Article

Dynamic trafficking and delivery of connexons to the plasma membrane and accretion to gap junctions in living cells

Authors:
To read the full-text of this research, you can request a copy directly from the authors.

Abstract

Certain membrane channels including acetylcholine receptors, gap junction (GJ) channels, and aquaporins arrange into large clusters in the plasma membrane (PM). However, how these channels are recruited to the clusters is unknown. To address this question, we have investigated delivery of GJ channel subunits (connexons) assembled from green fluorescent protein (GFP)-tagged connexin 43 (Cx43) to the PM and GJs in living cells. Fluorescence-photobleaching of distinct areas of Cx43-GFP GJs demonstrated that newly synthesized channels were accrued to the outer margins of channel clusters. Time-lapse microscopy further revealed that connexons were delivered in vesicular carriers traveling along microtubules from the Golgi to the PM. Routing and insertion of connexons occurred predominantly into the nonjunctional PM. These PM connexons can move laterally as shown by photo-bleaching and thus, can reach the margins of channel clusters. There, the apposing PMs are close enough to allow connexons to dock into complete GJ channels. When connexon delivery to the PM was inhibited by brefeldin A, or nocodazole pretreatment, the PM pool initially enabled connexon accrual to the clusters but further accrual was inhibited upon depletion. Taken together, our results indicate that GJ channel clusters grow by accretion at their outer margins from connexon subunits that were delivered to the nonjunctional PM, and explain how connexons in the PM can function in intra-/extracellular signaling before GJ channel formation and direct cell-cell communication.

No full-text available

Request Full-text Paper PDF

To read the full-text of this research,
you can request a copy directly from the authors.

... It has been suggested that actin is directly involved in the Cx43 transport process [83][84][85]. The hemichannels synthesized in the trans-Golgi are first transported along microtubules to the perinexus region, a peripheral cell membrane containing unconnected hemichannels around the GJPs. ...
... First, Cx43-containing vesicles can fuse with the plasma membrane of isolated cells [92]. Second, the presence of Cx43 was detected on the non-connected surfaces of cells and the membrane mobility of Cx43 could be calculated [83]. Four pieces of evidence supporting the second transport model are as follows. ...
... In terms of the site that fuses to the membrane, Lauf et al. [83] suggested that Cx43 transport vesicles randomly fuse with the plasma membrane of non-GJPs and then directionally move to form GJPs, since no transport of hemichannels in any specific direction after leaving the Golgi apparatus was observed and hemichannels transported to the plasma membrane of non-GJPs could be observed to freely move ( Figure 1). This is consistent with electron microscopy images that indicate the presence of intermembrane particles dispersed around GJPs [93,94], which may act as a reserve pool of Cxs to complement GJPs. ...
Article
Full-text available
The connexin gene family is the most prevalent gene that contributes to hearing loss. Connexins 26 and 30, encoded by GJB2 and GJB6, respectively, are the most abundantly expressed connexins in the inner ear. Connexin 43, which is encoded by GJA1, appears to be widely expressed in various organs, including the heart, skin, the brain, and the inner ear. The mutations that arise in GJB2, GJB6, and GJA1 can all result in comprehensive or non-comprehensive genetic deafness in newborns. As it is predicted that connexins include at least 20 isoforms in humans, the biosynthesis, structural composition, and degradation of connexins must be precisely regulated so that the gap junctions can properly operate. Certain mutations result in connexins possessing a faulty subcellular localization, failing to transport to the cell membrane and preventing gap junction formation, ultimately leading to connexin dysfunction and hearing loss. In this review, we provide a discussion of the transport models for connexin 43, connexins 30 and 26, mutations affecting trafficking pathways of these connexins, the existing controversies in the trafficking pathways of connexins, and the molecules involved in connexin trafficking and their functions. This review can contribute to a new way of understanding the etiological principles of connexin mutations and finding therapeutic strategies for hereditary deafness.
... It has been suggested that actin is directly involved in the Cx43 transport process [61][62][63]. The hemichannels synthesized in the trans-Golgi are first transported along microtubules to the perinexus region, a peripheral cell membrane containing unconnected hemichannels around the GJPs. ...
... First, Cx43-containing vesicles can fuse with the plasma membrane of isolated cells [70]. Second, the presence of Cx43 was detected on the non-connected surfaces of cells and the membrane mobility of Cx43 could be calculated [61]. Four pieces of evidence supporting the second transport model are as follows. ...
... Then hemichannels interact with cortical actin via ZO-1/2 [15,28], which regulates the directional transport of hemichannels from the perinexus region to the GJPs. In terms of the site that fuses to the membrane, Lauf et al. [61] suggested that Cx43 transport vesicles fuse randomly with the plasma membrane of non-GJPs and then move directionally to form GJPs since no transport of hemichannels in any specific direction after leaving the Golgi apparatus was observed and hemichannels transported to the plasma membrane of non-GJPs could be observed to move freely ( Figure 1). This is consistent with electron microscopy images that indicate the presence of intermembrane particles dispersed around GJPs [71,72], which may act as a reserve pool of Cxs to complement GJPs. ...
Preprint
Full-text available
The connexin gene family is the most prevalent gene that contributes to hearing loss. Connexins 26 and 30, encoded by GJB2 and GJB6 respectively, are the most abundant expressed connexins in the inner ear. Connexin 43 which is encoded by GJA1 appears to be widely expressed in various organs, including the heart, skin, brain, and inner ear. The mutations that arise in GJB2, GJB6 and GJA1 can all result in comprehensive or non-comprehensive genetic deafness in newborns. As it is predicted that connexins include at least 20 isoforms in humans, the biosynthesis, structural composition, and degradation of connexins must be precisely regulated so that the gap junctions can operate properly. Certain mutations result in connexins possessing a faulty subcellular localization, failing to transport to the cell membrane and preventing gap junction formation, ultimately leading to connexin dysfunction and hearing loss. In this review, we provide a discussion of the transport models for connexin 43, connexins 30 and 26, mutations affecting trafficking pathways of connexin 26, the existing controversies in trafficking pathways of connexins, and the molecules involved in connexin trafficking and their functions. This review can contribute to a new way of understanding the etiological principles of connexin mutations and finding therapeutic strategies for hereditary deafness.
... Les études ultérieures, réalisées également par cryofracture et microscopie électronique ont montré que ces striations provenaient d'ensembles, constitués de centaines à plusieurs milliers de canaux intercellulaires, qui connectent le cytoplasme d'une cellule au cytoplasme de la cellule voisine (Goodenough et al., 1970, Benedetti et al., 1965. Les jonctions communicantes se regroupent ainsi pour former des plaques jonctionnelles constituées de milliers de jonctions gap rapprochées les unes des autres, donnant ainsi une concentration pouvant aller jusqu'à 10 4 canaux/µm 2 de membrane (Lauf et al., 2002). Le diamètre de ces plaques varie de 100 nm à plusieurs µm (Segretain et al., 2004). ...
... Lauf et al. ont étudié la distribution des connexons assemblés à partir de Cx43 couplées à la green fluorescent protein (GFP) dans des cellules HeLa transfectées (Lauf et al., 2002). La microscopie temporelle en marquages multiples a permis d'observer le transport des connexons à l'intérieur de vésicules, le long des microtubules, depuis l'appareil de Golgi vers la membrane cytoplasmique. ...
... Gaietta et al. ont démontré que les jonctions gap nouvellement formées se trouvent sur la couronne externe de la plaque tandis que les canaux les plus anciens sont eux au centre des plaques (Gaietta et al., 2002). Les canaux sont donc distinctement localisés en fonction de leur âge (Lauf et al., 2002). Actuellement, on suppose que les canaux au centre des plaques sont destinés à être internalisés puis dégradés (Laird, 2006). ...
Thesis
Ce travail de recherche s'inscrit dans un axe de diagnostic du cancer via le développement d'une méthode optique pour la caractérisation fonctionnelle de tissus. La technique de gap- FRAP (Fluorescence Recovery After Photobleaching) permet l'étude quantitative de la fonctionnalité des jonctions gap. La majorité des cellules néoplasiques se caractérisent par une modification du niveau d'expression et/ou de la fonctionnalité des jonctions gap par comparaison à leurs homologues saines. La technique de gap-FRAP permet en conséquence de discriminer les cellules cancéreuses en fonction de la communication intercellulaire gap jonctionnelle (CIGJ). Particulièrement utilisée in vitro, cette technique restait cependant anecdotique ex vivo. Nous avons validé la faisabilité du transfert de cette méthode sur tissus et organes ex vivo. A partir de cellules de statuts différents en expression et en distribution des connexines, nous avons caractérisé la calcéine-AM comme étant une sonde fluorescente adaptée pour des mesures sur tissus. Puis nous avons développé un modèle d'ingénierie systéme pour l'analyse comparative des données de recouvrement de fluorescence sur des modèles bi et tridimensionnels. Nous avons transposé ces conditions préalablement définies sur organe entier ex vivo : la vessie de rat. Un marquage multiple a été optimisé avec une sonde fluorescente pour le tracking des cellules cancéreuses dans la vessie ex vivo, un marqueur pour l'identification histologique de l'urothélium et la calcéine-AM pour mesurer la CIGJ. Le gap-FRAP a été utilisé pour la première fois pour différencier le degré de communication intercellulaire gap jonctionnelle entre le tissu sain et néoplasique sur un organe entier ex vivo, ouvrant des perspectives pour le diagnostic du cancer de la vessie corrélé à la modification de la CIGJ.
... The microscope was equipped with an incubation chamber to maintain cells at 37 °C in DMEM without phenol red. Since previous reports [20], [28] have described connexon replenishment occurring at the lateral ends of the gap-junction plaque, this was selected as the region of interest (ROI). ROIs were manually drawn at the non-junctional plasma membrane and lateral ends of the gap-junction plaque using the rectangle tool. ...
... Targeted direct delivery of connexons to the GJP relies on a complex of cytoskeletal and adherence-junction proteins [32]. Alternatively, connexons are routed to the plasma membrane and subsequently recruited into the GJP at the lateral ends [28], [33], [34]. This process occurs despite the ability of microtubules to anchor directly at the GJP [21]. ...
... After bleaching, we observed that the plasma membrane (PM) of wild-type Cx36 recovered more efficiently than at the GJP (mobile fraction (Mf) in %, GJP: 25.6 ± 0.6, n = 28; PM: 69.6 ± 3.6 n = 12, p < 0.01 × 10 −2 ; half time of recovery (T1/2), GJP: 11.5s; PM: 23.5s) ( Figure 4A,D). The higher Mf seen at the PM suggested that, under control conditions, Cx36 is initially delivered to the plasma membrane and subsequently routed to the plaque, where it becomes stabilized, supporting the mechanisms previously described [28], [34], [35]. In Cx36Δ279-292-expressing cells, the Mf was again higher at the PM than at the GJP (GJP: 50 ± 0.5%, n = 22; PM: 97.1 ± 1.9%, n = 10, p < 0.01 × 10 −2 ;), suggesting that no changes were made to the route of delivery. ...
Article
Full-text available
Connexin-36 (Cx36) electrical synapses strengthen transmission in a calcium/calmodulin (CaM)/calmodulin-dependent kinase II (CaMKII)-dependent manner similar to a mechanism whereby the N-methyl-D-aspartate (NMDA) receptor subunit NR2B facilitates chemical transmission. Since NR2B–microtubule interactions recruit receptors to the cell membrane during plasticity, we hypothesized an analogous modality for Cx36. We determined that Cx36 binding to tubulin at the carboxy-terminal domain was distinct from Cx43 and NR2B by binding a motif overlapping with the CaM and CaMKII binding motifs. Dual patch-clamp recordings demonstrated that pharmacological interference of the cytoskeleton and deleting the binding motif at the Cx36 carboxyl-terminal (CT) reversibly abolished Cx36 plasticity. Mechanistic details of trafficking to the gap-junction plaque (GJP) were probed pharmacologically and through mutational analysis, all of which affected GJP size and formation between cell pairs. Lys279, Ile280, and Lys281 positions were particularly critical. This study demonstrates that tubulin-dependent transport of Cx36 potentiates synaptic strength by delivering channels to GJPs, reinforcing the role of protein transport at chemical and electrical synapses to fine-tune communication between neurons.
... Many connexins have multiple phosphorylation sites for several kinases like the mitogen-activated protein kinases ERK1/2, the protein-kinase C (PKC), the casein kinase 1 (CK1), the serine/threonine-protein kinase Akt or the src protein-tyrosine kinase. Besides the channel opening prevalence, phosphorylation events determine disassembly and internalization of the gap junction plaque: Phosphorylated connexins disband from gap junction plaques, whereas unphosphorylated proteins remain in plaques and provide the means for intercellular gap junction communication [32,61]. [51]. ...
... As outlined below, cancerous cells undergo changes in connexin expression and these are likely to be reflected in morphological changes of gap junction plaques. Even in normal cells, gap junctions show a permanent assembly and turnover of particles and are therefore considered highly flexible components in the regulation of physiological processes like cell proliferation and differentiation as well as the maintenance of homeostasis [32,39,60,61]. Accordingly, gap junction morphology is crucial for gap junction function [62]. ...
... Many connexins have multiple phosphorylation sites for several kinases like the mitogen-activated protein kinases ERK1/2, the protein-kinase C (PKC), the casein kinase 1 (CK1), the serine/threonine-protein kinase Akt or the src protein-tyrosine kinase. Besides the channel opening prevalence, phosphorylation events determine disassembly and internalization of the gap junction plaque: Phosphorylated connexins disband from gap junction plaques, whereas unphosphorylated proteins remain in plaques and provide the means for intercellular gap junction communication [32,61]. ...
Article
Full-text available
Gap junction proteins are expressed in cancer stem cells and non-stem cancer cells of many tumors. As the morphology and assembly of gap junction channels are crucial for their function in intercellular communication, one focus of our review is to outline the data on gap junction plaque morphology available for cancer cells. Electron microscopic studies and freeze-fracture analyses on gap junction ultrastructure in cancer are summarized. As the presence of gap junctions is relevant in solid tumors, we exemplarily outline their role in glioblastomas and in breast cancer. These were also shown to contain cancer stem cells, which are an essential cause of tumor onset and of tumor transmission into metastases. For these processes, gap junctional communication was shown to be important and thus we summarize, how the expression of gap junction proteins and the resulting communication between cancer stem cells and their surrounding cells contributes to the dissemination of cancer stem cells via blood or lymphatic vessels. Based on their importance for tumors and metastases, future cancer-specific therapies are expected to address gap junction proteins. In turn, gap junctions also seem to contribute to the unattainability of cancer stem cells by certain treatments and might thus contribute to therapeutic resistance.
... First, hexameric connexons assembled in the trans-Golgi network are trafficked along microtubules to non-junctional plasma membrane. 21,22 Second, hemichannels associate with cortical actin through actin-binding proteins zonula occludens which regulate delivery of connexins from the periphery to preexisting gap junction plaque (GJP). 23,24 This peripheral membrane region containing non-junctional hemichannels and surrounding the GJP is called "perinexus." 25 Besides mutations that affect the channel function itself, many of the disease-causing mutations in GJB2 or GJB6 impair the trafficking and assembly of Cx26 and Cx30, what prevents the formation of gap junctions. ...
... 51 Because of the relatively short half-life of connexins (usually 1-5 h), the GJP is in a dynamic state, constantly remodeled through both recruitment of newly synthesized hemichannels to the outer periphery of the GJP and endocytosis of older components from the center of the plaque. 21,52 In agreement with the latter model, we could suggest that, owing to low affinity of Cx26 for cholesterol abundantly present in lipid rafts, 39,40 Cx26/Cx30 hemichannels rapidly diffuse laterally out of lipid raft-enriched tricellular junctions and accrue along the outer periphery of the pre-existing GJP, where each hemichannel docks with another from the neighboring cell to form a new heteromeric channel. The inhibition of N-cadherin-based homophilic interactions induced a significant reduction in the summed Cx26/Cx30 GJP length per inner sulcus cell and a concomitant accumulation of Cx26/Cx30 oligomers within the cytoplasm. ...
Article
Full-text available
In the cochlea, connexin 26 (Cx26) and connexin 30 (Cx30) co‐assemble into two types of homomeric and heteromeric gap junctions between adjacent non‐sensory epithelial cells. These channels provide a mechanical coupling between connected cells, and their activity is critical to maintain cochlear homeostasis. Many of the mutations in GJB2 or GJB6, which encode Cx26 and Cx30 in humans, impair the formation of membrane channels and cause autosomal syndromic and non‐syndromic hearing loss. Thus, deciphering the connexin trafficking pathways in situ should represent a major step forward in understanding the pathogenic significance of many of these mutations. A growing body of evidence now suggests that Cx26/Cx30 heteromeric and Cx30 homomeric channels display distinct assembly mechanisms. Here, we review the most recent advances that have been made toward unraveling the biogenesis and stability of these gap junctions in the cochlea.
... Since Cx43 s normal life cycle includes incorporation into a gap junction followed by degradation ([32] i.e., there does not seem to be a nonjunctional and junctional population of Cx43 with different half lives), gap junctions are in a constant state of flux involving assembly, remodeling, and Cx43 turnover [33][34][35]. Live cell imaging of Cx43-fluorescent protein fusions (Cx43-FP) show that gap junction plaques are highly dynamic exhibiting growth via lateral accretion of connexons into the plaque and fusion of small plaques [36][37][38]. The organization of channels within the plaque can also be quite dynamic but appear to be regulated; there are clear experiments showing organization where the oldest channels can be found in the innermost region of the plaque [36,38] while experiments utilizing fluorescence recovery after photobleaching (FRAP) show that connexon mobility within gap junctions is variable and dependent on the C-terminus [39,40]. ...
... Live cell imaging of Cx43-fluorescent protein fusions (Cx43-FP) show that gap junction plaques are highly dynamic exhibiting growth via lateral accretion of connexons into the plaque and fusion of small plaques [36][37][38]. The organization of channels within the plaque can also be quite dynamic but appear to be regulated; there are clear experiments showing organization where the oldest channels can be found in the innermost region of the plaque [36,38] while experiments utilizing fluorescence recovery after photobleaching (FRAP) show that connexon mobility within gap junctions is variable and dependent on the C-terminus [39,40]. Connexon organization within the plaque appears to be regulated through interactions with kinases as treatment with glycyrrhetinic acid altered channel spacing and organization in a manner involving Src kinase [41,42] and PKC [43]. ...
Article
Full-text available
The gap junction protein Connexin43 (Cx43) is highly regulated by phosphorylation at over a dozen sites by probably at least as many kinases. This Cx43 “kinome” plays an important role in gap junction assembly and turnover. We sought to gain a better understanding of the interrelationship of these phosphorylation events particularly related to src activation and Cx43 turnover. Using state-of-the-art live imaging methods, specific inhibitors and many phosphorylation-status specific antibodies, we found phospho-specific domains in gap junction plaques and show evidence that multiple pathways of disassembly exist and can be regulated at the cellular and subcellular level. We found Src activation promotes formation of connexisomes (internalized gap junctions) in a process involving ERK-mediated phosphorylation of S279/282. Proteasome inhibition dramatically and rapidly restored gap junctions in the presence of Src and led to dramatic changes in the Cx43 phospho-profile including to increased Y247, Y265, S279/282, S365, and S373 phosphorylation. Lysosomal inhibition, on the other hand, nearly eliminated phosphorylation on Y247 and Y265 and reduced S368 and S373 while increasing S279/282 phosphorylation levels. We present a model of gap junction disassembly where multiple modes of disassembly are regulated by phosphorylation and can have differential effects on cellular signaling.
... CIP75, which belongs to the UbL (ubiquitin-like)-UBA (ubiquitinassociated) domain-containing protein family, was found to interact with the Cterminal domain of Cx43 and to promote its proteosomal degradation286 . From the Golgi apparatus, Cx43 is packed into vesicles and delivered to the plasma membranes along microtubules287 . Two models regarding the targeting of Cx43 to the plasma membrane have been proposed. ...
... Two models regarding the targeting of Cx43 to the plasma membrane have been proposed. In the classic one, Cx43 channels are transported to the membrane where they freely diffuse laterally287 . An opposing view hypothesised that assembled Cx43 hemichannels are directly targeted to the area of adherent junctions through the interaction of Cx43 with the EB1 protein and the Dynactin complex288 . ...
Thesis
The Atypical Chemokine Receptor 3 (ACKR3) and CXCR4 are two G protein-coupled receptors (GPCR) belonging to the CXC chemokine receptor family. Both receptors are activated upon CXCL12 binding and are over-expressed in various tumours, including glioma, where they have been found to promote proliferation and invasive behaviours. Upon CXCL12 binding, CXCR4 activates canonical GPCR signalling pathways involving Gαi protein and β-arrestins. In addition, CXCR4 was found to interact with several proteins able to modify its signalling, trafficking and localization. In contrast, the cellular pathways underlying ACKR3-dependent effects remain poorly characterized. Several reports show that ACKR3 engages β-arrestin-dependent signalling pathways, but its coupling to G proteins is restricted to either specific cellular populations, including astrocytes, or occurs indirectly via its interaction with CXCR4. ACKR3 also associates with the epidermal growth factor receptor to promote proliferation of tumour cells in an agonist-independent manner. These examples suggest that the extensive characterization of ACKR3 and CXCR4 interactomes might be a key step in understanding or clarifying their roles in physiological and pathological contexts. This thesis addressed this issue employing an affinity purification coupled to high-resolution mass spectrometry proteomic strategy that identified 19 and 151 potential protein partners of CXCR4 and ACKR3 transiently expressed in HEK-293T cells, respectively. Amongst ACKR3 interacting proteins identified, we paid particular attention on the gap junction protein Connexin-43 (Cx43), in line with its overlapping roles with the receptor in the control of leukocyte entry into the brain, interneuron migration and glioma progression. Western blotting and BRET confirmed the specific association of Cx43 with ACKR3 compared to CXCR4. Likewise, Cx43 is co-localized with ACKR3 but not CXCR4 in glioma initiating cell lines, and ACKR3 and Cx43 are co-expressed in astrocytes of the sub-ventricular zone and surrounding blood vessels in adult mouse brain, suggesting that both proteins form a complex in authentic cell or tissue contexts. Further functional studies showed that ACKR3 influences Cx43 trafficking and functionality at multiple levels. Transient expression of ACKR3 in HEK-293T cells to mimic ACKR3 overexpression detected in several cancer types, induces Gap Junctional Intercellular Communication (GJIC) inhibition in an agonist-independent manner. In addition, agonist stimulation of endogenously expressed ACKR3 in primary cultured astrocytes inhibits Cx43-mediated GJIC through a mechanism that requires activation of Gαi protein, and dynamin- and β-arrestin2-dependent Cx43 internalisation. Therefore, this thesis work provides the first functional link between the CXCL11/CXCL12/ACKR3 axis and gap junctions that might underlie their critical role in glioma progression.
... The present study showed that the actin cytoskeleton and RhoA signaling pathway dynamically modulates the traffic of HCs and GJCs plaque formation for Cx43 and Cx26. The role of RhoA and actin fibers in the traffic [7][8][9][32][33][34], and function of connexins [21,[35][36][37], have been centered on GJCs. Few studies focus on HCs [26,38,41]. ...
Preprint
Full-text available
Connexins (Cxs) are transmembrane proteins that assemble into gap junction channels (GJCs) and hemichannels (HCs). Previous research supports the involvement of Rho GTPases and actin microfilaments in the trafficking of Cxs, formation of GJCs plaques, and regulation of channel activity. Nonetheless, it remains uncertain whether distinct types of Cxs HCs and GJCs respond differently to Rho GTPases or changes in actin polymerization/depolymerization dynamics. Our investigation revealed that inhibiting RhoA, a small GTPase that controls actin polymerization, or disrupting actin microfilaments with cytochalasin B (Cyto-B), resulted in reduced GJCs plaque size at appositional membranes and increased transport of HCs to non-appositional plasma membrane regions. Notably, these effects were consistent across different Cx types, since Cx26 and Cx43 exhibited similar responses despite having distinct trafficking routes to the plasma membrane. Functional assessments showed that RhoA inhibition and actin depolymerization decreased the activity of Cx43 GJCs while significantly increasing HCs activity. However, the functional status of GJCs and HCs composed of Cx26 remained unaffected. This study supports the hypothesis that RhoA, through its control of the actin cytoskeleton, facilitates the transport of HCs to appositional cell membranes for GJCs formation while simultaneously limiting the positioning of free HCs at non-appositional cell membranes, independently of Cx type. This dynamic regulation promotes intercellular communication and reduces non-selective plasma membrane permeability through a Cx-type dependent mechanism, whereby Cx43 HCs and GJCs are affected, but Cx26 channels are not.
... The connexin protein pool that constitutes the gap junctions of a cell is constantly renewed, as newly formed intercellular channels are continually added to the outer edges of gap junctions, whereas older channels are cleared from the center of the gap junction plaque through endocytosis [6][7][8][9][10][11]. In line with these findings, increasing experimental evidence suggests that intercellular communication via gap junctions can be regulated through modulation of the endocytosis and degradation rates of connexins [12][13][14][15][16][17][18][19]. ...
Article
Full-text available
Intercellular communication via gap junctions has a fundamental role in regulating cell growth and tissue homeostasis, and its dysregulation may be involved in cancer development and radio- and chemotherapy resistance. Connexin43 (Cx43) is the most ubiquitously expressed gap junction channel protein in human tissues. Emerging evidence indicates that dysregulation of the sorting of Cx43 to lysosomes is important in mediating the loss of Cx43-based gap junctions in cancer cells. However, the molecular basis underlying this process is currently poorly understood. Here, we identified the E3 ubiquitin ligase ITCH as a novel regulator of intercellular communication via gap junctions. We demonstrate that ITCH promotes loss of gap junctions in cervical cancer cells, which is associated with increased degradation of Cx43 in lysosomes. The data further indicate that ITCH interacts with and regulates Cx43 ubiquitination and that the ITCH-induced loss of Cx43-based gap junctions requires its catalytic HECT (homologous to E6-AP C-terminus) domain. The data also suggest that the ability of ITCH to efficiently promote loss of Cx43-based gap junctions and degradation of Cx43 depends on a functional PY (PPXY) motif in the C-terminal tail of Cx43. Together, these data provide new insights into the molecular basis underlying the degradation of Cx43 and have implications for the understanding of how intercellular communication via gap junctions is lost during cancer development.
... In addition, previous studies have found that at the embryonic stage, desmosome forms new intercellular junctions before the gap junction protein Cx43. 12,50 The disruption of desmosome directly leads to rearrangement of Cx43 distribution, resulting in Cx43 lateralization. It further suggests a close relationship between desmosome and Cx43 distribution. ...
Article
Full-text available
Background: While desmosomal junctions and gap junction remodeling are among the arrhythmogenic substrates, the fate of desmosomal and gap junctions in high-pacing-induced heart failure remains unclear. This aim of this study was to determine the fate of desmosomal junctions in high-pacing-induced heart failure. Methods: Dogs were randomly divided into 2 equal groups, a high-pacing-induced heart failure model group (heart failure group, n = 6) and a sham operation group (control group, n = 6). Echocardiography and cardiac electrophysiological examination were performed. Cardiac tissue was analyzed by immunofluorescence and transmission electron microscopy. The expression of desmoplakin and desmoglein-2 proteins was detected by western blot. Results: A significant decrease in ejection fraction, significant cardiac dilatation, diastolic and systolic dysfunction, and ventricular thinning occurred after 4 weeks in high-pacing-induced dog model of heart failure. Effective refractory period action potential duration at 90% repolarization was prolonged in the heart failure group. Immunofluorescence analysis and transmission electron microscopy demonstrated connexin-43 lateralization accompanies desmoglein-2 and desmoplakin remodeling in the heart failure group. Western blotting showed that the expression of desmoplakin and desmoglein-2 proteins was higher in heart failure than in normal tissue. Conclusion: Desmosome (desmoglein-2 and desmoplakin) redistribution and desmosome (desmoglein-2) overexpression accompanying connexin-43 lateralization were parts of a complex remodeling in high-pacing-induced heart failure.
... Intracellular trafficking targets connexin proteins from the trans-Golgi network to the cell membrane and guides their internalization for degradation, recycling, and signal transduction (7,8). These events are thought to be regulated by the posttranslational modifications of the Cx protein C-terminal tail (9,10,11,12), and Cx endocytosis is often modulated by microtubule (MT)binding proteins on MT-filament tracks (13,14). Connexins, and GJ channels that they comprise, regulate growth and differentiation of many cell types (15,16,17). ...
Article
Full-text available
Connexin37-mediated regulation of cell cycle modulators and, consequently, growth arrest lack mechanistic understanding. We previously showed that arterial shear stress up-regulates Cx37 in endothelial cells and activates a Notch/Cx37/p27 signaling axis to promote G1 cell cycle arrest, and this is required to enable arterial gene expression. However, how induced expression of a gap junction protein, Cx37, up-regulates cyclin-dependent kinase inhibitor p27 to enable endothelial growth suppression and arterial specification is unclear. Herein, we fill this knowledge gap by expressing wild-type and regulatory domain mutants of Cx37 in cultured endothelial cells expressing the Fucci cell cycle reporter. We determined that both the channel-forming and cytoplasmic tail domains of Cx37 are required for p27 up-regulation and late G1 arrest. Mechanistically, the cytoplasmic tail domain of Cx37 interacts with, and sequesters, activated ERK in the cytoplasm. This then stabilizes pERK nuclear target Foxo3a, which up-regulates p27 transcription. Consistent with previous studies, we found this Cx37/pERK/Foxo3a/p27 signaling axis functions downstream of arterial shear stress to promote endothelial late G1 state and enable up-regulation of arterial genes.
... Connexin43 (Cx43), the most widely studied connexin isoform, is extensively regulated by protein partner interactions and multiple phosphorylation events in the carboxyl terminal domain (CT) [2]. For example, microtubules and the actin cytoskeleton are necessary for Cx43 trafficking from the trans-Golgi network to the plasma membrane [3][4][5][6][7]. ZO-1 (binds to actin) regulates the size of the gap junction plaques by controlling the rate of channel accretion at the plaque perimeters [8]. ...
Article
Full-text available
T and B cell receptor signaling involves the activation of Akt, MAPKs, and PKC as well as an increase in intracellular Ca2+ and calmodulin activation. While these coordinate the rapid turnover of gap junctions, also implicated in this process is Src, which is not activated as part of T and B cell receptor signaling. An in vitro kinase screen identified that Bruton’s tyrosine kinase (BTK) and interleukin-2-inducible T-cell kinase (ITK) phosphorylate Cx43. Mass spectroscopy revealed that BTK and ITK phosphorylate Cx43 residues Y247, Y265, and Y313, which are identical to the residues phosphorylated by Src. Overexpression of BTK or ITK in the HEK-293T cells led to increased Cx43 tyrosine phosphorylation as well as decreased gap junction intercellular communication (GJIC) and Cx43 membrane localization. In the lymphocytes, activation of the B cell receptor (Daudi cells) or T cell receptor (Jurkat cells) increased the BTK and ITK activity, respectively. While this led to increased tyrosine phosphorylation of Cx43 and decreased GJIC, the cellular localization of Cx43 changed little. We have previously identified that Pyk2 and Tyk2 also phosphorylate Cx43 at residues Y247, Y265, and Y313 with a similar cellular fate to that of Src. With phosphorylation critical to Cx43 assembly and turnover, and kinase expression varying between different cell types, there would be a need for different kinases to achieve the same regulation of Cx43. The work presented herein suggests that in the immune system, ITK and BTK have the capacity for the tyrosine phosphorylation of Cx43 to alter the gap junction function in a similar manner as Pyk2, Tyk2, and Src.
... This is associated with a continuous Cx43 stream through the endoplasmic reticulum/Golgi network, where Cx43 oligomerizes (6), followed by its arrival in the plasma membrane assembled as hexameric HCs. From there, HCs migrate laterally in the sarcolemmal lipid bilayer toward a zone called the perinexus where they coalesce with the edges of existing GJs, known as the nexus zone (4,7). HCs subsequently dock head to head with their counterparts on neighboring cells, forming GJ channels that combine into channel arrays organized as junctional plaques. ...
Article
Full-text available
Connexins are crucial cardiac proteins that form hemichannels and gap junctions. Gap junctions are responsible for the propagation of electrical and chemical signals between myocardial cells and cells of the specialized conduction system in order to synchronize the cardiac cycle and steer cardiac pump function. Gap junctions are normally open, while hemichannels are closed, but pathological circumstances may close gap junctions and open hemichannels, thereby perturbing cardiac function and homeostasis. Current evidence demonstrates an emerging role of hemichannels in myocardial ischemia and arrhythmia, and tools are now available to selectively inhibit hemichannels without inhibiting gap junctions as well as to stimulate hemichannel incorporation into gap junctions. We review available experimental evidence for hemichannel contributions to cellular pro-arrhythmic events in ventricular and atrial cardiomyocytes, and link these to insights at the level of molecular control of connexin-43-based hemichannel opening. We conclude that a double-edged approach of both preventing hemichannel opening and preserving gap junctional function will be key for further research and development of new connexin-based experimental approaches for treating heart disease.
... These data emphasize that the formation of gap junctions requires strong intercellular adhesion. The forward trafficking of Cx43 (FIGURE 6) occurs along microtubules (152,153), and at least in cell culture, the tethering of microtubules to the membrane occurs by a complex involving N-cadherin, the microtubule plus end tracking protein EB1, and p150 (Glued) (77). The link to N-cadherin may explain why Cx43 predominantly traffics to the ID, as knockout of Ncadherin disrupts not only the ID but also reduces membrane expression of Cx43 (24,25). ...
Article
Full-text available
The intercalated disc (ID) is a highly specialized structure, connecting cardiomyocytes via mechanical and electrical junctions. Although described in some detail by light microscopy in the 19th century, it was in 1966 that electron microscopy images showed that the ID represented apposing cell borders, and provided detailed insight into the complex ID nanostructure. Since then, much has been learned about the ID and its molecular composition, and it has become evident that a large number of proteins, not all of them involved in direct cell-to-cell coupling via mechanical or gap junctions, reside at the ID. Furthermore, an increasing number of functional interactions between ID components are emerging, leading to the concept that the ID is not the sum of isolated molecular silos, but an interacting molecular complex, an "organelle" where components work in concert to bring about electrical and mechanical synchrony. The aim of the present review is to give a short historical account of the ID's discovery, an updated overview of its composition and organization, followed by a discussion of the physiological implications of the ID architecture and the local intermolecular interactions. The latter will focus on both the importance for normal conduction of cardiac action potentials as well as the impact on the pathophysiology of arrhythmias.
... Cx43 hemichannels then dock in the perinexus and translocate into the gap junction plaque (nexus) proper. Older channels are internalized and degraded from the central region of the gap junction plaque (26, 73,74). Trafficking of Cx43 within the plasma membrane is also regulated by C-terminal tail ...
Article
Full-text available
Intercellular communication mediated by gap junction channels and hemichannels composed of Connexin 43 (Cx43) is vital for the propagation of electrical impulses through cardiomyocytes. The carboxyl terminal tail of Cx43 undergoes various post-translational modifications including phosphorylation of its Serine-368 (S368) residue. Protein Kinase C isozymes directly phosphorylate S368 to alter Cx43 function and stability through inducing conformational changes affecting channel permeability or promoting internalization and degradation to reduce intercellular communication between cardiomyocytes. Recent studies have implicated this PKC/Cx43-pS368 circuit in several cardiac-associated diseases. In this review, we describe the molecular and cellular basis of PKC-mediated Cx43 phosphorylation and discuss the implications of Cx43 S368 phosphorylation in the context of various cardiac diseases, such as cardiomyopathy, as well as the therapeutic potential of targeting this pathway.
... 03 of 1 µm of confocal stack images using NIS elements software (Nikon Instruments). Plasma membrane was not labeled directly, instead we estimated its localization between cellular appositions (cell-cell contacts) and cell surface, observed using the bright field light from confocal microscope images, similar criteria has been used before (Lauf et al., 2002). ...
Article
Full-text available
Some mutations in gap junction protein Connexin 26 (Cx26) lead to syndromic deafness, where hearing impairment is associated with skin disease, like in Keratitis Ichthyosis Deafness (KID) syndrome. This condition has been linked to hyperactivity of connexin hemichannels but this has never been demonstrated in cochlear tissue. Moreover, some KID mutants, like Cx26S17F, form hyperactive HCs only when co-expressed with other wild-type connexins. In this work, we evaluated the functional consequences of expressing a KID syndromic mutation, Cx26S17F, in the transgenic mouse cochlea and whether co-expression of Cx26S17F and Cx30 leads to the formation of hyperactive HCs. Indeed, we found that cochlear explants from a constitutive knock-in Cx26S17F mouse or conditional in vitro cochlear expression of Cx26S17F produces hyperactive HCs in supporting cells of the organ of Corti. These conditions also produce loss of hair cells stereocilia. In supporting cells, we found high co-localization between Cx26S17F and Cx30. The functional properties of HCs formed in cells co-expressing Cx26S17F and Cx30 were also studied in oocytes and HeLa cells. Under the recording conditions used in this study Cx26S17F did not form functional HCs and GJCs, but cells co-expressing Cx26S17F and Cx30 present hyperactive HCs insensitive to HCs blockers, Ca2+ and La3+, resulting in more Ca2+ influx and cellular damage. Molecular dynamic analysis of putative heteromeric HC formed by Cx26S17F and Cx30 presents alterations in extracellular Ca2+ binding sites. These results support that in KID syndrome, hyperactive HCs are formed by the interaction between Cx26S17F and Cx30 in supporting cells probably causing damage to hair cells associated to deafness.
... Instead, experimental results have found that fiber cell elongation occurred in the chick in the presence of inhibitors of microtubule assembly(354). In the mouse lens, elongation was poorly correlated with volume regulation(355) suggesting an indirect mechanism, possibly involving microtubule mediated aquaporin or connexin functions in the regulation of hydration and osmotic stress(356)(357)(358)(359), or involvement of the cytoskeleton (76,124) in FGF signaling(360). The alignment of the cytoskeleton to the plasma membranes and integral membrane proteins provides stability for elongating lens fiber cells(73,124,326,327,(343)(344)(345)(346)(347)(348)(349)353,361).In addition to this structural function, one of the lens intermediate filament proteins, the Beaded Filament Structural Protein 1 (BFSP1, aka filensin) regulates the activity of the major lens fiber cell water channel, aquaporin 0 (AQP0;(362)(363)(364)). ...
Article
Full-text available
In the human eye, a transparent cornea and lens combine to form the “refracton” to focus images on the retina. This requires the refracton to have a high refractive index “n”, mediated largely by extracellular collagen fibrils in the corneal stroma and the highly concentrated crystallin proteins in the cytoplasm of the lens fiber cells. Transparency is a result of short-range order (SRO) in the spatial arrangement of corneal collagen fibrils and lens crystallins, generated in part by post-translational modifications (PTMs). However, while corneal collagen is remodeled continuously and replaced, lens crystallins are very long-lived and are not replaced, and so accumulate PTMs over a lifetime. Eventually, a tipping point is reached when protein aggregation results in increased light scatter, inevitably leading to the iconic protein condensation-based disease, age-related cataract (ARC). Cataracts account for 50% of vision impairment world-wide, affecting far more people than other well-known protein aggregation-based diseases. However, because accumulation of crystallin PTMs begins before birth and long before ARC presents, we postulate that the lens protein PTMs contribute to a “cataractogenic load” (CL) that increases with age but also has protective effects on optical function by stabilizing lens crystallins until a tipping point is reached. In this review, we highlight decades of experimental findings that support the potential for PTMs to be protective during normal development. We hypothesize that ARC is preventable by protecting the biochemical and biophysical properties of lens proteins needed to maintain transparency, refraction, and optical function.
... The exposed β tubulin at the plus end binds to and hydrolyses GTP to GDP, thereby allowing dynamic treadmilling of the microtubule, while the exposed α tubulin does not hydrolyse its attached GTP (Grego et al., 2001;WatermanStorer & Salmon, 1997). It is important to note that though microtubules do not extend to the plasma membrane (Conacci-Sorrell et al., 2002;Dejana, 2004;Dudek & Garcia, 2001;Lampugnani et al., 2002) and also do not directly interact with cellular junctions (Vincent et al., 2004), they are capable of delivering cell junction components to the cell surface through the vesicular trafficking of p120 catenin, a component of AJ as well as the delivery of connexin hemi-channels for the gap junction channels (Chen et al., 2003;Lauf et al., 2002;Shaw et al., 2007;Yanagisawa et al., 2004). Microtubules are always in a dynamic state of assembly and disassembly and initially, microtubules and actin filaments were regarded as individual entities. ...
Article
During metastasis, tumour cells must become migratory and travel towards a capillary within the tumour. They then degrade the matrix surrounding the pericytes and endothelial cells, insert themselves between endothelial cells, transverse the capillary wall, to then enter the blood stream. This process depends on the motile behaviour of the tumour cells as well as the role of endothelial cell-cell junctions, both including adherens junctions and tight junctions. Circulating tumour cells must next, adhere to the walls of the capillary at the site of secondary tumour formation. Here, they again traverse the capillary wall to enter tissues distant from the primary tumour. This review aim to discuss the basic architecture of the endothelial junctional complex as well as the role played by these components towards the transendothelial migration of cancer cells from the primary site to the secondary site. Proper understanding of the role played by each of these components could invariably lead to the development of novel adjuvant cancer chemotherapy.
... Further, in cell culture, αCT1 has been shown to inhibit binding with its protein partner ZO1 [163]. Under physiological conditions, the PDZ2 domain of ZO1 has been shown to interact with the Cx43 CT and studies have demonstrated that ZO1 regulates the connexin to GJ transition, by slowing the rate at which hemichannels move into established GJ plaques [163][164][165]. Further, αCT1 treatment has been shown to increase GJ plaque size and GJ coupling, and decrease hemichannel activity [163]. ...
Article
Full-text available
Gap junctions (GJ) and connexins play integral roles in cellular physiology and have been found to be involved in multiple pathophysiological states from cancer to cardiovascular disease. Studies over the last 60 years have demonstrated the utility of altering GJ signaling pathways in experimental models, which has led to them being attractive targets for therapeutic intervention. A number of different mechanisms have been proposed to regulate GJ signaling, including channel blocking, enhancing channel open state, and disrupting protein-protein interactions. The primary mechanism for this has been through the design of numerous peptides as therapeutics, that are either currently in early development or are in various stages of clinical trials. Despite over 25 years of research into connexin targeting peptides, the overall mechanisms of action are still poorly understood. In this overview, we discuss published connexin targeting peptides, their reported mechanisms of action, and the potential for these molecules in the treatment of disease.
... HDAC6 activation initiates deacetylation of αtubulin, leading to derailment of α-tubulin proteostasis and disruption of the cardiomyocyte microtubule structure. 11) Lauf, et al. 31) demonstrated that microtubules have an important role in the trafficking of CX43 to the gap junction. We demonstrated that the protein expression levels of acetylated α-tubulin decreased significantly in HDAC6 active TG compared with NTG mouse atria (Figure 1A and B). ...
Article
Atrial fibrillation (AF) is a relatively common complication of hypertension. Chronic hypertension induces cardiac HDAC6 catalytic activity. However, whether HDAC6 activation contributes to hypertension-induced AF is still uncertain. We examined whether chronic cardiac HDAC6 activation-induced atrial remodeling, leading to AF induction. The HDAC6 constitutively active transgenic (TG) (HDAC6 active TG) mouse overexpressing the active HDAC6 protein, specifically in cardiomyocytes, was created to examine the effects of chronic HDAC6 activation on atrial electrical and structural remodeling and AF induction in HDAC6 active TG and non-transgenic (NTG) mice. Left atrial burst pacing (S1S1 = 30 msec) for 15-30 sec significantly increased the frequency of sustained AF in HDAC6 active-TG mice compared with NTG mice. Left steady-state atrial pacing (S1S1 = 80 msec) decreased the atrial conduction velocity in isolated HDAC6 active TG compared with NTG mouse atria. The atrial size was similar between HDAC6 active TG and NTG mice. In contrast, atrial interstitial fibrosis increased in HDAC6 active TG compared with that of NTG mouse atria. While protein expression levels of both CX40 and CX43 were similar between HDAC6 active TG and NTG mouse atria, a heterogeneous distribution of CX40 and CX43 occurred in HDAC6 active-TG mouse atria but not in NTG mouse atria. Gene expression of interleukin 6 increased in HDAC6 active TG compared with NTG mouse atria. Chronic cardiac HDAC6 activation induced atrial electrical and structural remodeling, and sustained AF. Hypertension-induced cardiac HDAC6 catalytic activity may play important roles in the development of AF.
... Connexins are synthesized in the endoplasmic reticulum and trafficking to the plasma membrane is by the secretory pathway involving the Golgi apparatus and the trans-Golgi network, although Golgi independent trafficking has also been reported for Cx26 [7][8][9]. Most connexins have short half-lives (1-5 h) when compared to typical integral membrane proteins [10][11][12][13][14][15][16]. Although the reason for this is unknown, it has been proposed that a highly regulated short half-life allows for more precise control of expression. ...
Article
Connexins are membrane proteins involved directly in cell-to-cell communication through the formation of gap-junctional channels. These channels result from the head-to-head docking of two hemichannels, one from each of two adjacent cells. Undocked hemichannels are also present at the plasma membrane where they mediate the efflux of molecules that participate in autocrine and paracrine signaling, but abnormal increase in hemichannel activity can lead to cell damage in disorders such as cardiac infarct, stroke, deafness, cataracts, and skin diseases. For this reason, connexin hemichannels have emerged as a valid therapeutic target. Know small molecule hemichannel inhibitors are not ideal leads for the development of better drugs for clinical use because they are not specific and/or have toxic effects. Newer inhibitors are more selective and include connexin mimetic peptides, anti-connexin antibodies and drugs that reduce connexin expression such as antisense oligonucleotides. Re-purposed drugs and their derivatives are also promising because of the significant experience with their clinical use. Among these, aminoglycoside antibiotics have been identified as inhibitors of connexin hemichannels that do not inhibit gap-junctional channels. In this review, we discuss connexin hemichannels and their inhibitors, with a focus on aminoglycoside antibiotics and derivatives of kanamycin A that inhibit connexin hemichannels, but do not have antibiotic effect.
... The life cycle of Cx43 involves posttranslational insertion of Cx43 monomers into the ER membrane followed by their oligomerization into hexameric hemichannels or connexons in the Golgi. Preassembled hemichannels are translocated to the cell surface, which then dock with connexons from an apposing cell and undergo channel clustering to form morphologically identifiable gap junction plaques (41). Cx43 is unique in that it does not oligomerize in the ER unlike most multimeric membrane proteins. ...
Article
Full-text available
Altered expression and function of astroglial gap junction protein Connexin 43 (Cx43) has increasingly been associated to neurotoxicity in Alzheimer disease (AD). While earlier studies have examined the effect of increased amyloid-β (Aβ) on Cx43 expression and function leading to neuronal damage, underlying mechanisms by which Aβ modulates Cx43 in astrocytes remain elusive. Here, using mouse primary astrocyte cultures, we have examined the cellular processes by which Aβ can alter Cx43 gap junctions. We show that Aβ 25-35 impairs functional gap junction coupling yet increases hemichannel activity. Interestingly, Aβ 25-35 increased the intracellular pool of Cx43 with a parallel decrease in gap junction assembly at the surface. Intracellular Cx43 was found to be partly retained in the endoplasmic reticulum-associated cell compartments. However, forward trafficking of the newly synthesized Cx43 that already reached the Golgi was not affected in Aβ 25-35 exposed astrocytes. Supporting this, treatment with 4-phenylbutyrate, a well-known chemical chaperone that improves trafficking of several transmembrane proteins, restored Aβ-induced impaired gap junction coupling between astrocytes. We further show that interruption of Cx43 endocytosis in Aβ 25-35 exposed astrocytes resulted in their retention at the cell surface in the form of functional gap junctions indicating that Aβ 25-35 causes rapid internalization of Cx43 gap junctions. Additionally, in silico molecular docking suggests that Aβ can bind favorably to Cx43. Our study thus provides novel insights into the cellular mechanisms by which Aβ modulates Cx43 function in astrocytes, the basic understanding of which is vital for the development of alternative therapeutic strategy targeting connexin channels in AD.
... Gap junctions are usually much larger than lipid rafts [2], and the internalization of entire plaques by this alternative pathway is unlikely. Instead, under normal physiological conditions, connexins destined for the degradation are removed from the center of the plaque [44,45]. Endocytosis of Cx36 by a caveolin-mediated pathway might be one of the different pathways used by cells for dynamic control of gap junction mediated communication. ...
Article
Full-text available
The gap junctional protein connexin 36 (Cx36) has been co-purified with the lipid raft protein caveolin-1 (Cav-1). The relevance of an interaction between the two proteins is unknown. In this study, we explored the significance of Cav-1 interaction in the context of intracellular and membrane transport of Cx36. Coimmunoprecipitation assays and Förster resonance energy transfer analysis (FRET) were used to confirm the interaction between the two proteins in the Neuro 2a cell line. We found that the Cx36 and Cav-1 interaction was dependent on the intracellular calcium levels. By employing different microscopy techniques, we demonstrated that Cav-1 enhances the vesicular transport of Cx36. Pharmacological interventions coupled with cell surface biotinylation assays and FRET analysis revealed that Cav-1 regulates membrane localization of Cx36. Our data indicate that the interaction between Cx36 and Cav-1 plays a role in the internalization of Cx36 by a caveolin-dependent pathway.
... Many Connexins (protein subunits of vertebrate gap junctions) are dependent on actin and microtubule networks for localization (George et al., 1999;Martin et al., 2001;Lauf et al., 2002), while Pannexins (chordate homologues of Innexins) are dependent on actin but not 1 2 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 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93 94 95 96 97 98 99 100 101 102 103 104 105 106 107 108 microtubules (Bhalla-Gehi et al., 2010). However, there are no previous reports indicating whether insect Innexins require the actin and tubulin cytoskeleton for proper localization. ...
Article
Full-text available
Polydnaviruses associated with ichneumonid parasitoid wasps (Ichnoviruses) encode large numbers of genes, often in multigene families. The Ichnovirus Vinnexin gene family, which is expressed in parasitized lepidopteran larvae, encodes homologues of Innexins, the structural components of insect gap junctions. Here, we have examined intracellular behaviors of the Campoletis sonorensis Ichnovirus (CsIV) Vinnexins, alone and in combination with a host Innexin orthologue, Innexin2 (Inx2). QRT‐PCR verified that transcription of CsIV vinnexins occurs contemporaneously with inx2 , implying co‐occurrence of Vinnexin and Inx2 proteins. Confocal microscopy demonstrated that epitope‐tagged VinnexinG (VnxG) and VinnexinQ2 (VnxQ2) exhibit similar subcellular localization as Spodoptera frugiperda Inx2 (Sf‐ Inx2). Surface biotinylation assays verified that all three proteins localize to the cell surface, and cytochalasin B and nocodazole that they rely on actin and microtubule cytoskeletal networks for localization. Immunomicroscopy following co‐transfection of constructs indicates extensive co‐localization of Vinnexins with each other and Sf‐ Inx2, and live‐cell imaging of mCherry‐labelled Inx2 supports that Vinnexins may affect Sf ‐Inx2 distribution in a Vinnexin‐specific fashion. Our findings support that the Vinnexins may disrupt host cell physiology in a protein‐specific manner through altering gap junctional intercellular channel communication, as well as indirectly by affecting multicellular junction characteristics. This article is protected by copyright. All rights reserved.
... Les GJ sont formées de l'apposition de deux membranes plasmiques, séparées par une mince fente de 2 à 3 nm de large, avec présence de structures pentalaminaires caractéristiques des GJ ( Figure 28). Par des études en microscopie électronique et de cryofracture, le coeur opaque observé s'est révélé être un pore : des centaines à plusieurs milliers de canaux intercellulaires forme ainsi des plaques « jonctionnelles » où la concentration de GJ peut aller jusqu'à 10 4 canaux/µm 2 de membrane (Lauf et al., 2002). Figure 28 : Image en microscopie électronique de jonction communicante D'après Revel and Karnovsky, 1967 tel-00935261, version 1 -23 Jan 2014 ...
... La démonstration de l'existence d'une communication astrocytaire a été rendue possible grâce à l'avènement des techniques de microscopie. En particulier, l'utilisation de la microscopie électronique et de la cryofracture a notamment permis la visualisation des JC et des HC et de dissocier leurs rôles respectifs(Lauf et al 2002). ...
Thesis
Les cellules gliales dont les astrocytes - au moins aussi nombreux que les neurones dans le cerveau - joueraient un rôle important dans l'anxiété, la dépression et probablement dans la réponse aux antidépresseurs. Plusieurs études menées chez l'Homme et l'animal vont dans ce sens puisqu'elles mettent en évidence une association entre des changements d'expression de différents marqueurs astrocytaires et la sévérité de ces troubles psychiatriques. C'est notamment le cas de la connexine 43 (Cx43), une protéine transmembranaire impliquée dans la formation de deux unités fonctionnelles distinctes : les jonctions communicantes (JCs) qui assurent la communication entre deux astrocytes voisins et les hémicanaux (HCs) dont le rôle est de libérer de molécules neuro-actives (gliotransmetteurs i.e. glutamate, ATP, D-sérine) dans la fente synaptique. En effet, une diminution de l'expression des Cx43 a été rapportée dans différentes régions cérébrales de patients dépressifs et dans des modèles murins de dépression. En revanche, d'un point de vue fonctionnel, l'induction d'un phénotype "anxio-dépressif" serait associée à une diminution de l'activité des JCs et à une augmentation de l'activité des HCs. Face à ces effets opposés, l'objectif de cette thèse était de caractériser plus finement le rôle des Cx43 dans les comportements "anxio-dépressifs" et la réponse aux psychotropes en utilisant des approches d'inactivation génétiques et pharmacologiques de ces protéines. Nos résultats montrent que l'inactivation génétique des Cx43 dans l'hippocampe n'entraine aucun effet neuro-comportemental. En revanche, dans un modèle de dépression basé sur l'exposition chronique des souris à la corticostérone (modèle CORT), l'inactivation génétique des Cx43 exerce des effets de type anxiolytiques-antidépresseurs. D'un point de vue mécanistique, ces effets seraient liés à une diminution de la libération hippocampique de glutamate par les HCs et à une atténuation de la réactivité de l'axe hypothalamo-hypophysaire (HPA). Concernant l'inactivation pharmacologique des Cx43, nos travaux apportent des évidences expérimentales sur le fait que l'administration systémique de carbenoxolone, un bloqueur des connexines, potentialise la réponse aiguë d'un inhibiteur de recapture de la sérotonine en conditions basales, mais s'oppose à ses effets bénéfiques chroniques dans un modèle de dépression. Le microenvironnement cellulaire semble donc essentiel dans la manière dont les Cx43 influencent la réponse aux antidépresseurs. L'ensemble de ces résultats de thèse laissent entrevoir un rôle des Cx43 astrocytaires dans la régulation de l'humeur via la modulation de circuits neuronaux convergeant vers l'axe HPA. Ils soulignent également l'intérêt de moduler les Cx43 pour renforcer l'activité thérapeutiques des antidépresseurs actuellement disponibles. Les futures recherches devront préciser les modalités de ces nouvelles stratégies combinant des agents pharmacologiques à tropisme astrocytaire et neuronal.
... Connexins arrive at the synapse as hemichannels that are inserted at the boundaries of existing gap junction plaques where they then find a partner hemichannel in the adjoining neuron. Over time, the channels migrate towards the center of the plaque where they are endocytosed and sent to the lysosome for degradation (Lauf et al., 2002;Flores et al., 2012;Wang et al., 2015). The half-life of Cx36 is estimated to be between 1 and 3 h in vivo, so to maintain the electrical synapse, Cx36 must continuously be made and trafficked to the correct location (Flores et al., 2012;Wang et al., 2015). ...
Article
Full-text available
In this review article, we will describe the recent advances made towards understanding the molecular and cell biological mechanisms of electrical synapse formation. New evidence indicates that electrical synapses, which are gap junctions between neurons, can have complex molecular compositions including protein asymmetries across joined cells, diverse morphological arrangements, and overlooked similarities with other junctions, all of which indicate new potential roles in neurodevelopmental disease. Aquatic organisms, and in particular the vertebrate zebrafish, have proven to be excellent models for elucidating the molecular mechanisms of electrical synapse formation. Zebrafish will serve as our main exemplar throughout this review and will be compared with other model organisms. We highlight the known cell biological processes that build neuronal gap junctions and compare these with the assemblies of adherens junctions, tight junctions, non-neuronal gap junctions, and chemical synapses to explore the unknown frontiers remaining in our understanding of the critical and ubiquitous electrical synapse.
... On the basis of these results, the authors already suggested that lipid rafts may be involved in trafficking non-junctional membrane Cx26 to non-lipid raft GJPs ( Locke et al., 2005 ). Because of the relatively short half-life of connexins (usually 1 -5 h), the GJP is in a dynamic state, constantly remodeled through both recruitment of newly synthesized hemichannels to the outer periphery of the GJP and endocytosis of older components from the center of the plaque ( Gaietta et al., 2002 ;Lauf et al., 2002 ). In agreement with the latter model, we could suggest that, owing to low affinity of Cx26 for cholesterol abundantly present in lipid rafts ( Hung and Yarovsky, 2011 ;Locke and Harris, 2009 ), Cx26/Cx30 hemichannels rapidly diffuse laterally out of lipid raft-enriched tricellular junctions and accrue along the outer periphery of the pre-existing GJP, where each hemichannel docks with another from the neighbouring cell to form a new heteromeric channel. ...
Article
In the cochlea, connexins 26 (Cx26) and 30 (Cx30) largely co-assemble into heteromeric gap junctions, which connect adjacent non-sensory epithelial cells. These channels are believed to ensure the rapid removal of K⁺ away from the base of sensory hair cells, resulting in K⁺ recycling back to the endolymph to maintain cochlear homeostasis. Many of the mutations in GJB2 and GJB6, which encode CX26 and CX30, impair the formation of membrane channels and cause autosomal hearing loss in humans. Although recent advances have been made, several important questions remain about connexin trafficking and gap junction biogenesis. Here we show that tricellular adherens junctions present at the crossroad between adjacent gap junction plaques, provide an unexpected cell surface delivery platform for Cx26/Cx30 oligomers. Using an in situ proximity ligation assay, we detected the presence of non-junctional Cx26/Cx30 oligomers within lipid raft-enriched tricellular junction sites. In addition, we observed that cadherin homophilic interactions are critically involved in microtubule-mediated trafficking of Cx26/Cx30 oligomers to the cell surface. Overall, our results unveil an unexpected role for tricellular junctions in the trafficking and assembly of membrane channels.
... Many GJIC cluster to form a tightly packed semi-crystalline arrays referred as GJ plaques. The outer edges of the GJ plaque are composed of newly synthesized connexons, while the older GJ channels are found in the center of the plaque, where they get internalized and destined for degradation [42][43][44]. The degradation of GJ plaques occurs via the formation of connexosomes (annular junction), where an entire GJ or a fragment of GJ are removed by lysosomal, phagolysosomal, autophagy, or ubiquitin dependent proteasomal pathways [33,45]. ...
Article
Full-text available
The crosstalk between hematopoietic stem cells (HSC) and bone marrow (BM) microenvironment is critical for homeostasis and hematopoietic regeneration in response to blood formation emergencies after injury, and has been associated with leukemia transformation and progression. Intercellular signals by the BM stromal cells in the form of cell-bound or secreted factors, or by physical interaction, regulate HSC localization, maintenance, and differentiation within increasingly defined BM HSC niches. Gap junctions (GJ) are comprised of arrays of membrane embedded channels formed by connexin proteins, and control crucial signaling functions, including the transfer of ions, small metabolites, and organelles to adjacent cells which affect intracellular mechanisms of signaling and autophagy. This review will discuss the role of GJ in both normal and leukemic hematopoiesis, and highlight some of the most novel approaches that may improve the efficacy of cytotoxic drugs. Connexin GJ channels exert both cell-intrinsic and cell-extrinsic effects on HSC and BM stromal cells, involved in regenerative hematopoiesis after myelosuppression, and represent an alternative system of cell communication through a combination of electrical and metabolic coupling as well as organelle transfer in the HSC niche. GJ intercellular communication (GJIC) in the HSC niche improves cellular bioenergetics, and rejuvenates damaged recipient cells. Unfortunately, they can also support leukemia proliferation and survival by creating leukemic niches that provide GJIC dependent energy sources and facilitate chemoresistance and relapse. The emergence of new strategies to disrupt self-reinforcing malignant niches and intercellular organelle exchange in leukemic niches, while at the same time conserving normal hematopoietic GJIC function, could synergize the effect of chemotherapy drugs in eradicating minimal residual disease. An improved understanding of the molecular basis of connexin regulation in normal and leukemic hematopoiesis is warranted for the re-establishment of normal hematopoiesis after chemotherapy.
... Les connexons exprimés à la membrane et non apposés à d'autres connexons forment les hémicanaux. Puis l'accrétion de nombreux connexons qui s'apposent aux connexons d'une cellule voisine forme les plaques de jonction communicantes (Lauf et al., 2002). ...
Thesis
La Neuromyélite Optique (NMO) est une maladie auto-immune démyélinisante, rare et grave, du système nerveux central (SNC). Elle est caractérisée par une démyélinisation et une perte axonale ciblant principalement le nerf optique et la moelle épinière. La découverte d'un auto-anticorps (IgG-NMO) dirigé contre l'aquaporine-4 (AQP4), un canal hydrique exprimé par l'astrocyte, a été une étape clé dans la compréhension de la physiopathologie de la NMO, actuellement définie comme une astrocytopathie. La pathogénicité de l'IgG-NMO a été démontrée : il induit une internalisation d'AQP4 et des transporteurs au glutamate, provoquant une altération de la fonction astrocytaire. Cependant les mécanismes permettant de lier la dysfonction astrocytaire aux altérations caractéristiques de la NMO, notamment la démyélinisation, restent méconnus. Les astrocytes sont des cellules gliales essentielles à l'établissement et au maintien de l'homéostasie du SNC. Ils permettent la régulation des flux hydriques et ioniques, le contrôle extracellulaire des neuromédiateurs ainsi que l'apport de métabolites énergétiques aux neurones et aux oligodendrocytes. Ils sont aussi caractérisés par une très forte expression de connexines (Cx), des molécules transmembranaires s'assemblant sous une forme hexamérique : le connexon. Les connexines forment soit des hémicanaux, permettant l'échange de petites molécules entre les milieux intra- et extra-cellulaires, soit des jonctions communicantes par la juxtaposition de connexons appartenant à deux cellules, assurant le couplage intercellulaire avec le passage de petites molécules et d'ions (ATP, glutamate, lactate, calcium). Les fonctions hemicanal et jonction communicante sont fortement régulées en condition physiologique et altérées en condition pathologique, notamment en contexte neuroinflammatoire. Nous émettons l'hypothèse que les IgG-NMO altèrent l'expression et la fonction des connexines, et conduisent ainsi à la production d'un environnement toxique pour les oligodendrocytes et la myéline, et délétère pour le fonctionnement neuronal. Mon projet de thèse avait trois objectifs : i) la caractérisation du phénotype astrocytaire induit par les IgGNMO ; ii) l'identification d'altérations des connexines et leur implication dans la pathologie ; iii) la mise en évidence d'altérations de la transmission synaptique induites par les IgG-NMO et l'implication de connexines dans cet effet. Des modèles de cultures primaires gliales traitées par des IgG-NMO issue d'une cohorte de patients m'ont permis de caractériser le phénotype acquis par les astrocytes, et de proposer le concept d'un astrocyte réactif spécifique de pathologie. Les astrocytes réactifs spécifiques de la NMO induisent un milieu inflammatoire spécifique et toxique, provoquant une démyélinisation. Grâce au développement d'une coculture gliale et neuronale produisant des neurones myélinisés, et à l'utilisation de peptides inhibiteurs des Cx, j'ai pu montrer que les NMO-IgG ont un effet démyélinisant et que celui-ci implique les Cx. La démyélinisation est en effet associée à des modifications structurales et fonctionnelles des Cx astrocytaires, observées à la fois in vitro et dans notre modèle in vivo, le rat-NMO. Enfin, la mise en place d'une étude électrophysiologique en potentiel de champs local sur des tranches d'hippocampe de rats m'a permis d'étudier l'effet des IgG-NMO sur la transmission glutamatergique basale. J'ai pu mettre en évidence un effet dépresseur des IgG-NMO, partiellement bloqué par un inhibiteur de connexines, la carbenoxolone. Comme il a déjà été démontré par des études cliniques dans des pathologies neurodégénératives, l'utilisation de modulateurs de Cx semble être une voie thérapeutique prometteuse afin de prévenir la démyélinisation et les altérations du fonctionnement neuronal de la NMO
... In differentiated neurons, detyr-α-tub assists in the polarization of neurons by increasing specific motor binding to decorated microtubules and by guiding motor proteins to specific sites, probably for transport of particular cargoes (Janke and Bulinski, 2011). In HeLa and T51B rat liver epithelial cells, connexins are packaged in vesicular carriers traveling along microtubules from the Golgi to insertion sites all over the cell surface (Lauf et al., 2002). ...
Article
Full-text available
Human trophoblast syncytialization is one of the most important yet least understood events during placental development. In this study, we found that detyrosinated α-tubulin (detyr-α-tub), which is negatively regulated by tubulin tyrosine ligase (TTL), was elevated during human placental cytotrophoblast fusion. Correspondingly, relatively high expression of TTL protein was observed in first-trimester human placental cytotrophoblast cells, but fusing trophoblast cells exhibited much lower levels of TTL. Notably, fusion of preeclamptic cytotrophoblast cells was compromised but could be partially rescued by knockdown of TTL levels. Mechanistically, chronic downregulation of TTL in trophoblast cells resulted in significantly elevated expression of detyr-α-tub. Restoration of detyr-α-tub thus contributed to the cell surface localization of the fusogenic protein Syncytin-2 and the gap junction protein Connexin 43 (Cx43), which in turn promoted successful fusion between trophoblast cells. Taken together, the results suggest that tubulin detyrosination plays an essential role in human trophoblast fusogenic protein aggregation and syncytialization. Insufficient tubulin detyrosination leads to defects in syncytialization and potentially to the onset of preeclampsia.
... It has been shown that Cx43 trafficking to the sarcolemma of cardiomyocytes is dependent on both the microtubular network (58) and actin cytoskeleton (59). Previously, the role of a-SMA stress fibers in myofibroblasts has been investigated, and it has been shown that pharmacological ablation of these cytoskeletal elements eliminates the arrhythmogenic effects of myofibroblasts on cardiomyocytes (12,41,59). ...
Article
Full-text available
Healthy cardiomyocytes are electrically coupled at the intercalated discs by gap junctions. In infarcted hearts, adverse gap‐junctional remodeling occurs in the border zone, where cardiomyocytes are chemically and electrically influenced by myofibroblasts. The physical movement of these contacts remains unquantified. Using scanning ion conductance microscopy, we show that intercellular contacts between cardiomyocytes and myofibroblasts are highly dynamic, mainly owing to the edge dynamics (lamellipodia) of the myofibroblasts. Decreasing the amount of functional connexin‐43 (Cx43) at the membrane through Cx43 silencing, suppression of Cx43 trafficking, or hypoxia‐induced Cx43 internalization attenuates heterocellular contact dynamism. However, we found decreased dynamism and stabilized membrane contacts when cellular coupling was strengthened using 4‐phenylbutyrate (4PB). Fluorescent‐dye transfer between cells showed that the extent of functional coupling between the 2 cell types correlated with contact dynamism. Intercellular calcein transfer from myofibroblasts to cardiomyocytes is reduced after myofibroblast‐specific Cx43 down‐regulation. Conversely, 4PB‐treated myofibroblasts increased their functional coupling to cardiomyocytes. Consistent with lamellipodia‐mediated contacts, latrunculin‐B decreases dynamism, lowers physical communication between heterocellular pairs, and reduces Cx43 intensity in contact regions. Our data show that heterocellular cardiomyocyte‐myofibroblast contacts exhibit high dynamism. Therefore, Cx43 is a potential target for prevention of aberrant cardiomyocyte coupling and myofibroblast proliferation in the infarct border zone.—Schultz, F., Swiatlowska, P., Alvarez‐Laviada, A., Sanchez‐Alonso, J. L., Song, Q., de Vries, A. A. F., Pijnappels, D. A., Ongstad, E., Braga, V. M. M., Entcheva, E., Gourdie, R. G., Miragoli, M., Gorelik, J. Cardiomyocyte–myofibroblast contact dynamism is modulated by connexin‐43. FASEB J. 33, 10453–10468 (2019). www.fasebj.org
... Gap junction assembly usually occurs in a "two-step mechanism" which requires successively microtubules and actin cytoskeletal components. First, hexameric connexons assembled in the trans-Golgi network are trafficked along microtubules to the non-junctional plasma membrane (Koval et al., 1997;Lauf et al., 2002;Musil and Goodenough, 1993). Secondly, hemichannels associate with cortical actin through actin-binding proteins zonula occludens (ZOs) which regulate delivery of connexins from the periphery to the GJP (Herv e et al., 2014;Th evenin et al., 2013). ...
Article
Hereditary hearing loss affects about 1 per 1000 children. Mutations in GJB2, which encodes the connexin 26 protein (Cx26) involved in cochlear homeostasis, are found in about 50% of patients with autosomal recessive non-syndromic hearing loss. Deciphering the trafficking pathway of cochlear Cx26 in situ should represent an advance in understanding the pathogenic significance of many of these mutations. Connexins trafficking and delivery to lipid raft-associated gap junction plaques usually requires successively microtubule and actin networks. Here we show that cochlear Cx26 exhibits an unusual trafficking pathway. We observed that Cx26 assembly occurs in non-lipid raft membrane domains and that junctional plaques are devoid of actin and associated zonula occludens proteins. Using cytoskeleton-disrupting drugs in organotypic culture, we found that cochlear Cx26 gap junction assembly requires microtubules but not actin filaments. Altogether, our data provide an unexpected insight into Cx26 trafficking pathway and gap junction assembly in the cochlea.
... These annular gap junctions were first identified with transmission electron microscopy (TEM) and were distinguished from other organelles by the presence of a double-membrane (pentalaminar membrane) and a central lumen [8,[13][14][15][16][17][18]. Annular gap junctions (also called connexosomes) have now been demonstrated with live cell imaging techniques to form from internalization at central regions of the gap junction plaque [19][20][21][22] or internalization of the entire gap junction plaque [23]. ...
Article
Full-text available
It is becoming clear that in addition to gap junctions playing a role in cell–cell communication, gap junction proteins (connexins) located in cytoplasmic compartments may have other important functions. Mitochondrial connexin 43 (Cx43) is increased after ischemic preconditioning and has been suggested to play a protective role in the heart. How Cx43 traffics to the mitochondria and the interactions of mitochondria with other Cx43-containing structures are unclear. In this study, immunocytochemical, super-resolution, and transmission electron microscopy were used to detect cytoplasmic Cx43-containing structures and to demonstrate their interactions with other cytoplasmic organelles. The most prominent cytoplasmic Cx43-containing structures—annular gap junctions—were demonstrated to form intimate associations with lysosomes as well as with mitochondria. Surprisingly, the frequency of associations between mitochondria and annular gap junctions was greater than that between lysosomes and annular gap junctions. The benefits of annular gap junction/mitochondrial associations are not known. However, it is tempting to suggest, among other possibilities, that the contact between annular gap junction vesicles and mitochondria facilitates Cx43 delivery to the mitochondria. Furthermore, it points to the need for investigating annular gap junctions as more than only vesicles destined for degradation.
Article
Connexins are channel forming proteins that function to facilitate gap junctional intercellular communication. Here we use dual cell voltage clamp and dye transfer studies to corroborate past findings that Cx31.1 is defective in gap junction channel formation; illustrating that Cx31.1 alone does not form functional gap junction channels in connexin-deficient cells. Rather Cx31.1 transiently localizes to the secretory pathway with a subpopulation reaching the cell surface which is rarely seen in puncta reminiscent of gap junctions. Intracellular retained Cx31.1 was subject to degradation as Cx31.1 accumulated in the presence of proteasomal inhibition, had a faster turnover when Cx43 was present, and ultimately reached lysosomes. While intracellularly retained Cx31.1 was found to interact with Cx43, this interaction did not rescue its delivery to the cell surface. Conversely, the co-expression of Cx31 dramatically rescued the assembly of Cx31.1 into gap junctions where gap junction-mediated dye transfer was enhanced. Collectively, our results indicate that the localization and functional status of Cx31.1 is altered through selective interplay with co-expressed connexins, perhaps suggesting Cx31.1 is a key regulator of intercellular signalling in keratinocytes.
Article
Non-junctional connexin43 (Cx43) plasma membrane hemichannels have been implicated in several inflammatory diseases, particularly playing a role in ATP release that triggers activation of the inflammasome. Therapies targeting the blocking of the hemichannels to prevent the pathological release or uptake of ions and signalling molecules through its pores are of therapeutic interest. To date, there is no close-to-native, high-definition documentation of the impact of Cx43 hemichannel-mediated inflammation on cellular ultrastructure, neither is there a robust account of the ultrastructural changes that occur following treatment with selective Cx43 hemichannel blockers such as Xentry-Gap19 (XG19). A combination of same-sample correlative high-resolution three-dimensional fluorescence microscopy and soft X-ray tomography at cryogenic temperatures, enabled in the identification of novel 3D molecular interactions within the cellular milieu when comparing behaviour in healthy states and during the early onset or late stages under inflammatory conditions. Notably, our findings suggest that XG19 blockage of connexin hemichannels under pro-inflammatory conditions may be crucial in preventing the direct degradation of connexosomes by lysosomes, without affecting connexin protein translation and trafficking. We also delineated fine and gross cellular phenotypes, characteristic of inflammatory insult or road-to-recovery from inflammation, where XG19 could indirectly prevent and reverse inflammatory cytokine-induced mitochondrial swelling and cellular hypertrophy through its action on Cx43 hemichannels. Our findings suggest that XG19 might have prophylactic and therapeutic effects on the inflammatory response, in line with functional studies.
Article
Full-text available
Gap junctions are specialized regions of the plasma membrane containing clusters of channels that provide for the diffusion of ions and small molecules between adjacent cells. A fundamental role of gap junctions is to coordinate the functions of cells in tissues. Cancer pathogenesis is usually associated with loss of intercellular communication mediated by gap junctions, which may affect tumor growth and the response to radio- and chemotherapy. Gap junction channels consist of integral membrane proteins termed connexins. In addition to their canonical roles in cell-cell communication, connexins modulate a range of signal transduction pathways via interactions with proteins such as β-catenin, c-Src, and PTEN. Consequently, connexins can regulate cellular processes such as cell growth, migration, and differentiation through both channel-dependent and independent mechanisms. Gap junctions are dynamic plasma membrane entities, and by modulating the rate at which connexins undergo endocytosis and sorting to lysosomes for degradation, cells rapidly adjust the level of gap junctions in response to alterations in the intracellular or extracellular milieu. Current experimental evidence indicates that aberrant trafficking of connexins in the endocytic system is intrinsically involved in mediating the loss of gap junctions during carcinogenesis. This review highlights the role played by the endocytic system in controlling connexin degradation, and consequently gap junction levels, and discusses how dysregulation of these processes contributes to the loss of gap junctions during cancer development. We also discuss the therapeutic implications of aberrant endocytic trafficking of connexins in cancer cells.
Article
Full-text available
Gap junction channels, composed of connexins, allow direct cell-to-cell communication. Connexin 43 (Cx43) is widely expressed in tissues, including the epidermis. In a previous study of human papillomavirus-positive cervical epithelial tumour cells, we identified Cx43 as a binding partner of the human homologue of Drosophila Discs large (Dlg1). Dlg1 is a member of the membrane associated-guanylate kinase (MAGUK) scaffolding protein family that is known to control cell shape and polarity. Here we show that Cx43 also interacts with Dlg1 in uninfected keratinocytes in vitro and in keratinocytes, dermal cells and adipocytes in normal human epidermis in vivo. Depletion of Dlg1 in keratinocytes did not alter Cx43 transcription but was associated with a reduction in Cx43 protein levels. Reduced Dlg1 levels in keratinocytes resulted in a reduction in Cx43 at the plasma membrane with a concomitant reduction in gap junctional intercellular communication and relocation of Cx43 to the Golgi compartment. Our data suggest a key role for Dlg1 in maintaining Cx43 at the plasma membrane in keratinocytes.
Thesis
L’exposition aux polluants peut durablement affecter la santé humaine. Parmi ces polluants, certains agissent comme perturbateurs endocriniens (PE) connus pour être à l’origine de diverses maladies chroniques. Bien que leur impact sur la survenue de cancers soit difficile à évaluer, certains d’entre eux, comme le Bisphénol A (BPA) sont classés parmi les cancérigènes.Le développement des tumeurs cancéreuses est souvent la conséquence de mécanismes et d’interactions complexes avec leur microenvironnement. Et certaines interactions potentielles entre les cellules cancéreuses et leur microenvironnement peuvent ainsi orienter l’évolution de la tumeur. Parmi celles-ci, les jonctions communicantes sont les seules permettant une communication directe entre les cellules et ont été souvent associées par leur absence ou par la présence aberrante de leur protéine de structure, les connexines, dans le développement des tumeurs solides. Ainsi, dans le cas des tumeurs cérébrales que sont les glioblastomes, la présence d’une connexine particulière, la connexine 43 (Cx43), semble favoriser leur forte capacité invasive.Prenant en compte ce contexte particulier, les effets du microenvironnement tumoral (astrocytes), de l’exposition au BPA et de ses dérivés chlorés ainsi que le rôle de la Cx43 dans la capacité invasive des cellules de glioblastome humains ont été étudiés par des approches in utero et in vitro. A cette fin, pour l’approche in utero, les effets d’une exposition au BPA et ses dérivés chlorés ont été évalués par injections quotidiennes dans des souris femelles (20μg/kg/jour) durant la gestation (E0 à E21) et la lactation (J0 à J6), et ce, dans des contextes différents d’expression de Cx43 par l’utilisation de souris WT ou Cx43+/-. Les effets in vitro du BPA et ses dérivés chlorés ont été testés sur des cellules de glioblastome humain (lignée U251) exprimant différents niveaux de Cx43 par shRNA. Les résultats montrent que le BPA et ses dérivés chlorés affectent l’expression de la Cx43 et la communication jonctionnelle mais aussi la migration des cellules U251 selon le niveau d’expression de la Cx43. Les traitements in utero affectent également l’expression de la Cx43 des astrocytes et augmentent la communication jonctionnelle sans effet notable sur la migration. Cependant, d’une manière générale, les traitements ne semblent pas agir sur la fonction hémicanal de la Cx43. Par ailleurs, si le BPA augmente la migration, l’invasion 3D et le nombre d’invadopodes des cellules U251, les effets sont plus hétérogènes sur les cellules dont l’expression de Cx43 est diminuée. Ainsi, sur ces cellules, la migration 3D est diminuée après traitement à des dérivés chlorés alors que l’invasion 3D augmente après traitement au BPA et que le nombre d’invadopodes est augmenté quelles que soient les conditions de traitement. De plus, l’ajout de milieu d’astrocytes WT ou Cx43+/- semble augmenter la capacité migratoire et invasive des cellules U251 avec une forte augmentation du nombre des invadopodes quelle que soit le niveau d’expression de la Cx43 sauf pour la migration, inchangée, lorsque l’expression de la Cx43 est diminuée.En conclusion, le BPA et ses dérivés chlorés sont capables d’affecter l’expression et la fonction communicante de la Cx43. De plus, ces traitements ainsi que les astrocytes semblent favoriser les capacités migratoire et invasive des cellules U251 en augmentant le nombre des invadopodes selon le niveau d’expression de la Cx43.
Article
Purpose Understanding the effects of antiepileptic drugs on glial cells and glia-mediated inflammation is a new approach to future treatment of epilepsy. Little is known about direct effects of the antiepileptic drug lacosamide (LCM) on glial cells. Therefore, we aimed to study the LCM effects on glial viability, microglial activation, expression of gap-junctional (GJ) protein Cx43 as well as intercellular communication in an in vitro astrocyte-microglia co-culture model of inflammation. Methods Primary rat astrocytes co-cultures containing 5% (M5, “physiological” conditions) or 30% (M30, “pathological inflammatory” conditions) of microglia were treated with different concentrations of LCM [5, 15, 30, and 90 μg/ml] for 24 h. Glial cell viability was measured by MTT assay. Immunocytochemistry was performed to analyze the microglial activation state. Western blot analysis was used to quantify the astroglial Cx43 expression. The GJ cell communication was studied via Scrape Loading. Results A concentration-dependent incubation with LCM did not affect the glial cell viability both under physiological and pathological conditions. LCM induced a significant concentration-dependent decrease of activated microglia with parallel increase of ramified microglia under pathological inflammatory conditions. This correlated with an increase in astroglial Cx43 expression. Nevertheless, the functional coupling via GJs was significantly reduced after incubation with LCM. Conclusion LCM has not shown effects on the glial cell viability. The reduced GJ coupling by LCM could be related to its anti-epileptic activity. The anti-inflammatory glial features of LCM with inhibition of microglial activation under inflammatory conditions support beneficial role in epilepsy associated with neuroinflammation.
Article
Full-text available
Gap junctions mediate direct cell-to-cell communication by forming channels that physically couple cells, thereby linking their cytoplasm, permitting the exchange of molecules, ions, and electrical impulses. Gap junctions are assembled from connexin (Cx) proteins, with connexin 43 (Cx43) being the most ubiquitously expressed and best studied. While the molecular events that dictate the Cx43 life cycle have largely been characterized, the unusually short half-life of connexins of only 1-5 hours, resulting in constant endocytosis and biosynthetic replacement of gap junction channels has remained puzzling. The Cx43 C-terminal (CT) domain serves as the regulatory hub of the protein affecting all aspects of gap junction function. Here, deletion within the Cx43 CT (amino acids 256-289), a region known to encode key residues regulating gap junction turnover is employed to examine the effects of dysregulated Cx43 gap junction endocytosis using cultured cells (Cx43 ∆256-289 ) and a zebrafish model ( cx43 lh10 ). We report that this CT deletion causes defective gap junction endocytosis as well as increased gap junction intercellular communication (GJIC). Increased Cx43 protein content in cx 43 lh10 zebrafish, specifically in the cardiac tissue, larger gap junction plaques and longer Cx43 protein half-lives coincide with severely impaired development. Our findings demonstrate for the first time that Cx43 gap junction endocytosis is an essential aspect of gap junction function and when impaired, gives rise to significant physiological problems as revealed here for cardiovascular development and function. [Media: see text] [Media: see text] [Media: see text] [Media: see text] [Media: see text]
Article
Full-text available
Connexins are a family of transmembrane proteins that play a key role in cardiac physiology. Gap junctional channels put into contact the cytoplasms of connected cardiomyocytes, allowing the existence of electrical coupling. However, in addition to this fundamental role, connexins are also involved in cardiomyocyte death and survival. Thus, chemical coupling through gap junctions plays a key role in the spreading of injury between connected cells. Moreover, in addition to their involvement in cell-to-cell communication, mounting evidence indicates that connexins have additional gap junction-independent functions. Opening of unopposed hemichannels, located at the lateral surface of cardiomyocytes, may compromise cell homeostasis and may be involved in ischemia/reperfusion injury. In addition, connexins located at non-canonical cell structures, including mitochondria and the nucleus, have been demonstrated to be involved in cardioprotection and in regulation of cell growth and differentiation. In this review, we will provide, first, an overview on connexin biology, including their synthesis and degradation, their regulation and their interactions. Then, we will conduct an in-depth examination of the role of connexins in cardiac pathophysiology, including new findings regarding their involvement in myocardial ischemia/reperfusion injury, cardiac fibrosis, gene transcription or signaling regulation.
Article
Gap junctions are present in most tissues and play essential roles in various biological processes. However, we know surprisingly little about the molecular mechanisms underlying gap junction formation. Here, we uncover the essential role of a conserved EGF- and laminin-G-domain-containing protein nlr-1/CASPR in the regulation of gap junction formation in multiple tissues across different developmental stages in C. elegans. NLR-1 is located in the gap junction perinexus, a region adjacent to but not overlapping with gap junctions, and forms puncta before the clusters of gap junction channels appear on the membrane. We show that NLR-1 can directly bind to actin to recruit F-actin networks at the gap junction formation plaque, and the formation of F-actin patches plays a critical role in the assembly of gap junction channels. Our findings demonstrate that nlr-1/CASPR acts as an early stage signal for gap junction formation through anchoring of F-actin networks.
Article
Multifunctional matrix protein (M) of rabies virus (RABV) plays essential roles in the pathogenesis of rabies infection. Identification of M protein interacting partners in target hosts could help to elucidate the biological pathways and molecular mechanisms involved in the pathogenesis of this virus. In this study, two-dimensional Far-western blotting (2D-Far-WB) technique was applied to find possible matrix protein partners in the rat brainstem. Recombinant RABV M was expressed in Pichia pastoris (P. pastoris) and was partially purified. Subsequently, 2D-Far-WB determined six rat brainstem proteins interacted with recombinant M protein which were identified by mass spectrometry. Functional annotation by gene ontology analysis determined these proteins were involved in the regulation of synaptic transmission processes, metabolic process, and cell morphogenesis-cytoskeleton organization. The interaction of viral M protein with selected host proteins in mouse Neuro-2a cells infected with RABV was verified by super-resolution confocal microscopy. Molecular docking simulations also demonstrated the formation of RABV M complexes. However, further confirmation with co-immunoprecipitation (Co-IP), was only successful for M-actin cytoplasmic1 interaction. Totally, our study revealed actin cytoplasmic1 as a binding partner of M protein, which might have important role(s) in rabies pathogenesis.
Article
Lead (Pb) is one of the most common heavy metal contaminants in the environment. Pb can cause pathophysiological changes in several organ systems, including the cardiovascular system, but the molecular mechanism remains elusive. The study aimed to study the effects of Pb on Gap junction intercellular communication (GJIC) and its role in Pb-induced apoptosis. The present study aims to determine whether Pb-induced autophagy promotes apoptosis of rat cardiac myocytes (H9c2 cells) by downregulating GJIC using CCK-8 Kit, scrape loading/dye transfer assay, Annexin V/PI assays, Western blot analysis and double-immunofluorescence experiments. The results showed that Pb elicited cytotoxicity in a time- and concentration-dependent manner and led to increased apoptosis in a concentration-dependent manner in H9c2 cells. Pb also reduced GJIC in H9c2 cells in a concentration-dependent manner through the downregulation of connexin (Cx) 43. Inhibition of gap junctions by gap junction blocker carbenoxolone disodium (CBX) resulted in increased apoptosis. Furthermore, Pb increased autophagy in a concentration-dependent manner in H9c2 cells, promoting the internalization of Cx43 and targeting the autophagosome via light chain 3 (LC3). However, autophagy inhibitor 3-Methyladenine (3-MA) can slow down the downregulation of Cx43 induced by Pb in H9c2 cells. In conclusion, our results provide that Pb-decreased GJIC promotes apoptosis in cardiomyocytes. It is probably because Pb-induced autophagy exacerbates GJIC inhibition and downregulation of Cx43. These results suggest that GJIC plays a protective role in Pb-induced apoptosis in rat cardiomyocytes, and GJIC may be one of the targets of Pb-induced biological effects.
Thesis
Gap junctions in the normal and regenerating inner ear of chick hatchlings have been immunohistochemically and functionally examined. Additionally, the effect of blocked gap junction channels on the proliferation of supporting cells in response to hair cell death has been assessed. Studies have been conducted primarily in organotypic cultures of the auditory and vestibular sensory epithelium of chicken hatchlings. For regeneration studies, explants of basilar papilla and utricle were exposed to gentamicin, an ototoxic antibiotic, to induce hair cell loss. Following hair cell death, proliferation of supporting cells was upregulated and after 5 days immature, new hair cells were apparent. Blocking of gap junctions by carbenoxolone led to a significant reduction in the proliferation of supporting cells, suggesting an involvement of intercellular communication in the regeneration of hair cells. Expression of connexin 43 (Cx43) and the chicken-specific connexin 31 (cCx31) was examined by immunohistochemistry and confocal microscopy. cCx31 was strongly expressed in the normal basilar papilla and utricle. Cx43 was confined to the supporting cells of the auditory sensory epithelium, where its immunolabelling co-localised with cCx31. In response to hair cell loss, Cx43 was transiently downregulated. This finding, together with the absence of Cx43 in the sensory epithelium of the utricle, which has a constant turnover of hair cells, might point to an inhibitory effect of Cx43 on supporting cell proliferation. A dye-coupling assay, based on fluorescence recovery after photobleaching (FRAP), has been developed to examine the diffusion of the fluorescent tracer, calcein, between supporting cells in the intact tissue. Recovery of fluorescence occurred in supporting cells, but not in hair cells and was inhibited by the presence of carbenoxolone. Most notably, an asymmetric dye transfer across the basilar papilla was observed. The absence of directional permeability in the utricular macula, and in the drug-damaged basilar papilla, strongly suggests that the co-expression of cCx31 and Cx43 results in chemically rectifying gap junctions.
Article
Aim: To investigate the possible epigenetic pathogenesis of sporadic congenital cataract. Materials & methods: We conducted whole genome bisulfite sequencing on peripheral blood from sporadic binocular or monocular congenital cataract patients and cataract-free participants. Results: We found massive differentially methylated regions within the whole genomes between any two groups. Meanwhile, we identified five genes ( ACTN4, ACTG1, TUBA1A, TUBA1C, TUBB4B) for the binocular and control groups and TUBA1A for the monocular and control groups as the core differentially methylated region-related genes. The proteins encoded by these core genes are involved in building cytoskeleton and intercellular junctions. Conclusion: Changes in the methylation levels of core genes may disturb the function of cytoskeleton and intercellular junctions, eventually leading to sporadic congenital cataract.
Article
Full-text available
Gap junctions consist of arrays of intercellular channels that enable adjacent cells to communicate both electrically and metabolically. Gap junctions have a wide diversity of physiological functions, playing critical roles in both excitable and non-excitable tissues. Gap junction channels are formed by integral membrane proteins called connexins. Inherited or acquired alterations in connexins are associated with numerous diseases, including heart failure, neuropathologies, deafness, skin disorders, cataracts and cancer. Gap junctions are highly dynamic structures and by modulating the turnover rate of connexins, cells can rapidly alter the number of gap junction channels at the plasma membrane in response to extracellular or intracellular cues. Increasing evidence suggests that ubiquitination has important roles in the regulation of endoplasmic reticulum-associated degradation of connexins as well as in the modulation of gap junction endocytosis and post-endocytic sorting of connexins to lysosomes. In recent years, researchers have also started to provide insights into the physiological roles of connexin ubiquitination in specific tissue types. This review provides an overview of the advances made in understanding the roles of connexin ubiquitination in the regulation of gap junction intercellular communication and discusses the emerging physiological and pathophysiological implications of these processes.
Article
Full-text available
Connexins, the proteins that form gap junction channels, are polytopic plasma membrane (PM) proteins that traverse the plasma membrane bilayer four times. The insertion of five different connexins into the membrane of the ER was studied by synthesizing connexins in translation-competent cell lysates supplemented with pancreatic ER-derived microsomes, and by expressing connexins in vivo in several eucaryotic cell types. In addition, the subcellular distribution of the connexins was determined. In vitro-synthesis in the presence of microsomes resulted in the signal recognition particle-dependent membrane insertion of the connexins. The membrane insertion of all connexins was accompanied by an efficient proteolytic processing that was dependent on the microsome concentration. Endogenous unprocessed connexins were detectable in the microsomes used, indicating that the pancreatic microsomes serve as a competent recipient in vivo for unprocessed full length connexins. Although oriented with their amino terminus in the cytoplasm, the analysis of the cleavage reaction indicated that an unprecedented processing by signal peptidase resulted in the removal of an amino-terminal portion of the connexins. Variable amounts of similar connexin cleavage products were also identified in the ER membranes of connexin overexpressing cells. The amount generated correlated with the level of protein expression. These results demonstrate that the connexins contain a cryptic signal peptidase cleavage site that can be processed by this enzyme in vitro and in vivo in association with their membrane insertion. Consequently, a specific factor or condition must be required to prevent this aberrant processing of connexins under normal conditions in the cell.
Article
Full-text available
A correlation is made between the appearances of the nexus ("gap junction") as revealed by thin-section and by freeze-cleave electron microscopy techniques. These methods reveal different aspects of a complex subunit assembly forming the nexus membranes. In thin sections, the nexus is formed by the very close apposition of two "unit" membranes. The electron-opaque tracer, colloidal lanthanum hydroxide, outlines an aspect of electron-lucent subunits that project into the central region of the nexus. The freeze-cleave technique demonstrates novel membrane faces that are generated from within the interior of plasma membranes by splitting them into two lamellae (Lm): Lm 1 adjacent to the cytoplasm, and Lm 2 adjacent to the extracellular space. Each of the two membranes forming the nexus can be split into these two lamellae. On the new face of Lm 1, particles approximately 50 A in diameter are closely packed in an array which is often hexagonal with a 90–100 A center-to-center spacing. The two apposed lamellae (Lm 2-Lm 2) of the nexus are constructed of sheets of subunits in a similar array. The Lm 1 particles appear to extend into the Lm 2 subunits to form macromolecular complexes. The Lm 2 subunits extend to the center of the nexus to form the contacts outlined by lanthanum in sections. It is postulated that central hydrophilic channels may extend through the subunit assembly to provide a direct route for intercellular communication.
Article
Full-text available
The biogenesis and maintenance of asymmetry is crucial to many cellular functions including absorption and secretion, signalling, development and morphogenesis. Here we have directly visualized the segregation and trafficking of apical (glycosyl phosphatidyl inositol-anchored) and basolateral (vesicular stomatitis virus glycoprotein) cargo in living cells using multicolour imaging of green fluorescent protein variants. Apical and basolateral cargo segregate progressively into large domains in Golgi/trans-Golgi network structures, exclude resident proteins, and exit in separate transport containers. These remain distinct and do not merge with endocytic structures suggesting that lateral segregation in the trans-Golgi network is the primary sorting event. Fusion with the plasma membrane was detected by total internal reflection microscopy and reveals differences between apical and basolateral carriers as well as new 'hot spots' for exocytosis.
Article
Full-text available
We previously demonstrated that the gap junction protein connexin43 is translated as a 42-kD protein (connexin43-NP) that is efficiently phosphorylated to a 46,000-Mr species (connexin43-P2) in gap junctional communication-competent, but not in communication-deficient, cells. In this study, we used a combination of metabolic radiolabeling and immunoprecipitation to investigate the assembly of connexin43 into gap junctions and the relationship of this event to phosphorylation of connexin43. Examination of the detergent solubility of connexin43 in communication-competent NRK cells revealed that processing of connexin43 to the P2 form was accompanied by acquisition of resistance to solubilization in 1% Triton X-100. Immunohistochemical localization of connexin43 in Triton-extracted NRK cells demonstrated that connexin43-P2 (Triton-insoluble) was concentrated in gap junctional plaques, whereas connexin43-NP (Triton-soluble) was predominantly intracellular. Using either a 20 degrees C intracellular transport block or cell-surface protein biotinylation, we determined that connexin43 was transported to the plasma membrane in the Triton-soluble connexin43-NP form. Cell-surface biotinylated connexin43-NP was processed to Triton-insoluble connexin43-P2 at 37 degrees C. Connexin43-NP was also transported to the plasma membrane in communication defective, gap junction-deficient S180 and L929 cells but was not processed to Triton-insoluble connexin43-P2. Taken together, these results demonstrate that gap junction assembly is regulated after arrival of connexin43 at the plasma membrane and is temporally associated with acquisition of insolubility in Triton X-100 and phosphorylation to the connexin43-P2 form.
Article
Full-text available
In the plasma membrane of animal cells, many membrane-spanning proteins exhibit lower lateral mobilities than glycosylphosphatidylinositol (GPI)-linked proteins. To determine if the GPI linkage was a major determinant of the high lateral mobility of these proteins, we measured the lateral diffusion of chimeric membrane proteins composed of normally transmembrane proteins that were converted to GPI-linked proteins, or GPI-linked proteins that were converted to membrane-spanning proteins. These studies indicate that GPI linkage contributes only marginally (approximately twofold) to the higher mobility of several GPI-linked proteins. The major determinant of the high mobility of these proteins resides instead in the extracellular domain. We propose that lack of interaction of the extracellular domain of this protein class with other cell surface components allows diffusion that is constrained only by the diffusion of the membrane anchor. In contrast, cell surface interactions of the ectodomain of membrane-spanning proteins exemplified by the vesicular stomatitis virus G glycoprotein reduces their lateral diffusion coefficients by nearly 10-fold with respect to many GPI-linked proteins.
Article
Full-text available
Zonulae occludentes and gap junctions were examined both in the intact mouse liver and in a junction-rich membrane fraction from homogenized mouse liver. These preparations were visualized with the techniques of uranyl acetate staining en bloc, staining with colloidal lanthanum, negative staining with phosphotungstate, and freeze-cleaving. The zonula occludens is arranged as a meshwork of branching and anastomosing threadlike contacts sealing the lumen of the bile canaliculus from the liver intercellular space. The gap junction is characterized in section by a 20 A gap between the apposed junctional membrane outer leaflets, and permeation of this space with lanthanum or phosphotungstate reveals a polygonal lattice of subunits with a center-to-center spacing of 90-100 A. Freeze-cleaved gap junctions show a similar lattice. Extraction of junction-rich fractions with 60% aqueous acetone results in a disappearance of the 20 A gap in sectioned pellets and an inability to demonstrate the polygonal lattice with either the freeze-cleave or negative staining techniques. Extraction of the membranes with 50% acetone does not produce this effect. Thin-layer chromatography of the acetone extracts reveals a group of phospholipids in the 60% extract that are not detectable in the 50% extract. Acetone does not cause any detectable change in the structure of the zonula occludens, but the occluding junction becomes leaky to lanthanum following acetone treatment. The effects of other reagents on the junctions are reported.
Article
Full-text available
The fine structure and distribution of tight (zonula occludens) and gap junctions in epithelia of the rat pancreas, liver, adrenal cortex, epididymis, and duodenum, and in smooth muscle were examined in paraformaldehyde-glutaraldehyde-fixed, tracer-permeated (K-pyroantimonate and lanthanum), and freeze-fractured tissue preparations. While many pentalaminar and septilaminar foci seen in thin-section and tracer preparations can be recognized as corresponding to well-characterized freeze-fracture images of tight and gap junction membrane modifications, many others cannot be unequivocally categorized-nor can all freeze-etched aggregates of membrane particles. Generally, epithelia of exocrine glands (pancreas and liver) have moderate-sized tight junctions and large gap junctions, with many of their gap junctions basal to the junctional complex. In contrast, the adrenal cortex, a ductless gland, may not have a tight junction but does possess large gap junctions. Mucosal epithelia (epididymis and intestine) have extensive tight junctions, but their gap junctions are not as well developed as those of glandular tissue. Smooth muscle contains numerous small gap junctions The incidence, size, and configuration of the junctions we observed correlate well with the known functions of the junctions and of the tissues where they are found.
Article
Full-text available
A correlation is made between the appearances of the nexus ("gap junction") as revealed by thin-section and by freeze-cleave electron microscopy techniques. These methods reveal different aspects of a complex subunit assembly forming the nexus membranes. In thin sections, the nexus is formed by the very close apposition of two "unit" membranes. The electron-opaque tracer, colloidal lanthanum hydroxide, outlines an aspect of electron-lucent subunits that project into the central region of the nexus. The freeze-cleave technique demonstrates novel membrane faces that are generated from within the interior of plasma membranes by splitting them into two lamellae (Lm): Lm 1 adjacent to the cytoplasm, and Lm 2 adjacent to the extracellular space. Each of the two membranes forming the nexus can be split into these two lamellae. On the new face of Lm 1, particles approximately 50 A in diameter are closely packed in an array which is often hexagonal with a 90-100 A center-to-center spacing. The two apposed lamellae (Lm 2-Lm 2) of the nexus are constructed of sheets of subunits in a similar array. The Lm 1 particles appear to extend into the Lm 2 subunits to form macromolecular complexes. The Lm 2 subunits extend to the center of the nexus to form the contacts outlined by lanthanum in sections. It is postulated that central hydrophilic channels may extend through the subunit assembly to provide a direct route for intercellular communication.
Article
Full-text available
Connexins, the proteins that form gap junction channels, are polytopic plasma membrane (PM) proteins that traverse the plasma membrane bilayer four times. The insertion of five different connexins into the membrane of the ER was studied by synthesizing connexins in translation-competent cell lysates supplemented with pancreatic ER-derived microsomes, and by expressing connexins in vivo in several eucaryotic cell types. In addition, the subcellular distribution of the connexins was determined. In vitro-synthesis in the presence of microsomes resulted in the signal recognition particle-dependent membrane insertion of the connexins. The membrane insertion of all connexins was accompanied by an efficient proteolytic processing that was dependent on the microsome concentration. Endogenous unprocessed connexins were detectable in the microsomes used, indicating that the pancreatic microsomes serve as a competent recipient in vivo for unprocessed full length connexins. Although oriented with their amino terminus in the cytoplasm, the analysis of the cleavage reaction indicated that an unprecedented processing by signal peptidase resulted in the removal of an amino-terminal portion of the connexins. Variable amounts of similar connexin cleavage products were also identified in the ER membranes of connexin overexpressing cells. The amount generated correlated with the level of protein expression. These results demonstrate that the connexins contain a cryptic signal peptidase cleavage site that can be processed by this enzyme in vitro and in vivo in association with their membrane insertion. Consequently, a specific factor or condition must be required to prevent this aberrant processing of connexins under normal conditions in the cell.
Article
Full-text available
Intercellular gap junction channels are thought to form when oligomers of connexins from one cell (connexons) register and pair with connexons from a neighboring cell en route to forming tightly packed arrays (plaques). In the current study we used the rat mammary BICR-M1Rk tumor cell line to examine the trafficking, maturation, and kinetics of connexin43 (Cx43). Cx43 was conclusively shown to reside in the Golgi apparatus in addition to sites of cell-cell apposition in these cells and in normal rat kidney cells. Brefeldin A (BFA) blocked Cx43 trafficking to the surface of the mammary cells and also prevented phosphorylation of the 42-kD form of Cx43 to 44- and 46-kD species. However, phosphorylation of Cx43 occurred in the presence of BFA while it was still a resident of the ER or Golgi apparatus yielding a 43-kD form of Cx43. Moreover, the 42- and 43-kD forms of Cx43 trapped in the ER/Golgi compartment were available for gap junction assembly upon the removal of BFA. Mammary cells treated with BFA for 6 h lost preexisting gap junction "plaques," as well as the 44- and 46-kD forms of Cx43 and functional coupling. These events were reversible 1 h after the removal of BFA and not dependent on protein synthesis. In summary, we provide strong evidence that in BICR-M1Rk tumor cells: (a) Cx43 is transiently phosphorylated in the ER/Golgi apparatus, (b) Cx43 trapped in the ER/Golgi compartment is not subject to rapid degradation and is available for the assembly of new gap junction channels upon the removal of BFA, (c) the rapid turnover of gap junction plaques is correlated with the loss of the 44- and 46-kD forms of Cx43.
Article
Full-text available
The mechanism by which Golgi membrane proteins are retained within the Golgi complex in the midst of a continuous flow of protein and lipid is not yet understood. The diffusional mobilities of mammalian Golgi membrane proteins fused with green fluorescent protein from Aequorea victoria were measured in living HeLa cells with the fluorescence photobleaching recovery technique. The diffusion coefficients ranged from 3 × 10−9 square centimeters per second to 5 × 10−9 square centimeters per second, with greater than 90 percent of the chimeric proteins mobile. Extensive lateral diffusion of the chimeric proteins occurred between Golgi stacks. Thus, the chimeras diffuse rapidly and freely in Golgi membranes, which suggests that Golgi targeting and retention of these molecules does not depend on protein immobilization.
Article
Full-text available
Aquaporin-1 is present in the apical and basolateral membranes in proximal tubules and descending limbs of Henlé's loop. In order ot be able to study the routing of Aquaporin-1 and the regulation of Aquaporin-1-mediated transcellular water flow, we stably transfected LLC-PK1 and MDCK-HRS cell lines with an Aquaporin-1 expression construct. LLC-PK1 clone 7 and MDCK clone K integrated two and one copies, respectively, which was reflected in the amount of Aquaporin-1 mRNA expressed in both clones. The Aquaporin-1 protein levels, however, were similar. In both clones, immuno-electronmicroscopy showed extensive labelling of Aquaporin-1 on the basolateral plasma membrane, endosomal vesicles and the apical plasma membrane, including the microvilli. To measure transcellular water permeation, a simple method was applied using phenol-red as a cell-impermeant marker of concentration. In contrast to the native cell lines, both clones revealed a high transcellular osmotic water permeability, which could not be influenced by forskolin add/3-isobutyl-1-methylxanthine (IBMX) or the phorbol ester 12-O-tetradecanoyl 13-acetate (TPA). After glutaraldehyde fixation, it was inhibitable by HgCl2. These results indicate that targeting of Aquaporin-1 to the apical and basolateral plasma membrane is independent of cell type and show for the first time that water flow through a cultured epithelium can be blocked by mercurial compounds.
Article
Full-text available
Several different gap junction channel subunit isotypes, known as connexins, were synthesized in a cell-free translation system supplemented with microsomal membranes to study the mechanisms involved in gap junction channel assembly. Previous results indicated that the connexins were synthesized as membrane proteins with their relevant transmembrane topology. An integrated biochemical and biophysical analysis indicated that the connexins assembled specifically with other connexin subunits. No interactions were detected between connexin subunits and other co-translated transmembrane proteins. The connexins that were integrated into microsomal vesicles assembled into homo- and hetero-oligomeric structures with hydrodynamic properties of a 9S particle, consistent with the properties reported for hexameric gap junction connexons derived from gap junctions in vivo. Further, cell-free assembled homo-oligomeric connexons composed of beta1 or beta2 connexin were reconstituted into synthetic lipid bilayers. Single channel conductances were recorded from these bilayers that were similar to those measured for these connexons produced in vivo. Thus, this is the first direct evidence that the synthesis and assembly of a gap junction connexon can take place in microsomal membranes. Finally, the cell-free system has been used to investigate the properties of alpha1, beta1 and beta2 connexin to assemble into hetero-oligomers. Evidence has been obtained for a selective interaction between individual connexin isotypes and that a signal determining the potential hetero-oligomeric combinations of connexin isotypes may be located in the N-terminal sequence of the connexins.
Article
Full-text available
The mechanisms of localization and retention of membrane proteins in the inner nuclear membrane and the fate of this membrane system during mitosis were studied in living cells using the inner nuclear membrane protein, lamin B receptor, fused to green fluorescent protein (LBR-GFP). Photobleaching techniques revealed the majority of LBR-GFP to be completely immobilized in the nuclear envelope (NE) of interphase cells, suggesting a tight binding to heterochromatin and/or lamins. A subpopulation of LBR-GFP within ER membranes, by contrast, was entirely mobile and diffused rapidly and freely (D = 0. 41 +/- 0.1 microm2/s). High resolution confocal time-lapse imaging in mitotic cells revealed LBR-GFP redistributing into the interconnected ER membrane system in prometaphase, exhibiting the same high mobility and diffusion constant as observed in interphase ER membranes. LBR-GFP rapidly diffused across the cell within the membrane network defined by the ER, suggesting the integrity of the ER was maintained in mitosis, with little or no fragmentation and vesiculation. At the end of mitosis, nuclear membrane reformation coincided with immobilization of LBR-GFP in ER elements at contact sites with chromatin. LBR-GFP-containing ER membranes then wrapped around chromatin over the course of 2-3 min, quickly and efficiently compartmentalizing nuclear material. Expansion of the NE followed over the course of 30-80 min. Thus, selective changes in lateral mobility of LBR-GFP within the ER/NE membrane system form the basis for its localization to the inner nuclear membrane during interphase. Such changes, rather than vesiculation mechanisms, also underlie the redistribution of this molecule during NE disassembly and reformation in mitosis.
Article
Full-text available
The Golgi complex is a dynamic organelle engaged in both secretory and retrograde membrane traffic. Here, we use green fluorescent protein-Golgi protein chimeras to study Golgi morphology in vivo. In untreated cells, membrane tubules were a ubiquitous, prominent feature of the Golgi complex, serving both to interconnect adjacent Golgi elements and to carry membrane outward along microtubules after detaching from stable Golgi structures. Brefeldin A treatment, which reversibly disassembles the Golgi complex, accentuated tubule formation without tubule detachment. A tubule network extending throughout the cytoplasm was quickly generated and persisted for 5-10 min until rapidly emptying Golgi contents into the ER within 15-30 s. Both lipid and protein emptied from the Golgi at similar rapid rates, leaving no Golgi structure behind, indicating that Golgi membranes do not simply mix but are absorbed into the ER in BFA-treated cells. The directionality of redistribution implied Golgi membranes are at a higher free energy state than ER membranes. Analysis of its kinetics suggested a mechanism that is analogous to wetting or adsorptive phenomena in which a tension-driven membrane flow supplements diffusive transfer of Golgi membrane into the ER. Such nonselective, flow-assisted transport of Golgi membranes into ER suggests that mechanisms that regulate retrograde tubule formation and detachment from the Golgi complex are integral to the existence and maintenance of this organelle.
Article
Full-text available
To examine the trafficking, assembly, and turnover of connexin43 (Cx43) in living cells, we used an enhanced red-shifted mutant of green fluorescent protein (GFP) to construct a Cx43-GFP chimera. When cDNA encoding Cx43-GFP was transfected into communication-competent normal rat kidney cells, Cx43-negative Madin-Darby canine kidney (MDCK) cells, or communication-deficient Neuro2A or HeLa cells, the fusion protein of predicted length was expressed, transported, and assembled into gap junctions that exhibited the classical pentalaminar profile. Dye transfer studies showed that Cx43-GFP formed functional gap junction channels when transfected into otherwise communication-deficient HeLa or Neuro2A cells. Live imaging of Cx43-GFP in MDCK cells revealed that many gap junction plaques remained relatively immobile, whereas others coalesced laterally within the plasma membrane. Time-lapse imaging of live MDCK cells also revealed that Cx43-GFP was transported via highly mobile transport intermediates that could be divided into two size classes of <0.5 microm and 0.5-1.5 microm. In some cases, the larger intracellular Cx43-GFP transport intermediates were observed to form from the internalization of gap junctions, whereas the smaller transport intermediates may represent other routes of trafficking to or from the plasma membrane. The localization of Cx43-GFP in two transport compartments suggests that the dynamic formation and turnover of connexins may involve at least two distinct pathways.
Article
Full-text available
Hemichannels in the overlapping regions of apposing cells plasma membranes join to form gap junctions and provide an intercellular communication pathway. Hemichannels are also present in the nonjunctional regions of individual cells and their activity is gated by several agents, including calcium. However, their physiological roles are unknown. Using techniques of atomic force microscopy (AFM), fluorescent dye uptake assay, and laser confocal immunofluorescence imaging, we have examined the extracellular calcium-dependent modulation of cell volume. In response to a change in the extracellular physiological calcium concentration (1.8 to </=1.6 mM) in an otherwise isosmotic condition, real-time AFM imaging revealed a significant and reversible increase in the volume of cells expressing gap-junctional proteins (connexins). Volume change did not occur in cells that were not expressing connexins. However, after the transient or stable transfection of connexin43, volume change did occur. The volume increase was accompanied by cytochalasin D-sensitive higher cell stiffness, which helped maintain cell integrity. These cellular physical changes were prevented by gap-junctional blockers, oleamide and beta-glycyrrhetinic acid, or were reversed by returning extracellular calcium to the normal level. We conclude that nongap-junctional hemichannels regulate cell volume in response to the change in extracellular physiological calcium in an otherwise isosmotic situation.
Article
Full-text available
Communication-incompetent cell lines were transfected with connexin (Cx) 43 fused with enhanced green fluorescent protein (EGFP) to examine the relation between Cx distribution determined by fluorescence microscopy and electrical coupling measured at single-channel resolution in living cell pairs. Cx43-EGFP channel properties were like those of wild-type Cx43 except for reduced sensitivity to transjunctional voltage. Cx43-EGFP clustered into plaques at locations of cell-cell contact. Coupling was always absent in the absence of plaques and even in the presence of small plaques. Plaques exceeding several hundred channels always conferred coupling, but only a small fraction of channels were functional. These data indicate that clustering may be a requirement for opening of gap junction channels.
Article
Full-text available
Monitoring the fusion of constitutive traffic with the plasma membrane has remained largely elusive. Ideally, fusion would be monitored with high spatial and temporal resolution. Recently, total internal reflection (TIR) microscopy was used to study regulated exocytosis of fluorescently labeled chromaffin granules. In this technique, only the bottom cellular surface is illuminated by an exponentially decaying evanescent wave of light. We have used a prism type TIR setup with a penetration depth of ∼50 nm to monitor constitutive fusion of vesicular stomatitis virus glycoprotein tagged with the yellow fluorescent protein. Fusion of single transport containers (TCs) was clearly observed and gave a distinct analytical signature. TCs approached the membrane, appeared to dock, and later rapidly fuse, releasing a bright fluorescent cloud into the membrane. Observation and analysis provided insight about their dynamics, kinetics, and position before and during fusion. Combining TIR and wide-field microscopy allowed us to follow constitutive cargo from the Golgi complex to the cell surface. Our observations include the following: (1) local restrained movement of TCs near the membrane before fusion; (2) apparent anchoring near the cell surface; (3) heterogeneously sized TCs fused either completely; or (4) occasionally larger tubular-vesicular TCs partially fused at their tips.
Article
Full-text available
Total internal reflection fluorescence microscopy has been applied to image the final stage of constitutive exocytosis, which is the fusion of single post-Golgi carriers with the plasma membrane. The use of a membrane protein tagged with green fluorescent protein allowed the kinetics of fusion to be followed with a time resolution of 30 frames/s. Quantitative analysis allowed carriers undergoing fusion to be easily distinguished from carriers moving perpendicularly to the plasma membrane. The flattening of the carriers into the plasma membrane is seen as a simultaneous rise in the total, peak, and width of the fluorescence intensity. The duration of this flattening process depends on the size of the carriers, distinguishing small spherical from large tubular carriers. The spread of the membrane protein into the plasma membrane upon fusion is diffusive. Mapping many fusion sites of a single cell reveals that there are no preferred sites for constitutive exocytosis in this system.
Article
Full-text available
An essential feature of the first synapse in the retina is a negative feedback pathway from horizontal cells to cones. Here we show that at this synapse, connexin26 forms hemichannels on horizontal cell dendrites near the glutamate release site of the cones. Blocking these hemichannels hyperpolarizes horizontal cells, modulates the Ca2+ channels of the cones, and abolishes all feedback-mediated responses. We propose a feedback mechanism in which the activity of the Ca2+ channels and the subsequent glutamate release of the cones are modulated by a current through these hemichannels. Because the current through the hemichannels depends on the polarization of the horizontal cells, their activity modulates the output of the cones.
Article
Full-text available
Rat cortical astrocytes in pure culture are functionally coupled to neighboring cells via connexin (Cx) 43 gap junctions under ordinary conditions. Small fluorescent molecules such as Lucifer yellow (LY) pass between cell interiors via gap junctions, but do not enter the cells when externally applied. Subjecting rat and mouse cortical astrocytes to "chemical ischemia" by inhibition of glycolytic and oxidative metabolism induced permeabilization of cells to Lucifer yellow and ethidium bromide before loss of membrane integrity determined by dextran uptake and lactate dehydrogenase release. The gap junction blockers octanol and 18alpha-glycyrrhetinic acid markedly reduced dye uptake, suggesting that uptake was mediated by opening of unapposed hemichannels. Extracellular La(3+) also reduced dye uptake and delayed cell death. The purinergic blocker, oxidized ATP, was ineffective. Astrocytes isolated from mice with targeted deletion of the Cx43 coding DNA exhibited greatly reduced dye coupling and ischemia-induced dye uptake, evidence that dye uptake is mediated by Cx43 hemichannels. Dye coupling was reduced but not blocked by metabolic inhibition. Blockade of lipoxygenases or treatment with free radical scavengers reduced dye uptake by rat astrocytes, suggesting a role for arachidonic acid byproducts in hemichannel opening. Furthermore, permeabilization was accompanied by reduction in ATP levels and dephosphorylation of Cx43. Although hemichannel opening would tend to collapse electrochemical and metabolic gradients across the plasma membrane of dying cells, healthy cells might rescue dying cells by transfer of ions and essential metabolites via Cx43 gap junctions. Alternatively, dying astrocytes might compromise the health of neighboring cells via Cx43 gap junctions, thereby promoting the propagation of cell death.
Article
Full-text available
Recombinant proteins containing tetracysteine tags can be successively labeled in living cells with different colors of biarsenical fluorophores so that older and younger protein molecules can be sharply distinguished by both fluorescence and electron microscopy. Here we used this approach to show that newly synthesized connexin43 was transported predominantly in 100- to 150-nanometer vesicles to the plasma membrane and incorporated at the periphery of existing gap junctions, whereas older connexins were removed from the center of the plaques into pleiomorphic vesicles of widely varying sizes. Selective imaging by correlated optical and electron microscopy of protein molecules of known ages will clarify fundamental processes of protein trafficking in situ.
Article
Full-text available
Gap junctions are clustered channels between contacting cells through which direct intercellular communication via diffusion of ions and metabolites can occur. Two hemichannels, each built up of six connexin protein subunits in the plasma membrane of adjacent cells, can dock to each other to form conduits between cells. We have recently screened mouse and human genomic data bases and have found 19 connexin (Cx) genes in the mouse genome and 20 connexin genes in the human genome. One mouse connexin gene and two human connexin genes do not appear to have orthologs in the other genome. With three exceptions, the characterized connexin genes comprise two exons whereby the complete reading frame is located on the second exon. Targeted ablation of eleven mouse connexin genes revealed basic insights into the functional diversity of the connexin gene family. In addition, the phenotypes of human genetic disorders caused by mutated connexin genes further complement our understanding of connexin functions in the human organism. In this review we compare currently identified connexin genes in both the mouse and human genome and discuss the functions of gap junctions deduced from targeted mouse mutants and human genetic disorders.
Article
Intercellular gap junction channels are thought to form when oligomers of connexins from one cell (connexons) register and pair with connexons from a neighboring cell en route to forming tightly packed arrays (plaques). In the current study we used the rat mammary BICR-M1Rk tumor cell line to examine the trafficking, maturation, and kinetics of connexin43 (Cx43). Cx43 was conclusively shown to reside in the Golgi apparatus in addition to sites of cell-cell apposition in these cells and in normal rat kidney cells. Brefeldin A (BFA) blocked Cx43 trafficking to the surface of the mammary cells and also prevented phosphorylation of the 42-kD form of Cx43 to 44- and 46-kD species. However, phosphorylation of Cx43 occurred in the presence of BFA while it was still a resident of the ER or Golgi apparatus yielding a 43-kD form of Cx43. Moreover, the 42- and 43-kD forms of Cx43 trapped in the ER/Golgi compartment were available for gap junction assembly upon the removal of BFA. Mammary cells treated with BFA for 6 h lost preexisting gap junction "plaques," as well as the 44- and 46-kD forms of Cx43 and functional coupling. These events were reversible 1 h after the removal of BFA and not dependent on protein synthesis. In summary, we provide strong evidence that in BICR-M1Rk tumor cells: (a) Cx43 is transiently phosphorylated in the ER/Golgi apparatus, (b) Cx43 trapped in the ER/Golgi compartment is not subject to rapid degradation and is available for the assembly of new gap junction channels upon the removal of BFA, (c) the rapid turnover of gap junction plaques is correlated with the loss of the 44- and 46-kD forms of Cx43.
Article
Gap junction membrane channels mediate electrical and metabolic coupling between adjacent cells. The structure of a recombinant cardiac gap junction channel was determined by electron crystallography at resolutions of 7.5 angstroms in the membrane plane and 21 angstroms in the vertical direction. The dodecameric channel was formed by the end-to-end docking of two hexamers, each of which displayed 24 rods of density in the membrane interior, which is consistent with an α-helical conformation for the four transmembrane domains of each connexin subunit. The transmembrane α-helical rods contrasted with the double-layered appearance of the extracellular domains. Although not indicative for a particular type of secondary structure, the protein density that formed the extracellular vestibule provided a tight seal to exclude the exchange of substances with the extracellular milieu.
Article
Hemichannels in the overlapping regions of apposing cells plasma membranes join to form gap junctions and provide an intercellular communication pathway. Hemichannels are also present in the nonjunctional regions of individual cells and their activity is gated by several agents, including calcium. However, their physiological roles are unknown. Using techniques of atomic force microscopy (AFM), fluorescent dye uptake assay, and laser confocal immunofluorescence imaging, we have examined the extracellular calcium-dependent modulation of cell volume. In response to a change in the extracellular physiological calcium concentration (1.8 to ≤1.6 mM) in an otherwise isosmotic condition, real-time AFM imaging revealed a significant and reversible increase in the volume of cells expressing gap-junctional proteins (connexins). Volume change did not occur in cells that were not expressing connexins. However, after the transient or stable transfection of connexin43, volume change did occur. The volume increase was accompanied by cytochalasin D-sensitive higher cell stiffness, which helped maintain cell integrity. These cellular physical changes were prevented by gap-junctional blockers, oleamide and β-glycyrrhetinic acid, or were reversed by returning extracellular calcium to the normal level. We conclude that nongap-junctional hemichannels regulate cell volume in response to the change in extracellular physiological calcium in an otherwise isosmotic situation.
Article
In cells treated with brefeldin A (BFA), movement of newly synthesized membrane proteins from the endoplasmic reticulum (ER) to the Golgi apparatus was blocked. Surprisingly, the glycoproteins retained in the ER were rapidly processed by cis/medial Golgi enzymes but not by trans Golgi enzymes. An explanation for these observations was provided from morphological studies at both the light and electron microscopic levels using markers for the cis/medial and trans Golgi. They revealed a rapid and dramatic redistribution to the ER of components of the cis/medial but not the trans Golgi in response to treatment with BFA. Upon removal of BFA, the morphology of the Golgi apparatus was rapidly reestablished and proteins normally transported out of the ER were efficiently and rapidly sorted to their final destinations. These results suggest that BFA disrupts a dynamic membrane-recycling pathway between the ER and cis/medial Golgi, effectively blocking membrane transport out of but not back to the ER.
Article
Observations using electron microscopy and freeze-etching techniques show that both metabolic coupling and ionic coupling in fibroblasts are associated with the appearance of ``gap'' junctions.
Article
Connexin43 (Cx43) is an integral plasma membrane protein that forms gap junctions between vertebrate cells. We have used sucrose gradient fractionation and chemical cross-linking to study the first step in gap junction assembly, oligomerization of Cx43 monomers into connexon channels. In contrast with other plasma membrane proteins, multisubunit assembly of Cx43 was specifically and completely blocked when endoplasmic reticulum (ER)-to-Golgi transport was inhibited by 15 degrees C incubation, carbonyl cyanide m-chloro-phenylhydrazone, or brefeldin A or in CHO cell mutants with temperature-sensitive defects in secretion. Additional experiments indicated that connexon assembly occurred intracellularly, most likely in the trans-Golgi network. These results describe a post-ER assembly pathway for integral membrane proteins and have implications for the relationship between membrane protein oligomerization and intracellular transport.
Article
Fluorescence recovery after photobleaching (FRAP) has been a powerful tool for characterizing the mobility of cell surface membrane proteins. However, the application of FRAP to the study of intracellular membrane proteins has been hampered by the lack of specific probes and their physical inaccessibility in the cytoplasm. We have measured the mobility of a model transmembrane protein, the temperature-sensitive vesicular stomatitis viral membrane glycoprotein (ts-O45-G), in transit from the endoplasmic reticulum (ER) to the Golgi complex. ts-O45-G accumulates in the ER at nonpermissive temperature (39.5 degrees C) and is transported via the Golgi complex to the surface upon shifting cells to the permissive temperature (31 degrees C). Rhodamine-labeled Fab fragments against a cytoplasmic epitope of ts-O45-G (rh-P5D4-Fabs) were microinjected into cells to visualize the intracellular viral membrane protein and to determine its mobility by FRAP with a confocal microscope. Moreover, we have measured the effects of microinjected antibodies against beta-COP on the mobility of ts-O45-G following release of the temperature block. FRAP was essentially complete when rh-P5D4-Fab-injected cells were bleached either following release of labeled ts-O45-G from the ER or upon its accumulation at 20 degrees C in the trans-Golgi network (TGN). In contrast, recovery was reduced by about one third when infected cells had been injected with antibodies that bind to beta-COP in vivo. The diffusion constant of mobile ts-O45-G under all conditions was approximately 10 x 10(-10) cm2/s. These results validate the feasibility of FRAP for the study of an intracellular transmembrane protein and provide the first evidence that such a protein is highly mobile.
Article
An indirect immunogold labeling technique was applied to replicas of freeze-fractured membranes of rapidly frozen unfixed cells. The endogenous gap junction protein Cx43 of BICR/M1Rk rat mammary tumor cells was preferentially identified in quasi-crystalline gap junction plaques as were the transfected connexins Cx40, Cx43, and Cx45 in HeLa (human cervical carcinoma) cells. With this method we also detected contact areas with dispersed gap junction channels which are the only structural correlation for endogenous Cx45 in HeLa wild-type cells where no gap junction plaques exist. In double-transfected HeLa cells a colocalization of Cx40 and Cx43 was occasionally detected in quasi-crystalline gap junction plaques, whereas in contact areas with dispersed particles only one Cx type was present. Our results indicate that functional gap junction channels exist outside the quasi-crystalline plaques.
Article
The mechanisms and carriers responsible for exocytic protein trafficking between the trans-Golgi network (TGN) and the plasma membrane remain unclear. To investigate the dynamics of TGN-to-plasma membrane traffic and role of the cytoskeleton in these processes we transfected cells with a GFP-fusion protein, vesicular stomatitis virus G protein tagged with GFP (VSVG3-GFP). After using temperature shifts to block VSVG3-GFP in the endoplasmic reticulum and subsequently accumulate it in the TGN, dynamics of TGN-to-plasma membrane transport were visualized in real time by confocal and video microscopy. Both small vesicles (<250 nm) and larger vesicular-tubular structures (>1.5 µm long) are used as transport containers (TCs). These TCs rapidly moved out of the Golgi along curvilinear paths with average speeds of ∼0.7 µm/second. Automatic computer tracking objectively determined the dynamics of different carriers. Fission and fusion of TCs were observed, suggesting that these late exocytic processes are highly interactive. To directly determine the role of microtubules in post-Golgi traffic, rhodamine-tubulin was microinjected and both labeled cargo and microtubules were simultaneously visualized in living cells. These studies demonstrated that exocytic cargo moves along microtubule tracks and reveals that carriers are capable of switching between tracks. Key words: VSVG, GFP, Golgi, TGN, Cytoskeleton, Microtubule, Exocytosis
Article
Gap junction membrane channels mediate electrical and metabolic coupling between adjacent cells. The structure of a recombinant cardiac gap junction channel was determined by electron crystallography at resolutions of 7.5 angstroms in the membrane plane and 21 angstroms in the vertical direction. The dodecameric channel was formed by the end-to-end docking of two hexamers, each of which displayed 24 rods of density in the membrane interior, which is consistent with an alpha-helical conformation for the four transmembrane domains of each connexin subunit. The transmembrane alpha-helical rods contrasted with the double-layered appearance of the extracellular domains. Although not indicative for a particular type of secondary structure, the protein density that formed the extracellular vestibule provided a tight seal to exclude the exchange of substances with the extracellular milieu.
Article
Gap junction channels assemble as dodecameric complexes, in which a hexameric connexon (hemichannel) in one plasma membrane docks end-to-end with a connexon in the membrane of a closely apposed cell to provide direct cell-to-cell communication. Synthesis, assembly, and trafficking of the gap junction channel subunit proteins referred to as connexins, largely appear to follow the general secretory pathway for membrane proteins. The connexin subunits can assemble into homo-, as well as distinct hetero-oligomeric connexons. Assembly appears to be based on specific signals located within the connexin polypeptides. Plaque formation by the clustering of gap junction channels in the plane of the membrane, as well as channel degradation are poorly understood processes that are topics of current research. Recently, we tagged connexins with the autofluorescent reporter green fluorescent protein (GFP), and its cyan (CFP), and yellow (YFP) color variants and combined this reporter technology with single, and dual-color, high resolution deconvolution microscopy, computational volume rendering, and time-lapse microscopy to examine the detailed organization, structural composition, and dynamics of gap junctions in live cells. This technology provided for the first time a realistic, three-dimensional impression of gap junctions as they appear in the plasma membranes of adjoining cells, and revealed an excitingly detailed structural organization of gap junctions never seen before in live cells. Here, I summarize recent progress in areas encompassing the synthesis, assembly and structural composition of gap junctions with a special emphasis on the recent results we obtained using cell-free translation/ membrane-protein translocation, and autofluorescent reporters in combination with live-cell deconvolution microscopy.
Article
To study the organization of gap junctions in living cells, the connexin isotypes α1(Cx43), β1(Cx32) and β2(Cx26) were tagged with the autofluorescent tracer green fluorescent protein (GFP) and its cyan (CFP) and yellow (YFP) color variants. The cellular fate of the tagged connexins was followed by high-resolution fluorescence deconvolution microscopy and time-lapse imaging. Comprehensive analyses demonstrated that the tagged channels were functional as monitored by dye transfer, even under conditions where the channels were assembled solely from tagged connexins. High-resolution images revealed a detailed structural organization, and volume reconstructions provided a three-dimensional view of entire gap junction plaques. Specifically, deconvolved dual-color images of gap junction plaques assembled from CFP- and YFP-tagged connexins revealed that different connexin isotypes gathered within the same plaques. Connexins either codistributed homogeneously throughout the plaque, or each connexin isotype segregated into well-separated domains. The studies demonstrate that the mode of channel distribution strictly depends on the connexin isotypes. Based on previous studies on the synthesis and assembly of connexins I suggest that channel distribution is regulated by intrinsic connexin isotype specific signals. Movies available on-line: http://www.biologists.com/JCS/movies/jcs1735.html
Article
Gap junctions are specialized cell-cell junctions that mediate intercellular communication. They are composed of connexin proteins, which form transmembrane channels for small molecules [1, 2]. The C-terminal tail of connexin-43 (Cx43), the most widely expressed connexin member, has been implicated in the regulation of Cx43 channel gating by growth factors [3-5]. The Cx43 tail contains various protein interaction sites, but little is known about binding partners. To identify Cx43-interacting proteins, we performed pull-down experiments using the C-terminal tail of Cx43 fused to glutathione-S-transferase. We find that the Cx43 tail binds directly to tubulin and, like full-length Cx43, sediments with microtubules. Tubulin binding to Cx43 is specific in that it is not observed with three other connexins. We established that a 35-amino acid juxtamembrane region in the Cx43 tail, which contains a presumptive tubulin binding motif, is necessary and sufficient for microtubule binding. Immunofluorescence and immunoelectron microscopy studies reveal that microtubules extend to Cx43-based gap junctions in contacted cells. However, intact microtubules are dispensable for the regulation of Cx43 gap-junctional communication. Our findings suggest that, in addition to its well-established role as a channel-forming protein, Cx43 can anchor microtubule distal ends to gap junctions and thereby might influence the properties of microtubules in contacted cells.
  • D F Hulser
  • B Rehkopf
  • O Traub
Hulser, D. F., Rehkopf, B. & Traub, O. (1997) Exp. Cell Res. 233, 240–251.
  • P M Deen
  • S Nielsen
  • R J Bindels
  • C H Van Os
Deen, P. M., Nielsen, S., Bindels, R. J. & van Os, C. H. (1997) Pflugers Arch. 433, 780–787.
  • K Willecke
  • J Eiberger
  • J Degen
  • D Eckardt
  • A Romualdi
  • M Guldenagel
  • U Deutsch
  • G Sohl
Willecke, K., Eiberger, J., Degen. J., Eckardt, D., Romualdi, A., Guldenagel, M., Deutsch, U. & Sohl, G. (2002) Biol. Chem. 383, 725–737.
  • B N Giepmans
  • I Verlaan
  • T Hengeveld
  • H Janssen
  • J Calafat
  • M M Falk
  • W H Moolenaar
Giepmans, B. N., Verlaan, I., Hengeveld, T., Janssen, H., Calafat, J., Falk, M. M. & Moolenaar, W. H. (2001) Curr. Biol. 11, 1364–1368.
  • D A Goodenough
  • J P Revel
Goodenough, D. A. & Revel, J. P. (1970) J. Cell Biol. 45, 272–290.
  • L S Musil
  • D A Goodenough
Musil, L. S. & Goodenough, D. A. (1991) J. Cell Biol. 115, 1357–1374.
  • D W Laird
  • M Castillo
  • L Kasprzak
Laird, D. W., Castillo, M. & Kasprzak, L. (1995) J. Cell Biol. 131, 1193–1203.
  • Falk
  • Toomre
  • Storrie