregulation on occludin and ZO-1 expression and tight junction barrier function in retinal endothelial cells. Investig. Ophthalmol. Vis. Sci. 2013, 54, 6518–6525.
  50. Tien, T.; Muto, T.; Zhang, J.; Sohn, E.H.; Mullins, R.F.; Roy, S. Association of reduced Connexin 43 expression with retinal vascular lesions in human diabetic retinopathy. Exp. Eye Res. 2016, 146, 103–106.
  51. Toychiev, A.H.; Batsuuri, K.; Srinivas, M. Gap Junctional Coupling Between Retinal Astrocytes Exacerbates Neuronal Damage in Ischemia-Reperfusion Injury. Investig. Ophthalmol. Vis. Sci. 2021, 62, 27.
  52. Abudara, V.; Bechberger, J.; Freitas-Andrade, M.; De Bock, M.; Wang, N.; Bultynck, G.; Naus, C.C.; Leybaert, L.; Giaume, C. The connexin43 mimetic peptide Gap19 inhibits hemichannels without altering gap junctional communication in astrocytes. Front. Cell Neurosci. 2014, 8, 306.
  53. Orellana, J.A.; Retamal, M.A.; Moraga-Amaro, R.; Stehberg, J. Role of Astroglial Hemichannels and Pannexons in Memory and Neurodegenerative Diseases. Front. Integr. Neurosci. 2016, 10, 26.
  54. Acosta, M.L.; Mat Nor, M.N.; 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.
  55. 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.
  56. Guo, C.X.; Mat Nor, M.N.; Danesh-Meyer, H.V.; Vessey, K.A.; Fletcher, E.L.; O’Carroll, S.J.; Acosta, M.L.; Green, C.R. Connexin43 Mimetic Peptide Improves Retinal Function and Reduces Inflammation in a Light-Damaged Albino Rat Model. Investig. Ophthalmol. Vis. Sci. 2016, 57, 3961–3973.
  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.
  58. Mat Nor, M.N.; Rupenthal, I.D.; Green, C.R.; Acosta, M.L. Connexin Hemichannel Block Using Orally Delivered Tonabersat Improves Outcomes in Animal Models of Retinal Disease. Neurotherapeutics 2020, 17, 371–387.
  59. Lyon, H.; Shome, A.; Rupenthal, I.D.; Green, C.R.; Mugisho, O.O. Tonabersat Inhibits Connexin43 Hemichannel Opening and Inflammasome Activation in an In Vitro Retinal Epithelial Cell Model of Diabetic Retinopathy. Int. J. Mol. Sci. 2020, 22, 298.
  60. Mugisho, O.O.; Aryal, J.; Shorne, A.; Lyon, H.; Acosta, M.L.; Green, C.R.; Rupenthal, I.D. Orally Delivered Connexin43 Hemichannel Blocker, Tonabersat, Inhibits Vascular Breakdown and Inflammasome Activation in a Mouse Model of Diabetic Retinopathy. Int. J. Mol. Sci. 2023, 24, 3876.
  61. Lyon, H.; Yin, N.; Rupenthal, I.D.; Green, C.R.; Mugisho, O.O. Blocking connexin43 hemichannels prevents TGF-beta2 upregulation and epithelial-mesenchymal transition in retinal pigment epithelial cells. Cell Biol. Int. 2022, 46, 323–330.
  62. Vicario, N.; Zappala, A.; Calabrese, G.; Gulino, R.; Parenti, C.; Gulisano, M.; Parenti, R. Connexins in the Central Nervous System: Physiological Traits and Neuroprotective Targets. Front. Physiol. 2017, 8, 1060.
  63. Giaume, C.; Theis, M. Pharmacological and genetic approaches to study connexin-mediated channels in glial cells of the central nervous system. Brain Res. Rev. 2010, 63, 160–176.
  64. Panattoni, G.; Amoriello, R.; Memo, C.; Thalhammer, A.; Ballerini, C.; Ballerini, L. Diverse inflammatory threats modulate astrocytes Ca(2+) signaling via connexin43 hemichannels in organotypic spinal slices. Mol. Brain 2021, 14, 159.
  65. Sanmarco, L.M.; Polonio, C.M.; Wheeler, M.A.; Quintana, F.J. Functional immune cell-astrocyte interactions. J. Exp. Med. 2021, 218, e20202715.
  66. Vicario, N.; Parenti, R. Connexins Signatures of the Neurovascular Unit and Their Physio-Pathological Functions. Int. J. Mol. Sci. 2022, 23, 9510.
  67. 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.
  68. Liu, Y.D.; Tang, G.; Qian, F.; Liu, L.; Huang, J.R.; Tang, F.R. Astroglial Connexins in Neurological and Neuropsychological Disorders and Radiation Exposure. Curr. Med. Chem. 2021, 28, 1970–1986.
  69. Goldshmit, Y.; Jona, G.; Schmukler, E.; Solomon, S.; Pinkas-Kramarski, R.; Ruban, A. Blood Glutamate Scavenger as a Novel Neuroprotective Treatment in Spinal Cord Injury. J. Neurotrauma 2018, 35, 2581–2590.
  70. Zorec, R.; Araque, A.; Carmignoto, G.; Haydon, P.G.; Verkhratsky, A.; Parpura, V. Astroglial excitability and gliotransmission: An appraisal of Ca2+ as a signalling route. ASN Neuro 2012, 4, e00080.
  71. Hansson, E.; Skiöldebrand, E. Coupled cell networks are target cells of inflammation, which can spread between different body organs and develop into systemic chronic inflammation. J. Inflamm. 2015, 12, 44.
  72. Karpuk, N.; Burkovetskaya, M.; Fritz, T.; Angle, A.; Kielian, T. Neuroinflammation leads to region-dependent alterations in astrocyte gap junction communication and hemichannel activity. J. Neurosci. 2011, 31, 414–425.
  73. Abou-Mrad, Z.; Alomari, S.O.; Bsat, S.; Moussalem, C.K.; Alok, K.; El Houshiemy, M.N.; Alomari, A.O.; Minassian, G.B.; Omeis, I.A. Role of connexins in spinal cord injury: An update. Clin. Neurol. Neurosurg. 2020, 197, 106102.
  74. Chen, M.J.; Kress, B.; Han, X.; Moll, K.; Peng, W.; Ji, R.R.; Nedergaard, M. Astrocytic CX43 hemichannels and gap junctions play a crucial role in development of chronic neuropathic pain following spinal cord injury. Glia 2012, 60, 1660–1670.
  75. Chen, G.; Park, C.K.; Xie, R.G.; Berta, T.; Nedergaard, M.; Ji, R.R. Connexin-43 induces chemokine release from spinal cord astrocytes to maintain late-phase neuropathic pain in mice. Brain 2014, 137, 2193–2209.
  76. O’Carroll, S.J.; Alkadhi, M.; Nicholson, L.F.; Green, C.R. Connexin 43 mimetic peptides reduce swelling, astrogliosis, and neuronal cell death after spinal cord injury. Cell Commun. Adhes. 2008, 15, 27–42.
  77. Cronin, M.; Anderson, P.N.; Cook, J.E.; Green, C.R.; Becker, D.L. Blocking connexin43 expression reduces inflammation and improves functional recovery after spinal cord injury. Mol. Cell Neurosci. 2008, 39, 152–160.
  78. Zhang, C.; Yan, Z.; Maknojia, A.; Riquelme, M.A.; Gu, S.; Booher, G.; Wallace, D.J.; Bartanusz, V.; Goswami, A.; Xiong, W.; et al. Inhibition of astrocyte hemichannel improves recovery from spinal cord injury. JCI Insight 2021, 6, e134611.
More
Video Production Service