Next Article in Journal
Molecular Dynamics Simulations to Investigate How PZM21 Affects the Conformational State of the μ-Opioid Receptor Upon Activation
Previous Article in Journal
A Multi-Scale Study of Water Dynamics under Confinement, Exploiting Numerical Simulations in Relation to NMR Relaxometry, PGSE and NMR Micro-Imaging Experiments: An Application to the Clay/Water Interface
Previous Article in Special Issue
Gap Junctions in the Bone Marrow Lympho-Hematopoietic Stem Cell Niche, Leukemia Progression, and Chemoresistance
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of ROS/RNS in Preeclampsia: Are Connexins the Missing Piece?

by
María F. Rozas-Villanueva
1,2,
Paola Casanello
3,4 and
Mauricio A. Retamal
1,5,*
1
Centro de Fisiología Celular e Integrativa, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago 7690000, Chile
2
Programa de Doctorado en Ciencias e Innovación en Medicina, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago 7690000, Chile
3
Department of Obstetrics, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 7690000, Chile
4
Department of Neonatology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 7690000, Chile
5
Programa de Comunicación Celular de Cáncer, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago 7690000, Chile
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2020, 21(13), 4698; https://doi.org/10.3390/ijms21134698
Submission received: 29 May 2020 / Revised: 18 June 2020 / Accepted: 28 June 2020 / Published: 30 June 2020
(This article belongs to the Special Issue Connexin-Based Channels in Inflammatory Processes)

Abstract

:
Preeclampsia is a pregnancy complication that appears after 20 weeks of gestation and is characterized by hypertension and proteinuria, affecting both mother and offspring. The cellular and molecular mechanisms that cause the development of preeclampsia are poorly understood. An important feature of preeclampsia is an increase in oxygen and nitrogen derived free radicals (reactive oxygen species/reactive nitrogen species (ROS/RNS), which seem to be central players setting the development and progression of preeclampsia. Cell-to-cell communication may be disrupted as well. Connexins (Cxs), a family of transmembrane proteins that form hemichannels and gap junction channels (GJCs), are essential in paracrine and autocrine cell communication, allowing the movement of signaling molecules between cells as well as between the cytoplasm and the extracellular media. GJCs and hemichannels are fundamental for communication between endothelial and smooth muscle cells and, therefore, in the control of vascular contraction and relaxation. In systemic vasculature, the activity of GJCs and hemichannels is modulated by ROS and RNS. Cxs participate in the development of the placenta and are expressed in placental vasculature. However, it is unknown whether Cxs are modulated by ROS/RNS in the placenta, or whether this potential modulation contributes to the pathogenesis of preeclampsia. Our review addresses the possible role of Cxs in preeclampsia, and the plausible modulation of Cxs-formed channels by ROS and RNS. We suggest these factors may contribute to the development of preeclampsia.

1. Introduction

Preeclampsia (PE) is a multisystemic disease of pregnancy, characterized by hypertension and proteinuria after 20 weeks of gestation [1]. Preeclampsia is associated with endothelial activation of the placental and maternal vasculature. The estimated prevalence of PE is 3–7% of pregnancies, which may vary according to the region, maternal age, ethnicity, and the socioeconomic status of the studied population [2]. Several risk factors for PE have been proposed, the most relevant being the presence of antiphospholipid antibodies, previous history of pre-eclampsia, and maternal diabetes [3,4]. PE is associated with several complications, such as hypertensive encephalopathy, eclampsia, renal and liver failure, pulmonary edema, coagulopathies, and uteroplacental dysfunction that may lead to intrauterine growth restriction [5]. Thus, PE is considered one of the leading causes of maternal and neonatal mortality and morbidity worldwide [6]. Furthermore, PE may exert its consequences weeks or years after pregnancy, in the form of cardiovascular risk and postpartum depression [7], chronic hypertension, metabolic syndrome, and cardiac structural changes leading to diastolic dysfunction [8]. Even though PE represents a major health problem, its etiology is not yet well understood. The main theory is based on a two-stage event: First, an inefficient invasion of the spiral arteries’ walls by the extravillous trophoblast during placentation, which leads to incomplete degradation of the muscle layer of the arterial wall, and therefore to high resistance and low flow of the placental vascular bed, generating a decreased uteroplacental perfusion. Second, the uteroplacental ischemia leads to hypoxia and necrotic damage of the trophoblast, which releases soluble factors such as fms-like tyrosine kinase 1 (sFLT1) and endoglin (sENG) and other inflammatory molecules, leading to endothelial dysfunction and its effects on maternal and fetal systemic disease [6,9,10]. However, further research is necessary to advance our understanding of the molecular mechanisms for PE and its sequels.

2. Inflammation and Redox Imbalance in Preeclampsia

Normal pregnancy has been identified as a transient inflammatory state because the site of implantation resembles an open wound. In this context, several groups have shown an improvement in in vitro fertilization success rates when a mechanical injury in the endometrium, like a biopsy, is done. It seems that the success of implantation may be dependent on an initial “injury-induced inflammatory reaction”, and in agreement with this idea, proinflammatory T helper-1 (Th)-1 cells and proinflammatory cytokines, such as IL-6, IL-8, and TNFα have been found in the implantation zone. However, this response has to be carefully regulated and is followed by an anti-inflammatory state, characterized by a shift towards Th-2 and IL-10 immunity, which allows the pregnancy to continue [11,12,13].
On the other hand, insufficient and/or inefficient remodeling of the maternal vascular bed in PE leads to placental ischemia, which promotes a chronic immune and redox imbalance, characterized by a reduced natural killer activity [14], and an increase of Th-1 response leading to an exacerbated inflammatory status [15,16]. This chronic inflammatory and hypoxic condition increase reactive oxygen species (ROS) and reactive nitrogen species (RNS) production such as O2-, OH, H2O2, NO, and ONOO-. These molecules are produced by cell metabolism, and under physiological conditions work as cell signaling molecules [17,18,19,20]. However, excess ROS and RNS production causes the inappropriate balance of the antioxidant defense, leading to cellular damage of proteins, lipids, and DNA, through different posttranslational modifications, such as peroxidation, S-nitrosylation, carbonylation, and protein nitration. To delve deeper into the matter of nitric oxide (NO), which is recognized as a significant endothelium-derived relaxing factor, evidence shows that the enzyme levels (eNOS) that synthesize NO are not modified in PE, yet NO bioavailability is reduced [21,22]. This observation could be explained by the transformation of NO to peroxynitrite (ONOO-), which is enabled by the high availability of superoxide anion (O2-) in this condition [23,24]. Therefore, the reduction in NO vascular levels decreases vascular relaxation and provides a partial explanation for the hypertensive response in PE. Moreover, molecules that increase ROS/RNS production, also decrease proliferation and invasion of the extravillous trophoblast (EVTs) [25], therefore distressing the peri-implantation period.
In PE many vasoactive factors are compromised. Thus, for example, in human omental arteries taken from women with PE, an increase in vasopressin and KCl-induced vasoconstriction and an absent acetylcholine-dependent relaxation compared with normotensive gravidas was observed [26]. Additionally, the plasma concentration of the vasoactive neuropeptide Y (NYP) was elevated in preeclamptic women compared with normal pregnancy [27]. Both vasopressin and NYP, increase Ca2+ concentrations in rat and human smooth muscle cells [28,29]. It is well known that intracellular Ca2+ plays an essential role in the endothelial-smooth muscle cell communication and vascular physiology. On the other hand, several groups have focused on how ROS/RNS and inflammatory molecules alter Ca2+ homeostasis and its signaling in PE [30]. In this review, we analyze how ROS/RNS could contribute to the generation of PE in the implantation period and the worsening of PE through changes in endothelial/smooth muscle cell communication mediated by connexin (Cx)-based channels.

3. General Characteristics of Cx-Based Channels

Cxs are a family of proteins present in almost all cell types in the human body [31]. Twenty-one different coding genes for Cxs have been described [32], which are named accordingly to their predicted molecular weight (i.e., Cx43, ~43 kDa). Despite the great diversity of the members of this family [33], they have a high amino acid sequence conservation. Cxs are formed by four transmembrane domains connected by two extracellular and one intracellular loop. Both the N- and C-terminus are located at the cytoplasmic side of the membrane [31,34]. In some Cxs, the N-terminal portion has been described inside the channel forming the intracellular pore funnel [35,36,37], where it acts as part of the voltage sensor [38].
Accordingly, mutations in the N-terminal modify the channel properties, such as permeability, selectivity, and open probability [39,40,41,42,43]. Concerning the C-terminal domain, it is the most variable region between Cxs, not only in their amino acid composition but also in its length. For instance, Cx23 has the shortest C-terminal, and Cx62 has the largest one among all Cxs. Additionally, the C-terminal domain is a prominent site for posttranslational modifications in Cxs [44]. Hence, most of phosphorylations [45,46,47], S-nitrosylation [48], and proteolytic cleavage sites [49,50] are found in the C-terminus. A particular feature of Cxs is that they form two types of channels, hemichannels and GJCs (Figure 1). Despite that these two types of channels can be formed by the same type of protein, they show several differences in terms of cellular localization, permeability, control mechanisms, and cell function [51].

3.1. Hemichannels

Hemichannels are formed by hexamers of Cxs [35,52]. These channels form a central pore by which ions and molecules up to 1.2 kDa move following their electrochemical gradient [53]. Thus, experiments performed mostly in vitro, have demonstrated that when hemichannels increase their open probability (mostly caused by a reduction in the divalent cation concentration in the extracellular media [54,55,56]), they mediate an increase in extracellular concentration of ATP [57], glutamate [58], GSH [59], lactate [60], and NAD+ [61], and intracellular glucose [62], Na+ [63], and Ca2+ [64,65]. Despite the initial thought that hemichannels were non-selective, it is clear now that they have selective permeability, mainly determined by the size, 3D structure, and charge of the molecule in transit [66]. For example, Cx43 hemichannels are permeable to ethidium bromide (1+, 394 g/mol), but are not permeable to urea or glycerol which are non-charged and smaller (60 and 92 g/mol respectively) [67]. Similarly, Cx30 hemichannels are permeable to glucose (180 g/mol) but not to water (18 g/mol) [67].
Under physiological conditions, hemichannels are mostly closed, probably to “prevent” leakage of molecules such as ATP and amino acids [68]. This low hemichannel activity is maintained mainly by extracellular Ca2+ and Mg2+ concentrations in the mM range [54,55,56] and by a negative membrane potential [53,56,69]. However, some molecules found in vivo may favor an increased but controlled hemichannel opening. Thus, hemichannels increase their activity under redox reducing conditions [70,71] as well as when stimulated by linoleic acid [72]. It is plausible that hemichannel activity in vivo is higher than reported due to a lack of appropriate conditions for determining hemichannel activity in vitro. Accordingly, there is increasing evidence that hemichannel activity in vivo is high enough to have a significant role in information processing in the central nervous system [73]. For instance, blocking Cx43 hemichannels, with a mimetic peptide, in the lateral amygdala of adult rats induced amnesia in animals under a fear conditioning protocol [74], while under normal conditions these animals would remember the distressing experience. This result was associated with a lower release of glutamate and D-serine from astrocytes [74]. Other examples of hemichannel activity under physiological conditions is the release of D-serine from astrocytic hemichannels, which regulates glutamate-induced currents in prefrontal mouse cortex neurons [75], and the release of ATP from hippocampal astrocytes through Cx43 hemichannels, which regulate the basal activity of CA1 pyramidal neurons in a mouse model [76]. These data strongly suggest that hemichannels activity is low under physiological conditions, but high enough to mediate critical cellular processes.
Under pathological conditions, hemichannel activity is notoriously increased [68,77]. Hemichannels with higher activity are known as “leaky hemichannels” [68] because they represent a leaked route for molecules from the cytosol to extracellular space through the plasma membrane. This uncontrolled leaky flux of molecules leads to cell malfunction, and in some cases, cell death [68,77]. Some mutations in Cx-genes have been associated with a massive hemichannel activity, which in turn has been linked to KID (Keratitis–Ichthyosis–Deafness) syndrome [78,79,80,81] and X-linked Charcot-Marie-Tooth disease [82]. In addition, lateralization of Cx43—forming hemichannels—contributes to cardiac arrhythmias and leads to lethality in animal models of Duchenne muscular dystrophy [77].
Our group and others have proposed that molecules associated to oxidative stress modulate hemichannel opening [70,83,84,85,86]. For example, Cx43 hemichannel S-nitrosylation exacerbates hemichannel activity in cerebral ischemia [87] and inflammatory models [62]. Despite that the molecular mechanism is unknown, Cx cysteine (Cys) oxidation has been proposed as a mediator in this phenomenon [70,86,88]. In particular, the effect of NO on Cx43 hemichannels was associated with an increase in their open probability [87], and in the case of Cx46 hemichannels, with modifications of their electrophysiological properties and changes in their permeability to large molecules [88]. On the other hand, the oxidation of a putative free Cys in one of the extracellular loops seems to induce the closure of the hemichannel by 4-HNE [86]. Interestingly, the Ca2+ sensor of Cx26 is located at the interface of transmembrane 1 and the extracellular loop 1 [89], and the Cx32 sensor is constituted of several asparagine residues in the extracellular loop 1 [55]. Suggesting that any posttranslational modification in the extracellular loop 1 could modulate (in any direction) the Ca2+ sensor effect. Recently, our group suggested that the S-nitrosylation of Cx46 hemichannels could play an important role in cataract formation [90]. In summary, pathological conditions, such as mutations and inflammation, can induce leaky hemichannels leading to cell injury.

3.2. GJCs

GJCs are formed by the serial docking of two hemichannels [35]. These types of channels are permeable to ions [91] and to second messenger molecules, such as Ca2+, IP3, and cAMP [92,93,94,95,96], glucose [97,98], small peptides [99,100] and miRNAs [101,102]. Under physiological conditions, GJCs allow metabolic and electrical coordination between neighboring cells, which are essential for correct tissue functioning. Proinflammatory conditions have been associated with a decrease in gap junctional communication between cells in different tissues, such as brain, lung, and liver [62,103,104,105]. In contrast, other pathological conditions such as brain ischemia, where gap junctional coupling is maintained, this route of communication serves as a way to spread damaging signals to healthy cells [106] in a sort of “bystander effect”. Accordingly, evidence shows that astroglial Cx43 GJCs mediate neuronal and glial death in regions far beyond the infarcted zone [106,107,108]. Therefore, maintaining cellular communication is critical for retaining normal tissue function. However, under pathological conditions, the characteristics of gap junction-mediated communication could enhance or diminish cell injury to neighboring cells, probably depending on the nature of the injury. The effect of free radicals on GJC communication is not clear, but in general, is associated with a decrease of intercellular coupling [62,109,110].

4. The Role of Hemichannels and GJCs in the Vasculature

Human and animal studies have evidenced that Cx37, Cx40, Cx43, and Cx45 are expressed in the vascular system, and play an important role in its physiology [111,112]. Despite some differences in localization due to vessel size, vascular territory, and species [111,113], it is accepted that Cx37 is mostly confined to endothelial cells and is expressed to a lesser degree in smooth muscle cells [114,115]. Cx40 is expressed in endothelial and smooth muscle cells [116,117], Cx43 is relatively evenly distributed in endothelial and smooth muscle cells [97,100], and Cx45 is only present in smooth muscle cells [118]. The coupling between smooth muscles via GJCs [119] allows not only metabolic but also electrical coordination between cells, which in turn helps to synchronize the muscle contraction to obtain a meaningful response [120]. Also, smooth muscle cells are coupled to endothelial cells [121,122,123], producing a cellular syncytium in which cellular interactions converge in the control of the vascular tone [124]. The role exerted by Cxs has been evaluated with Gap27, a mimetic peptide that blocks Cx37 and Cx43 channels. When applied to the ex vivo system of a mesenteric resistance artery, a decrease in basal myogenic vasoconstriction was observed [124], suggesting that a “signal” is transferred from endothelial cells to smooth muscle cells via GJCs [121]. As mentioned before, GJCs are permeable to ions and signaling molecules; therefore, there are several candidates to be “the signal” crossing from endothelial to smooth muscle cells, with the potential to control the myoendothelial tone. One of the most important signaling molecules which participates in the vascular tone is Ca+2, which initiates the contraction in smooth muscle. The elevation of Ca+2 concentrations within the endothelium activates a negative feedback loop by increasing the synthesis of vasoactive molecules such as NO and endothelium-derived hyperpolarization factor (EDHF), promoting the relaxation of the vessels [125]. EDHF, an endothelium-derived vasodilator, is crucial in small arteries, and although there is no clarity as to the precise nature of this molecule, it seems that it is permeable through gap junctions, since a good correlation between the presence of functional gap junction and the activity of EDHF [126,127,128] has been demonstrated. Another possible “signal” moving through GJCs is IP3. Indeed, GJCs formed y Cx43 are permeable to IP3 [48]. This allows IP3 to flow from smooth muscle to endothelial cells, where it increases intracellular Ca2+ concentration. In turn, this change in intracellular Ca2+ activates NO production [129] which travels back to smooth muscle cells inducing vasorelaxation [48,130]. There is a dogma that claims, that the NO produced by endothelial cells “crosses freely” the plasma membranes until it reaches the cytoplasm of smooth muscle cells [131]. However, Figueroa et al. [132] suggested that NO uses Cxs 37, 40, and 43-hemichannels to easily permeate the plasma membrane of endothelial and smooth muscle cells, playing a pivotal role in the NO-dependent control of vascular tone. Furthermore, at the myoendothelial junction, NO autoregulates its transit, modifying cysteine residues on GJ channels to alter its permeability, through posttranslational modifications such as S-nitrosylation or denitrosylation, which makes channels more and less permeable, respectively [48]. It should be noted that Cxs not only facilitate the movement of NO through membranes but in turn, they can affect NO production. For instance, Cx37 interacts with eNOS, decreasing its activity and therefore decreasing NO levels [133]. Additionally, Cx40-/- mice have lower levels of eNOS compared to wild type mice [134]. In summary, GJCs and hemichannels formed by Cxs, communicate and coordinate chemical and electrical signals involved in the contraction and relaxation of the vessels through protein–protein interactions and facilitating the diffusion of vasoactive molecules.

5. Role of Cx-Based Channels in the Placenta

The placenta is an organ that allows the exchange of metabolites, gases, and waste products between the fetus and the mother. Cx32, Cx37, Cx40, Cx43, and Cx45 are expressed in human placenta [135,136,137,138,139,140,141,142,143] (Table 1). While Cx32 and Cx43 were described in the syncytiotrophoblast, a multinucleated cell in the placental bed, yet no function has been attributed to these Cxs in this particular location [141]. On the other hand, Cx43 has been described mostly forming GJCs between cytotrophoblast cells and between cytotrophoblast cells and syncytiotrophoblast [135], where it seems to regulate the fusion of cytotrophoblast into syncytiotrophoblast [142], warranting placental growth and maturation [143]. Two possible mechanisms of action have been proposed in this process. Firstly, there might be a “signal(s)” that moves between cells that allows cell fusion and, therefore, syncytiotrophoblast formation. Secondly, Cx43 coordinates the effect of other proteins such as ZO-1 through protein–protein interactions [138]. Lastly, these two mechanisms may act together in a concerted manner to achieve cytotrophoblast fusion and syncyalization.
Cx37 and Cx40 have been described in endothelial cells derived from placental vessels [137]. Although there is no evidence of their role in this particular territory, it is likely that Cx37 and Cx40 do not deviate far from the function established in other vascular regions, i.e., working as key molecules in controlling vascular tone. Cx40 and Cx45 are prominently expressed in the EVT of anchoring cell columns [136]. In these cells, Cx40 supports EVT proliferation and prevents their differentiation into an invasive phenotype. Meanwhile, Cx45 expression is restricted to placentas before nine weeks of gestation, but its role is unknown [136,143,144].

6. Could Hemichannels and GJCs Mediate Part of the ROS/RNS Response in Preeclampsia?

We analyze here the effect of inflammatory mediators (ROS/RNS) in Cxs-based channels function and how these modifications might be associated with vascular alterations in preeclamptic placentas (Figure 2).
Cx37: Even though Cx37 has been found in endothelial cells of chorionic arteries, its function has not been characterized in this particular territory. Furthermore, the presence and/or expression of this protein has not been evaluated in the placenta of preeclamptic patients.
In the presence of NO, Cx37-mediated gap junction communication is diminished [125,145,146]. Pogoda and co-workers demonstrated that in HeLa cells, this mechanism includes the inhibition of a tyrosine phosphatase (SHP-2), which removes the phosphorylation in tyrosine residue 332 on the C-terminal region of Cx37. This reduction in SHP-2 activity associates with a decrease in intracellular Ca2+ exchange between endothelial and smooth muscle cells [125]. The latter may represent a reduction of intercellular communication with other molecules as well.
Since NO bioavailability in PE is decreased, it could be suggested that the intercellular communication mediated by Cx37 could be increased. If the whole issue was centered on NO, it would be very counter-intuitive for the PE setting, because it would mean that the pathway for NO and other vasodilator molecules would be increased. However, as previously stated, there is no evidence demonstrating the presence and/or variation in expression of Cx37 in PE placentas nor the effect of other ROS/RNS on the activity of Cx37 GJCs. Therefore, in PE, in addition to posttranslational modifications in this Cx, a reduction of its expression could be suggested.
Contrary to the effect of NO on GJCss, in rat endothelial cells, NO increases Cx37 hemichannels activity and is permeable through them [132], suggesting a sort of paracrine signaling pathway. In preeclampsia, where NO levels are decreased, Cx37 hemichannel activity would be reduced, therefore diminishing one of NO “paracrine” releasing pathways. This mechanism is supported by evidence in a mouse Cx37-KO model, where a reduction of NO release was observed when compared with wild type animals [147].
Cx40: As previously mentioned, Cx40 may be found in two key locations in the placenta. First, being a part of EVT anchoring columns, where it colocalizes with proliferation markers, such as ki67. In this site, the presence of Cx40 seems to prevent the invasive pathway of EVT. As far as we know, there are no reports showing an effect of redox changes upon Cx40 levels or activity in EVT in preeclampsia. However, in rat brain tissue, oxidative stress increases Cx40 protein levels, while maintaining the same amount of mRNA, and that prevention of oxidative stress with N-acetylcysteine inhibits the increase in Cx40 protein seen without treatment [148,149]. Hence, oxidative stress in PE may be the cause of posttranslational modifications and/or an increase of protein expression of Cx40, which prevents the invasive pathway of EVT.
Second, Cx40 is found in placental and maternal vasculature. It has been observed that the lack of Cx40 (Cx40-/- mice) decreases NO production in a renin-dependent hypertension model [150]. This finding suggests that Cx40 may play a role in NO production and that the lack of Cx40 may exacerbate the hypertension response through the downregulation of vasodilator molecules. However, in another study, lack of Cx40 was associated with angiotensin II independent hypertension, and it did not correlate to NO or another endothelial-derived vasodilator [151]. This suggests that there might be a non-canonical pathway by which Cx40 is associated with hypertension without interfering with vasodilator molecules passage between neighboring cells. Also, a NO/cGMP-dependent de novo formation of Cx40-mediated gap junction coupling was observed in HeLa and HUVEC cells [152,153]. Based on these results, we could suggest that the reduction of NO levels seen in preeclampsia could impact the Cx40-mediated gap junction by reducing its coupling. This might not only exacerbate the reduced levels of NO by interfering with its propagation in the muscle-endothelial junction but also exacerbate the hypertensive response by interfering with the action of other vasodilator molecules such as EDHF. Regarding hemichannels, in rat endothelial cells, NO increases Cx40-hemichannel activity and permeates through them [132]. Thus, a decrease of NO bioavailability in PE could decrease Cx40-hemichannel activity, while also reducing the release of NO and its mediated vasodilatation, exacerbating once again the hypertensive response.
Cx43: In pregnant sheep, increased Cx43 gap junction function was associated with an increase in sustained Ca2+ burst and NO-induced vasodilatation [154]. In this work, Gap27 (which has been used as a hemichannel and GJC blocker) was used as a specific inhibitor of Cx43 [155,156]. Even though the authors claim that the effect was mediated by GJCs [154], the participation of hemichannels cannot be discarded. Moreover, the opening of Cx43 hemichannels mediates the uptake of Ca2+ from the extracellular space [64]. Taking into account these possibilities, we suggest that in normal pregnancy, Cx43-hemichannel and/or GJC functions, increase intracellular Ca2+ dynamics, which in turn increases NO production and its vasodilatory effect. Therefore, the exacerbated prooxidant and inflammatory milieu in PE could induce posttranslational modifications in Cx43, decreasing (in permeability or open probability) the hemichannel and/or GJCs function, reducing the vasodilator capacity of endothelial cells. Additionally, proinflammatory molecules can decrease Cx43 expression [157,158], therefore in PE, a similar phenomenon could be expected.
On the other hand, NO can increase Cx43 hemichannel activity in astrocytes. If the same mechanism operates in PE placentas, a decrease in Cx43 hemichannel activity in the syncytiotrophoblast layer of the placenta, due to lower bioavailability of NO, could be expected. Additionally, since NO uses Cx43-hemichannels as a pathway between the intra and extracellular space, a decrease in the release of NO could be expected as well [132]. As mentioned before the IP3 permeability through Cx43 GJCs is modulated by the S-nitrosylation of C271 [48]: Therefore, a decrease in the bioavailability of NO in PE could maintain Cx43 GJCs mainly in a closed state [48]. This could affect, at least theoretically, the flow of IP3 between smooth muscle and endothelial cells and also reduce the diffusion of EDHF and other vasodilator molecules, and as a consequence an increase in the placental vascular resistance.
Lower Ca2+ and Mg2+ plasma levels correlate with a higher risk of developing PE [159,160]. Accordingly, Ca2+ supplementation might decrease the risk of hypertension and therefore PE [161,162]. The hemichannel opening is controlled by intracellular and extracellular Ca2+ and Mg2+ concentrations, where a decrease of extracellular divalent cations increases hemichannel opening [55,163]. In consequence, a reduction of the extracellular concentration of these two ions in PE, could increase hemichannel opening. On the other hand, Cx43 hemichannels are permeable to ATP [164], and in a condition with an increased hemichannel open probability, a higher extracellular ATP concentration could be expected. This extracellular nucleotide, activates P2X and some P2Y receptors, and both can increase intracellular Ca2+ concentration [165]. In a healthy pregnancy, extracellular ATP induces intracellular Ca2+ bursts, modulating NO production [166]. However, in human preeclamptic umbilical endothelial cells, the Ca2+ and NO response to extracellular ATP is much lower [167]. A possible explanation of this, is that a sustained efflux of ATP through hemichannels, could induce P2 receptors desensitization, and therefore a loss of Ca2+ response and NO production.
Additionally, Cx43 GJCs seems to potentiate the role of Ca2+ as an NO inductor, via coordination of endothelial cell responses [154]. Usually, in experiments that mimic inflammatory and/or oxidative conditions, Cx43 GJCs come close together [62,109,110]. Therefore, in PE inflammatory and/or oxidative conditions could decrease Ca2+-induced NO production, via inhibition of GJCs communication.
As mentioned before, GJCs are permeable to second messenger and molecules such as miRNAs. There are several miRNAs that are associated with trophoblastic differentiation, migration and invasion [168]. Cx43 GJCs are permeable to several miRNAs [102,169]. Interestingly, Cx43 GJCs allows the exchange of miRNAs between bone-marrow-derived mesenchymal stem cells or cancer cells and endothelial cells [101,170,171]. Therefore, if Cx43 GJCs communication is reduced in PE (either because they are closed or their expression is reduced), a decrease of the flux of these molecules could be expected and malfunctioning of trophoblast’s function.
Cx32 and Cx45: There is no information regarding the role of Cx32 and Cx45 in preeclampsia. Moreover, the information about how ROS and RNS molecules affect the activity of these proteins in the placental vasculature is scarce. The need for experiments that would shed light on the role of these channel proteins in the placental vascular tone in PE is evident.

7. Conclusions and Future Directions

There is extensive evidence demonstrating that Cxs play a pivotal role in the regulation of systemic vascular function. Although the information regarding the role and function of Cxs in the placental vascular bed is scarce, this does not mean that Cxs are less relevant in this organ. On the contrary, we propose that Cxs might be directly involved in the maternal hypertensive disorder in response to an insufficient trophoblast invasion in PE.
We have analyzed the effect of oxidative stress, a key feature of preeclampsia, on Cxs in other territories. It is plausible that Cxs posttranslational modifications induced by ROS/RNS might be key elements in the vascular pathophysiology in PE. Thus, we suggest that the overall effect of ROS/RNS in the placental vascular cells is to decrease Cx-formed channels activity, which in turn reduces NO bioavailability and vasodilatation (Figure 3). In addition, it is possible that ROS/RNS cause lateralization of Cxs so that Cxs may be found in an aberrant location, i.e., increasing the expression of hemichannels while reducing gap junction Cxs—as it occurs in cardiac muscle Cx43 of Duchenne muscular dystrophy mice [77]. The possible increase in mislocalized hemichannels would increase the loss of NO to extracellular space, and reduce NO vasodilatory capacity. This review included results obtained in animals as well as humans. Research focused on the role of Cxs in women with PE is scarce; therefore, future investigations regarding the molecular mechanisms that modulate the Cx function in PE are needed. This review is an attempt to provoke the study not only of the effect of Cx dysregulation in the human placenta, but also to investigate the factors that induce these changes. Additionally, we advocate for the evaluation of non-canonical Cx functions and channel properties, as there could be a direct role of these proteins in trophoblast invasion and proliferation. Consequently, we encourage developing and enhancing new research on the C-terminal tail of Cxs, i.e., the target of this type of protein modifications in the preeclamptic placenta.

Author Contributions

M.F.R.-V., P.C., and M.A.R. designed, wrote, and edited the paper. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by FONDECYT [1160227 (MAR)] and Proyecto Interno Universidad del Desarrollo [23.400.159 (MFR-V)].

Acknowledgments

We would like to thank Walter Duran for his enormous contribution to the edition of this work.

Conflicts of Interest

The authors have no conflict of interest to declare.

References

  1. ACOG Practice Bulletin No. 202. Obstet. Gynecol. 2019, 133, e1–e25.
  2. Geographic variation in the incidence of hypertension in pregnancy. World Health Organization International Collaborative Study of Hypertensive Disorders of Pregnancy. Am. J. Obstet. Gynecol. 1988, 158, 80–83.
  3. Noris, M.; Perico, N.; Remuzzi, G. Mechanisms of disease: Pre-eclampsia. Nat. Clin. Pract. Nephrol. 2005, 1, 98–114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Duckitt, K.; Harrington, D. Risk factors for pre-eclampsia at antenatal booking: Systematic review of controlled studies. Br. Med. J. 2005, 330, 565–567. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Kuklina, E.V.; Ayala, C.; Callaghan, W.M. Hypertensive disorders and severe obstetric morbidity in the United States. Obstet. Gynecol. 2009, 113, 1299–1306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Myatt, L.; Roberts, J.M. Preeclampsia: Syndrome or Disease? Curr. Hypertens. Rep. 2015, 17, 83. [Google Scholar] [CrossRef] [PubMed]
  7. Caropreso, L.; de Azevedo Cardoso, T.; Eltayebani, M.; Frey, B.N. Preeclampsia as a risk factor for postpartum depression and psychosis: A systematic review and meta-analysis. Arch. Women’s Ment. Health 2019. [Google Scholar] [CrossRef] [PubMed]
  8. Hauspurg, A.; Countouris, M.E.; Catov, J.M. Hypertensive Disorders of Pregnancy and Future Maternal Health: How Can the Evidence Guide Postpartum Management? Curr. Hypertens. Rep. 2019, 21, 96. [Google Scholar] [CrossRef]
  9. Brosens, I.A.; Robertson, W.B.; Dixon, H.G. The role of the spiral arteries in the pathogenesis of pre-eclampsia. J. Pathol. 1970, 101, Pvi. [Google Scholar] [PubMed]
  10. Rana, S.; Lemoine, E.; Granger, J.; Karumanchi, S.A. Preeclampsia: Pathophysiology, Challenges, and Perspectives. Circ. Res. 2019, 124, 1094–1112. [Google Scholar] [CrossRef]
  11. Dekel, N.; Gnainsky, Y.; Granot, I.; Racicot, K.; Mor, G. The role of inflammation for a successful implantation. Am. J. Reprod. Immunol. 2014, 72, 141–147. [Google Scholar] [CrossRef] [PubMed]
  12. Mor, G.; Cardenas, I.; Abrahams, V.; Guller, S. Inflammation and pregnancy: The role of the immune system at the implantation site. Ann. N. Y. Acad. Sci. 2011, 1221, 80–87. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Griffith, O.W.; Chavan, A.R.; Protopapas, S.; Maziarz, J.; Romero, R.; Wagner, G.P. Embryo implantation evolved from an ancestral inflammatory attachment reaction. Proc. Natl. Acad. Sci. USA 2017, 114, E6566–E6575. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Parham, P. NK cells and trophoblasts: Partners in pregnancy. J. Exp. Med. 2004, 200, 951–955. [Google Scholar] [CrossRef] [Green Version]
  15. Cubro, H.; Kashyap, S.; Nath, M.C.; Ackerman, A.W.; Garovic, V.D. The Role of Interleukin-10 in the Pathophysiology of Preeclampsia. Curr. Hypertens. Rep. 2018, 20, 36. [Google Scholar] [CrossRef] [PubMed]
  16. Harmon, A.C.; Cornelius, D.C.; Amaral, L.M.; Faulkner, J.L.; Cunningham, M.W.; Wallace, K.; LaMarca, B. The role of inflammation in the pathology of preeclampsia. Clin. Sci. 2016, 130, 409–419. [Google Scholar] [CrossRef] [Green Version]
  17. Chiarello, D.I.; Abad, C.; Rojas, D.; Toledo, F.; Vázquez, C.M.; Mate, A.; Sobrevia, L.; Marín, R. Oxidative stress: Normal pregnancy versus preeclampsia. Biochim. Biophys. Acta-Mol. Basis Dis. 2019, 1866, 165354. [Google Scholar] [CrossRef]
  18. Burton, G.J.; Jauniaux, E. Oxidative stress. Best Pract. Res. Clin. Obstet. Gynaecol. 2011, 25, 287–299. [Google Scholar] [CrossRef] [Green Version]
  19. Duhig, K.; Chappell, L.C.; Shennan, A.H. Oxidative stress in pregnancy and reproduction. Obstet. Med. 2016, 9, 113–116. [Google Scholar] [CrossRef] [Green Version]
  20. Movafagh, S.; Crook, S.; Vo, K. Regulation of hypoxia-inducible Factor-1a by reactive oxygen species: New developments in an old debate. J. Cell. Biochem. 2015, 116, 696–703. [Google Scholar] [CrossRef]
  21. Goulopoulou, S.; Davidge, S.T. Molecular mechanisms of maternal vascular dysfunction in preeclampsia. Trends Mol. Med. 2015, 21, 88–97. [Google Scholar] [CrossRef]
  22. Seligman, S.P.; Buyon, J.P.; Clancy, R.M.; Young, B.K.; Abramson, S.B. The role of nitric oxide in the pathogenesis of preeclampsia. Am. J. Obstet. Gynecol. 1994, 171, 944–948. [Google Scholar] [CrossRef]
  23. Tsukimori, K.; Maeda, H.; Ishida, K.; Nagata, H.; Koyanagi, T.; Nakano, H. The superoxide generation of neutrophils in normal and preeclamptic pregnancies. Obstet. Gynecol. 1993, 81, 536–540. [Google Scholar] [PubMed]
  24. Roggensack, A.M.; Zhang, Y.; Davidge, S.T. Evidence for Peroxynitrite Formation in the Vasculature of Women with Preeclampsia. Hypertension 1999, 33, 83–89. [Google Scholar] [CrossRef] [PubMed]
  25. Mendes, S.; Timóteo-Ferreira, F.; Almeida, H.; Silva, E. New insights into the process of placentation and the role of oxidative uterine microenvironment. Oxid. Med. Cell. Longev. 2019, 2019. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Pascoal, I.F.; Lindheimer, M.D.; Nalbantian-Brandt, C.; Umans, J.G. Preeclampsia selectively impairs endothelium-dependent relaxation and leads to oscillatory activity in small omental arteries. J. Clin. Investig. 1998, 101, 464–470. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Paiva, S.P.C.; Veloso, C.A.; Campos, F.F.C.; Carneiro, M.M.; Tilan, J.U.; Wang, H.; Umans, J.G.; Zukowska, Z.; Kitlinska, J. Elevated levels of neuropeptide Y in preeclampsia: A pilot study implicating a role for stress in pathogenesis of the disease. Neuropeptides 2016, 55, 127–135. [Google Scholar] [CrossRef] [Green Version]
  28. Wier, W.G.; Mauban, J.R.H. Imaging sympathetic neurogenic Ca2 + signaling in blood vessels. Auton. Neurosci. Basic Clin. 2017, 207, 59–66. [Google Scholar] [CrossRef]
  29. Gal, C.S.L.; Herbert, J.M.; Delisee, C.; Schaeffer, P.; Raufaste, D.; Garcia, C.; Dol, F.; Marty, E.; Maffrand, J.P.; Le Fur, G. Effect of SR-49059, a vasopressin V(1a) antagonist, on human vascular smooth muscle cells. Am. J. Physiol.-Hear. Circ. Physiol. 1995, 268, H404–H410. [Google Scholar]
  30. Steinert, J.R.; Wyatt, A.W.; Jacob, R.; Mann, G.E. Redox modulation of Ca2+ signaling in human endothelial and smooth muscle cells in pre-eclampsia. Antioxid. Redox Signal. 2009, 11, 1149–1163. [Google Scholar] [CrossRef]
  31. Rackauskas, M.; Neverauskas, V.; Skeberdis, V.A. Diversity and properties of connexin gap junction channels. Medicina (Kaunas) 2010, 46, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. SOHL, G.; Willecke, K. Gap junctions and the connexin protein family. Cardiovasc. Res. 2004, 62, 228–232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Bosco, D.; Haefliger, J.-A.; Meda, P. Connexins: Key mediators of endocrine function. Physiol. Rev. 2011, 91, 1393–1445. [Google Scholar] [CrossRef] [Green Version]
  34. Beyer, E.C.; Berthoud, V.M. Gap junction structure: Unraveled, but not fully revealed. F1000Research 2017, 6, 568. [Google Scholar] [CrossRef] [Green Version]
  35. Maeda, S.; Nakagawa, S.; Suga, M.; Yamashita, E.; Oshima, A.; Fujiyoshi, Y.; Tsukihara, T. Structure of the connexin 26 gap junction channel at 3.5 Å resolution. Nature 2009, 458, 597–602. [Google Scholar] [CrossRef]
  36. Maeda, S.; Tsukihara, T. Structure of the gap junction channel and its implications for its biological functions. Cell. Mol. Life Sci. 2011, 68, 1115–1129. [Google Scholar] [CrossRef] [PubMed]
  37. Oshima, A.; Tani, K.; Hiroaki, Y.; Fujiyoshi, Y.; Sosinsky, G.E. Three-dimensional structure of a human connexin26 gap junction channel reveals a plug in the vestibule. Proc. Natl. Acad. Sci. USA 2007, 104, 10034–10039. [Google Scholar] [CrossRef] [Green Version]
  38. Kronengold, J.; Srinivas, M.; Verselis, V.K. The N-Terminal Half of the Connexin Protein Contains the Core Elements of the Pore and Voltage Gates. J. Membr. Biol. 2012, 245, 453–463. [Google Scholar] [CrossRef] [Green Version]
  39. Oh, S.; Verselis, V.K.; Bargiello, T.A. Charges dispersed over the permeation pathway determine the charge selectivity and conductance of a Cx32 chimeric hemichannel. J. Physiol. 2008, 586, 2445–2461. [Google Scholar] [CrossRef]
  40. Essenfelder, G.M.; Bruzzone, R.; Lamartine, J.; Charollais, A.; Blanchet-Bardon, C.; Barbe, M.T.; Meda, P.; Waksman, G. Connexin30 mutations responsible for hidrotic ectodermal dysplasia cause abnormal hemichannel activity. Hum. Mol. Genet. 2004, 13, 1703–1714. [Google Scholar] [CrossRef]
  41. Gemel, J.; Lin, X.; Veenstra, R.D.; Beyer, E.C. N-terminal residues in Cx43 and Cx40 determine physiological properties of gap junction channels, but do not influence heteromeric assembly with each other or with Cx26. J. Cell Sci. 2006, 119, 2258–2268. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Dong, L.; Liu, X.; Li, H.; Vertel, B.M.; Ebihara, L. Role of the N-terminus in permeability of chicken connexin45.6 gap junctional channels. J. Physiol. 2006, 576, 787–799. [Google Scholar] [CrossRef] [PubMed]
  43. Batir, Y.; Bargiello, T.A.; Dowd, T.L. Structural studies of N-terminal mutants of Connexin 26 and Connexin 32 using 1H NMR spectroscopy. Arch. Biochem. Biophys. 2016, 608, 8–19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Aasen, T.; Johnstone, S.; Vidal-Brime, L.; Lynn, K.; Koval, M. Connexins: Synthesis, Post-Translational Modifications, and Trafficking in Health and Disease. Int. J. Mol. Sci. 2018, 19, 1296. [Google Scholar] [CrossRef] [Green Version]
  45. Lampe, P.D.; Lau, A.F. Regulation of Gap Junctions by Phosphorylation of Connexins. Arch. Biochem. Biophys. 2000, 384, 205–215. [Google Scholar] [CrossRef]
  46. Shin, J.L.; Solan, J.L.; Lampe, P.D. The regulatory role of the C-terminal domain of connexin43. Cell Commun. Adhes. 2001, 8, 271–275. [Google Scholar] [CrossRef]
  47. Grosely, R.; Kopanic, J.L.; Nabors, S.; Kieken, F.; Spagnol, G.; Al-Mugotir, M.; Zach, S.; Sorgen, P.L. Effects of Phosphorylation on the Structure and Backbone Dynamics of the Intrinsically Disordered Connexin43 C-terminal Domain. J. Biol. Chem. 2013, 288, 24857–24870. [Google Scholar] [CrossRef] [Green Version]
  48. Straub, A.C.; Billaud, M.; Johnstone, S.R.; Best, A.K.; Yemen, S.; Dwyer, S.T.; Looft-Wilson, R.; Lysiak, J.J.; Gaston, B.; Palmer, L.; et al. Compartmentalized connexin 43 s-nitrosylation/denitrosylation regulates heterocellular communication in the vessel wall. Arterioscler. Thromb. Vasc. Biol. 2011, 31, 399–407. [Google Scholar] [CrossRef] [Green Version]
  49. De Bock, M.; Wang, N.; Decrock, E.; Bultynck, G.; Leybaert, L. Intracellular Cleavage of the Cx43 C-Terminal Domain by Matrix-Metalloproteases: A Novel Contributor to Inflammation? Mediat. Inflamm. 2015, 2015, 1–18. [Google Scholar] [CrossRef] [Green Version]
  50. Kudo-Sakamoto, Y.; Akazawa, H.; Ito, K.; Takano, J.; Yano, M.; Yabumoto, C.; Naito, A.T.; Oka, T.; Lee, J.K.; Sakata, Y.; et al. Calpain-dependent cleavage of N-cadherin is involved in the progression of post-myocardial infarction remodeling. J. Biol. Chem. 2014, 289, 19408–19419. [Google Scholar] [CrossRef] [Green Version]
  51. Verselis, V.K.; Trexler, E.B.; Bukauskas, F.F. Connexin hemichannels and cell-cell channels: Comparison of properties. Braz. J. Med. Biol. Res. 2000, 33, 379–389. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Alizadeh, H.; Davoodi, J.; Rafii-Tabar, H. Deconstruction of the human connexin 26 hemichannel due to an applied electric field; A molecular dynamics simulation study. J. Mol. Graph. Model. 2017, 73, 108–114. [Google Scholar] [CrossRef] [PubMed]
  53. Sáez, J.C.; Retamal, M.A.; Basilio, D.; Bukauskas, F.F.; Bennett, M.V.L. Connexin-based gap junction hemichannels: Gating mechanisms. Biochim. Biophys. Acta-Biomembr. 2005, 1711, 215–224. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Verselis, V.K.; Srinivas, M. Divalent cations regulate connexin hemichannels by modulating intrinsic voltage-dependent gating. J. Gen. Physiol. 2008, 132, 315–327. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Gomez-Hernandez, J.M.; de Miguel, M.; Larrosa, B.; Gonzalez, D.; Barrio, L.C. Molecular basis of calcium regulation in connexin-32 hemichannels. Proc. Natl. Acad. Sci. USA 2003, 100, 16030–16035. [Google Scholar] [CrossRef] [Green Version]
  56. Contreras, J.E.; Saez, J.C.; Bukauskas, F.F.; Bennett, M.V.L. Gating and regulation of connexin 43 (Cx43) hemichannels. Proc. Natl. Acad. Sci. USA 2003, 100, 11388–11393. [Google Scholar] [CrossRef] [Green Version]
  57. Stout, C.; Charles, A. Modulation of intercellular calcium signaling in astrocytes by extracellular calcium and magnesium. Glia 2003, 43, 265–273. [Google Scholar] [CrossRef]
  58. Ye, Z.-C.; Wyeth, M.S.; Baltan-Tekkok, S.; Ransom, B.R. Functional hemichannels in astrocytes: A novel mechanism of glutamate release. J. Neurosci. 2003, 23, 3588–3596. [Google Scholar] [CrossRef] [Green Version]
  59. Rana, S.; Dringen, R. Gap junction hemichannel-mediated release of glutathione from cultured rat astrocytes. Neurosci. Lett. 2007, 415, 45–48. [Google Scholar] [CrossRef]
  60. Karagiannis, A.; Sylantyev, S.; Hadjihambi, A.; Hosford, P.S.; Kasparov, S.; Gourine, A. V Hemichannel-mediated release of lactate. J. Cereb. Blood Flow Metab. 2016, 36, 1202–1211. [Google Scholar] [CrossRef] [Green Version]
  61. De Flora, A.; Zocchi, E.; Guida, L.; Franco, L.; Bruzzone, S. Autocrine and paracrine calcium signaling by the CD38/NAD+/cyclic ADP-ribose system. Ann. N. Y. Acad. Sci. 2004, 1028, 176–191. [Google Scholar] [PubMed]
  62. Retamal, M.A.; Froger, N.; Palacios-Prado, N.; Ezan, P.; Sáez, P.J.; Sáez, J.C.; Giaume, C. Cx43 hemichannels and gap junction channels in astrocytes are regulated oppositely by proinflammatory cytokines released from activated microglia. J. Neurosci. 2007, 27, 13781–13792. [Google Scholar] [CrossRef] [PubMed]
  63. Kondo, R.P.; Wang, S.-Y.; John, S.A.; Weiss, J.N.; Goldhaber, J.I. Metabolic Inhibition Activates a Non-selective Current Through Connexin Hemichannels in Isolated Ventricular Myocytes. J. Mol. Cell. Cardiol. 2000, 32, 1859–1872. [Google Scholar] [CrossRef]
  64. Schalper, K.A.; Sánchez, H.A.; Lee, S.C.; Altenberg, G.A.; Nathanson, M.H.; Sáez, J.C. Connexin 43 hemichannels mediate the Ca 2+ influx induced by extracellular alkalinization. Am. J. Physiol. Physiol. 2010, 299, C1504–C1515. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Sánchez, H.A.; Meşe, G.; Srinivas, M.; White, T.W.; Verselis, V.K. Differentially altered Ca2+ regulation and Ca2+ permeability in Cx26 hemichannels formed by the A40V and G45E mutations that cause keratitis ichthyosis deafness syndrome. J. Gen. Physiol. 2010, 136, 47–62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Vargas, A.A.; Cisterna, B.A.; Saavedra-Leiva, F.; Urrutia, C.; Cea, L.A.; Vielma, A.H.; Gutierrez-Maldonado, S.E.; Martin, A.J.M.; Pareja-Barrueto, C.; Escalona, Y.; et al. On Biophysical Properties and Sensitivity to Gap Junction Blockers of Connexin 39 Hemichannels Expressed in HeLa Cells. Front. Physiol. 2017, 8, 38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Hansen, D.B.; Braunstein, T.H.; Nielsen, M.S.; MacAulay, N. Distinct permeation profiles of the connexin 30 and 43 hemichannels. FEBS Lett. 2014, 588, 1446–1457. [Google Scholar] [CrossRef]
  68. Retamal, M.A.; Reyes, E.P.; García, I.E.; Pinto, B.; Martínez, A.D.; González, C. Diseases associated with leaky hemichannels. Front. Cell. Neurosci. 2015, 9, 267. [Google Scholar] [CrossRef] [Green Version]
  69. Pinto, B.I.; García, I.E.; Pupo, A.; Retamal, M.A.; Martínez, A.D.; Latorre, R.; González, C. Charged residues at the first transmembrane region contribute to the voltage dependence of the slow gate of connexins. J. Biol. Chem. 2016, 291, 15740–15752. [Google Scholar] [CrossRef] [Green Version]
  70. Retamal, M.A.; García, I.E.; Pinto, B.I.; Pupo, A.; Báez, D.; Stehberg, J.; Del Rio, R.; González, C. Extracellular cysteine in connexins: Role as redox sensors. Front. Physiol. 2016, 7, 1. [Google Scholar] [CrossRef] [Green Version]
  71. Retamal, M.A.; Schalper, K.A.; Shoji, K.F.; Bennett, M.V.L.; Sáez, J.C. Opening of connexin 43 hemichannels is increased by lowering intracellular redox potential. Proc. Natl. Acad. Sci. USA 2007, 104, 8322–8327. [Google Scholar] [CrossRef] [Green Version]
  72. Retamal, M.A.; Evangelista-Martínez, F.; León-Paravic, C.G.; Altenberg, G.A.; Reuss, L. Biphasic effect of linoleic acid on connexin 46 hemichannels. Pflugers Arch. Eur. J. Physiol. 2011, 461, 635–643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Orellana, J.A. Physiological Functions of Glial Cell Hemichannels. In Advances in Experimental Medicine and Biology; Springer: Cham, Switzerland, 2016; Volume 949, pp. 93–108. [Google Scholar]
  74. Stehberg, J.; Moraga-Amaro, R.; Salazar, C.; Becerra, A.; Echeverría, C.; Orellana, J.A.; Bultynck, G.; Ponsaerts, R.; Leybaert, L.; Simon, F.; et al. Release of gliotransmitters through astroglial connexin 43 hemichannels is necessary for fear memory consolidation in the basolateral amygdala. FASEB J. 2012, 26, 3649–3657. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Meunier, C.; Wang, N.; Yi, C.; Dallerac, G.; Ezan, P.; Koulakoff, A.; Leybaert, L.; Giaume, C. Contribution of Astroglial Cx43 Hemichannels to the Modulation of Glutamatergic Currents by D-Serine in the Mouse Prefrontal Cortex. J. Neurosci. 2017, 37, 9064–9075. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Chever, O.; Lee, C.-Y.; Rouach, N. Astroglial connexin43 hemichannels tune basal excitatory synaptic transmission. J. Neurosci. 2014, 34, 11228–11232. [Google Scholar] [CrossRef] [Green Version]
  77. Patrick Gonzalez, J.; Ramachandran, J.; Xie, L.H.; Contreras, J.E.; Fraidenraich, D. Selective Connexin43 Inhibition Prevents Isoproterenol-Induced Arrhythmias and Lethality in Muscular Dystrophy Mice. Sci. Rep. 2015, 5, 13490. [Google Scholar] [CrossRef]
  78. Taki, T.; Takeichi, T.; Sugiura, K.; Akiyama, M. Roles of aberrant hemichannel activities due to mutant connexin26 in the pathogenesis of KID syndrome. Sci. Rep. 2018, 8, 12824. [Google Scholar] [CrossRef] [Green Version]
  79. García, I.E.; Bosen, F.; Mujica, P.; Pupo, A.; Flores-Muñoz, C.; Jara, O.; González, C.; Willecke, K.; Martínez, A.D. From Hyperactive Connexin26 Hemichannels to Impairments in Epidermal Calcium Gradient and Permeability Barrier in the Keratitis-Ichthyosis-Deafness Syndrome. J. Invest. Dermatol. 2016, 136, 574–583. [Google Scholar] [CrossRef] [Green Version]
  80. Stong, B.C.; Chang, Q.; Ahmad, S.; Lin, X. A Novel Mechanism for Connexin 26 Mutation Linked Deafness: Cell Death Caused by Leaky Gap Junction Hemichannels. Laryngoscope 2006, 116, 2205–2210. [Google Scholar] [CrossRef]
  81. Sanchez, H.A.; Verselis, V.K. Aberrant Cx26 hemichannels and keratitis-ichthyosis-deafness syndrome: Insights into syndromic hearing loss. Front. Cell. Neurosci. 2014, 8, 354. [Google Scholar] [CrossRef] [Green Version]
  82. Liang, G.S.L.; de Miguel, M.; Gómez-Hernández, J.M.; Glass, J.D.; Scherer, S.S.; Mintz, M.; Barrio, L.C.; Fischbeck, K.H. Severe neuropathy with leaky connexin32 hemichannels. Ann. Neurol. 2005, 57, 749–754. [Google Scholar] [CrossRef] [PubMed]
  83. León-Paravic, C.G.; Figueroa, V.A.; Guzmán, D.J.; Valderrama, C.F.; Vallejos, A.A.; Fiori, M.C.; Altenberg, G.A.; Reuss, L.; Retamal, M.A. Carbon monoxide (CO) is a novel inhibitor of connexin hemichannels. J. Biol. Chem. 2014, 289, 36150–36157. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Retamal, M.A. Carbon Monoxide Modulates Connexin Function through a Lipid Peroxidation-Dependent Process: A Hypothesis. Front. Physiol. 2016, 7, 259. [Google Scholar] [CrossRef] [PubMed]
  85. Soetkamp, D.; Nguyen, T.T.; Menazza, S.; Hirschhäuser, C.; Hendgen-Cotta, U.B.; Rassaf, T.; Schlüter, K.D.; Boengler, K.; Murphy, E.; Schulz, R. S-nitrosation of mitochondrial connexin 43 regulates mitochondrial function. Basic Res. Cardiol. 2014, 109, 433. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Retamal, M.A.; Fiori, M.C.; Fernandez-Olivares, A.; Linsambarth, S.; Peña, F.; Quintana, D.; Stehberg, J.; Altenberg, G.A. 4-Hydroxynonenal induces Cx46 hemichannel inhibition through its carbonylation. Biochim. Biophys. Acta-Mol. Cell Biol. Lipids 2020, 1865, 158705. [Google Scholar] [CrossRef]
  87. Retamal, M.A.; Cortés, C.J.; Reuss, L.; Bennett, M.V.L.; Sáez, J.C. S-nitrosylation and permeation through connexin 43 hemichannels in astrocytes: Induction by oxidant stress and reversal by reducing agents. Proc. Natl. Acad. Sci. USA 2006, 103, 4475–4480. [Google Scholar] [CrossRef] [Green Version]
  88. Retamal, M.A.; Yin, S.; Altenberg, G.A.; Reuss, L. Modulation of Cx46 hemichannels by nitric oxide. Am. J. Physiol.-Cell Physiol. 2009, 296, C1356–C1363. [Google Scholar] [CrossRef] [Green Version]
  89. Zhang, Y.; Hao, H. Conserved glycine at position 45 of major cochlear connexins constitutes a vital component of the Ca2+ sensor for gating of gap junction hemichannels. Biochem. Biophys. Res. Commun. 2013, 436, 424–429. [Google Scholar] [CrossRef]
  90. Retamal, M.A.; Orellana, V.P.; Arévalo, N.J.; Rojas, C.G.; Arjona, R.J.; Alcaíno, C.A.; González, W.; Canan, J.G.; Moraga-Amaro, R.; Stehberg, J.; et al. Cx46 hemichannel modulation by nitric oxide: Role of the fourth transmembrane helix cysteine and its possible involvement in cataract formation. Nitric Oxide 2019, 86, 54–62. [Google Scholar] [CrossRef]
  91. Suchyna, T.M.; Nitsche, J.M.; Chilton, M.; Harris, A.L.; Veenstra, R.D.; Nicholson, B.J. Different Ionic Selectivities for Connexins 26 and 32 Produce Rectifying Gap Junction Channels. Biophys. J. 1999, 77, 2968–2987. [Google Scholar] [CrossRef] [Green Version]
  92. Saez, J.C.; Connor, J.A.; Spray, D.C.; Bennett, M.V. Hepatocyte gap junctions are permeable to the second messenger, inositol 1,4,5-trisphosphate, and to calcium ions. Proc. Natl. Acad. Sci. USA 1989, 86, 2708–2712. [Google Scholar] [CrossRef] [Green Version]
  93. Bevans, C.G.; Kordel, M.; Rhee, S.K.; Harris, A.L. Isoform Composition of Connexin Channels Determines Selectivity among Second Messengers and Uncharged Molecules. J. Biol. Chem. 1998, 273, 2808–2816. [Google Scholar] [CrossRef] [Green Version]
  94. Kam, Y.; Kim, D.Y.; Koo, S.K.; Joe, C.O. Transfer of second messengers through gap junction connexin 43 channels reconstituted in liposomes. Biochim. Biophys. Acta-Biomembr. 1998, 1372, 384–388. [Google Scholar] [CrossRef] [Green Version]
  95. Chen, Q.; Boire, A.; Jin, X.; Valiente, M.; Er, E.E.; Lopez-Soto, A.S.; Jacob, L.; Patwa, R.; Shah, H.; Xu, K.; et al. Carcinoma–astrocyte gap junctions promote brain metastasis by cGAMP transfer. Nature 2016, 533, 493–498. [Google Scholar] [CrossRef] [PubMed]
  96. Harris, A.L. Connexin channel permeability to cytoplasmic molecules. Prog. Biophys. Mol. Biol. 2007, 94, 120–143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Mayan, M.D.; Gago-Fuentes, R.; Carpintero-Fernandez, P.; Fernandez-Puente, P.; Filgueira-Fernandez, P.; Goyanes, N.; Valiunas, V.; Brink, P.R.; Goldberg, G.S.; Blanco, F.J. Articular chondrocyte network mediated by gap junctions: Role in metabolic cartilage homeostasis. Ann. Rheum. Dis. 2015, 74, 275–284. [Google Scholar] [CrossRef] [Green Version]
  98. Wang, Q.; Chi, M.M.; Schedl, T.; Moley, K.H. An intercellular pathway for glucose transport into mouse oocytes. Am. J. Physiol. Metab. 2012, 302, E1511–E1518. [Google Scholar]
  99. Neijssen, J.; Herberts, C.; Drijfhout, J.W.; Reits, E.; Janssen, L.; Neefjes, J. Cross-presentation by intercellular peptide transfer through gap junctions. Nature 2005, 434, 83–88. [Google Scholar] [CrossRef]
  100. Pang, B.; Neijssen, J.; Qiao, X.; Janssen, L.; Janssen, H.; Lippuner, C.; Neefjes, J. Direct Antigen Presentation and Gap Junction Mediated Cross-Presentation during Apoptosis. J. Immunol. 2009, 183, 1083–1090. [Google Scholar] [CrossRef] [Green Version]
  101. Thuringer, D.; Jego, G.; Berthenet, K.; Hammann, A.; Solary, E.; Garrido, C. Gap junction-mediated transfer of miR-145-5p from microvascular endothelial cells to colon cancer cells inhibits angiogenesis. Oncotarget 2016, 7, 28160–28168. [Google Scholar] [CrossRef] [Green Version]
  102. Hong, X.; Sin, W.C.; Harris, A.L.; Naus, C.C. Gap junctions modulate glioma invasion by direct transfer of microRNA. Oncotarget 2015, 6, 15566–15577. [Google Scholar] [CrossRef] [Green Version]
  103. De Maio, A.; Gingalewski, C.; Theodorakis, N.G.; Clemens, M.G. Interruption of hepatic gap junctional communication in the rat during inflammation induced by bacterial lipopolysaccharide. Shock 2000, 14, 53–59. [Google Scholar] [CrossRef]
  104. Chanson, M.; Berclaz, P.-Y.; Scerri, I.; Dudez, T.; Wernke-Dollries, K.; Pizurki, L.; Pavirani, A.; Fiedler, M.A.; Suter, S. Regulation of Gap Junctional Communication by a Pro-Inflammatory Cytokine in Cystic Fibrosis Transmembrane Conductance Regulator-Expressing but Not Cystic Fibrosis Airway Cells. Am. J. Pathol. 2001, 158, 1775–1784. [Google Scholar] [CrossRef] [Green Version]
  105. Johnson, L.N.; Koval, M. Cross-Talk Between Pulmonary Injury, Oxidant Stress, and Gap Junctional Communication. Antioxid. Redox Signal. 2009, 11, 355–367. [Google Scholar] [CrossRef] [Green Version]
  106. Lin, J.H.; Weigel, H.; Cotrina, M.L.; Liu, S.; Bueno, E.; Hansen, A.J.; Hansen, T.W.; Goldman, S.; Nedergaard, M. Gap-junction-mediated propagation and amplification of cell injury. Nat. Neurosci. 1998, 1, 494–500. [Google Scholar] [CrossRef]
  107. Frantseva, M.V.; Kokarovtseva, L.; Velazquez, J.L.P. Ischemia-Induced Brain Damage Depends on Specific Gap-Junctional Coupling. J. Cereb. Blood Flow Metab. 2002, 22, 453–462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Talhouk, R.S.; Zeinieh, M.P.; Mikati, M.A.; El-Sabban, M.E. Gap junctional intercellular communication in hypoxia–ischemia-induced neuronal injury. Prog. Neurobiol. 2008, 84, 57–76. [Google Scholar] [CrossRef]
  109. Contreras, J.E.; Sánchez, H.A.; Eugenin, E.A.; Speidel, D.; Theis, M.; Willecke, K.; Bukauskas, F.F.; Bennett, M.V.L.; Sáez, J.C. Metabolic inhibition induces opening of unapposed connexin 43 gap junction hemichannels and reduces gap junctional communication in cortical astrocytes in culture. Proc. Natl. Acad. Sci. USA 2002, 99, 495–500. [Google Scholar] [CrossRef] [Green Version]
  110. Kang, K.S.; Kang, B.C.; Lee, B.J.; Che, J.H.; Li, G.X.; Trosko, J.E.; Lee, Y.S. Preventive effect of epicatechin and ginsenoside Rb2 on the inhibition of gap junctional intercellular communication by TPA and H2O2. Cancer Lett. 2000, 152, 97–106. [Google Scholar] [CrossRef]
  111. Figueroa, X.F.; Duling, B.R. Gap junctions in the control of vascular function. Antioxid. Redox Signal. 2009, 11, 251–266. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Pohl, U. Connexins: Key Players in the Control of Vascular Plasticity and Function. Physiol. Rev. 2020, 100, 525–572. [Google Scholar] [CrossRef] [PubMed]
  113. Hong, T.; Hill, C.E. Restricted expression of the gap junctional protein connexin 43 in the arterial system of the rat. J. Anat. 1998, 192 Pt 4, 583–593. [Google Scholar] [CrossRef]
  114. Kim, N.K.; Santos-Miranda, A.; Chen, H.; Aoyama, H.; Bai, D. Heterotypic docking compatibility of human connexin37 with other vascular connexins. J. Mol. Cell. Cardiol. 2019, 127, 194–203. [Google Scholar] [CrossRef] [PubMed]
  115. Reed, K.E.; Westphale, E.M.; Larson, D.M.; Wang, H.Z.; Veenstra, R.D.; Beyer, E.C. Molecular cloning and functional expression of human connexin37, an endothelial cell gap junction protein. J. Clin. Investig. 1993, 91, 997–1004. [Google Scholar] [CrossRef] [Green Version]
  116. Little, T.L.; Beyer, E.C.; Duling, B.R. Connexin 43 and connexin 40 gap junctional proteins are present in arteriolar smooth muscle and endothelium in vivo. Am. J. Physiol. Circ. Physiol. 2017, 268, H729–H739. [Google Scholar] [CrossRef] [PubMed]
  117. Beyer, E.C.; Reed, K.E.; Westphale, E.M.; Kanter, H.L.; Larson, D.M. Molecular cloning and expression of rat connexin40, a gap junction protein expressed in vascular smooth muscle. J. Membr. Biol. 1992, 127, 69–76. [Google Scholar] [CrossRef]
  118. Schmidt, V.J.; Jobs, A.; von Maltzahn, J.; Wörsdörfer, P.; Willecke, K.; de Wit, C. Connexin45 is expressed in vascular smooth muscle but its function remains elusive. PLoS ONE 2012, 7, e42287. [Google Scholar] [CrossRef] [Green Version]
  119. Tulenko, T.N. Regulating cross-talk between vascular smooth muscle cells. Arterioscler. Thromb. Vasc. Biol. 2003, 23, 1707–1709. [Google Scholar] [CrossRef] [Green Version]
  120. Christ, G.J.; Spray, D.C.; El-Sabban, M.; Moore, L.K.; Brink, P.R. Gap junctions in vascular tissues: Evaluating the role of intercellular communication in the modulation of vasomotor tone. Circ. Res. 1996, 79, 631–646. [Google Scholar] [CrossRef]
  121. Triggle, C.R.; Samuel, S.M.; Ravishankar, S.; Marei, I.; Arunachalam, G.; Ding, H. The endothelium: Influencing vascular smooth muscle in many ways. Can. J. Physiol. Pharmacol. 2012, 90, 713–738. [Google Scholar] [CrossRef]
  122. Beny, J.L.; Pacicca, C. Bidirectional electrical communication between smooth muscle and endothelial cells in the pig coronary artery. Am. J. Physiol. Circ. Physiol. 2017, 266, H1465–H1472. [Google Scholar] [CrossRef] [PubMed]
  123. Emerson, G.G.; Segal, S.S. Electrical coupling between endothelial cells and smooth muscle cells in hamster feed arteries: Role in vasomotor control. Circ. Res. 2000, 87, 474–479. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Earley, S.; Resta, T.C.; Walker, B.R. Disruption of smooth muscle gap junctions attenuates myogenic vasoconstriction of mesenteric resistance arteries. Am. J. Physiol. Circ. Physiol. 2004, 287, H2677–H2686. [Google Scholar] [CrossRef] [Green Version]
  125. Pogoda, K.; Mannell, H.; Blodow, S.; Schneider, H.; Schubert, K.M.; Qiu, J.; Schmidt, A.; Imhof, A.; Beck, H.; Tanase, L.I.; et al. NO augments endothelial reactivity by reducing myoendothelial calcium signal spreading: A novel role for C×37 (Connexin 37) and the protein tyrosine phosphatase SHP-2. Arterioscler. Thromb. Vasc. Biol. 2017, 37, 2280–2290. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Dora, K.A.; Sandow, S.L.; Gallagher, N.T.; Takano, H.; Rummery, N.M.; Hill, C.E.; Garland, C.J. Myoendothelial gap junctions may provide the pathway for EDHF in mouse mesenteric artery. J. Vasc. Res. 2003, 40, 480–490. [Google Scholar] [CrossRef] [PubMed]
  127. Sandow, S.L.; Hill, C.E. Incidence of myoendothelial gap junctions in the proximal and distal mesenteric arteries of the rat is suggestive of a role in endothelium-derived hyperpolarizing factor-mediated responses. Circ. Res. 2000, 86, 341–346. [Google Scholar] [CrossRef] [Green Version]
  128. Harris, D.; Martin, P.E.M.; Evans, W.H.; Kendall, D.A.; Griffith, T.M.; Randall, M.D. Role of gap junctions in endothelium-derived hyperpolarizing factor responses and mechanisms of K+-relaxation. Eur. J. Pharmacol. 2000, 402, 119–128. [Google Scholar] [CrossRef]
  129. Dora, K.A.; Doyle, M.P.; Duling, B.R. Elevation of intracellular calcium in smooth muscle causes endothelial cell generation of NO in arterioles. Proc. Natl. Acad. Sci. USA 1997, 94, 6529–6534. [Google Scholar] [CrossRef] [Green Version]
  130. Ignarro, L.J.; Buga, G.M.; Wood, K.S.; Byrns, R.E.; Chaudhuri, G. Endothelium-derived relaxing factor produced and released from artery and vein is nitric oxide. Proc. Natl. Acad. Sci. USA 1987, 84, 9265–9269. [Google Scholar] [CrossRef] [Green Version]
  131. Loscher, T.F. Endothelium-derived nitric oxide: The endogenous nitrovasodilator in the human cardiovascular system. Eur. Heart J. 1991, 12, 2–11. [Google Scholar] [CrossRef] [Green Version]
  132. Figueroa, X.F.; Lillo, M.A.; Gaete, P.S.; Riquelme, M.A.; Sáez, J.C. Diffusion of nitric oxide across cell membranes of the vascular wall requires specific connexin-based channels. Neuropharmacology 2013, 75, 471–478. [Google Scholar] [CrossRef] [PubMed]
  133. Pfenniger, A.; Derouette, J.P.; Verma, V.; Lin, X.; Foglia, B.; Coombs, W.; Roth, I.; Satta, N.; Dunoyer-Geindre, S.; Sorgen, P.; et al. Gap junction protein Cx37 interacts with endothelial nitric oxide synthase in endothelial cells. Arterioscler. Thromb. Vasc. Biol. 2010, 30, 827–834. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Alonso, F.; Boittin, F.X.; Bény, J.L.; Haefliger, J.A. Loss of connexin40 is associated with decreased endothelium-dependent relaxations and eNOS levels in the mouse aorta. Am. J. Physiol.-Hear. Circ. Physiol. 2010, 299, H1365–H1373. [Google Scholar] [CrossRef] [Green Version]
  135. Cronier, L. Connexin expression and gap junctional intercellular communication in human first trimester trophoblast. Mol. Hum. Reprod. 2002, 8, 1005–1013. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Nishimura, T.; Dunk, C.; Lu, Y.; Feng, X.; Gellhaus, A.; Winterhager, E.; Rossant, J.; Lye, S.J. Gap junctions are required for trophoblast proliferation in early human placental development. Placenta 2004, 25, 595–607. [Google Scholar] [CrossRef] [PubMed]
  137. Lang, I.; Schweizer, A.; Hiden, U.; Ghaffari-Tabrizi, N.; Hagendorfer, G.; Bilban, M.; Pabst, M.A.; Korgun, E.T.; Dohr, G.; Desoye, G. Human fetal placental endothelial cells have a mature arterial and a juvenile venous phenotype with adipogenic and osteogenic differentiation potential. Differentiation 2008, 76, 1031–1043. [Google Scholar] [CrossRef]
  138. Malassiné, A.; Frendo, J.L.; Evain-Brion, D. Trisomy 21- Affected placentas highlight prerequisite factors for human trophoblast fusion and differentiation. Int. J. Dev. Biol. 2010, 54, 475–482. [Google Scholar] [CrossRef]
  139. Cronier, L.; Bastide, B.; Defamie, N.; Niger, C.; Pointis, G.; Gasc, J.M.; Malassiné, A. Involvement of gap junctional communication and connexin expression in trophoblast differentiation of the human placenta. Histol. Histopathol. 2001, 16, 285–295. [Google Scholar]
  140. Malassiné, A.; Cronier, L. Involvement of gap junctions in placental functions and development. Biochim. Biophys. Acta-Biomembr. 2005, 1719, 117–124. [Google Scholar] [CrossRef] [Green Version]
  141. Al-Lamki, R.S.; Skepper, J.N.; Burton, G.J. Are human placental bed giant cells merely aggregates of small mononuclear trophoblast cells? An ultrastructural and immunocytochemical study. Hum. Reprod. 1999, 14, 496–504. [Google Scholar] [CrossRef]
  142. Frendo, J.L.; Cronier, L.; Bertin, G.; Guibourdenche, J.; Vidaud, M.; Evain-Brion, D.; Malassiné, A. Involvement of connexin 43 in human trophoblast cell fusion and differentiation. J. Cell Sci. 2003, 116, 3413–3421. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Winterhager, E.; Kidder, G.M. Gap junction connexins in female reproductive organs: Implications for women’s reproductive health. Hum. Reprod. Update 2015, 21, 340–352. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Winterhager, E.; Von Ostau, C.; Gerke, M.; Gruemmer, R.; Traub, O.; Kaufmann, P. Connexin expression patterns in human trophoblast cells during placental development. Placenta 1999, 20, 627–638. [Google Scholar] [CrossRef] [PubMed]
  145. McKinnon, R.L.; Bolon, M.L.; Wang, H.X.; Swarbreck, S.; Kidder, G.M.; Simon, A.M.; Tyml, K. Reduction of electrical coupling between microvascular endothelial cells by NO depends on connexin37. Am. J. Physiol.-Hear. Circ. Physiol. 2009, 297, H93–H101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Kameritsch, P.; Khandoga, N.; Nagel, W.; Hundhausen, C.; Lidington, D.; Pohl, U. Nitric oxide specifically reduces the permeability of Cx37-containing gap junctions to small molecules. J. Cell. Physiol. 2005, 203, 233–242. [Google Scholar] [CrossRef]
  147. Meens, M.J.; Alonso, F.; Le Gal, L.; Kwak, B.R.; Haefliger, J.A. Endothelial Connexin37 and Connexin40 participate in basal but not agonist-induced NO release. Cell Commun. Signal. 2015, 13, 1–11. [Google Scholar] [CrossRef] [Green Version]
  148. Avila, M.A.; Sell, S.L.; Hawkins, B.E.; Hellmich, H.L.; Boone, D.R.; Crookshanks, J.M.; Prough, D.S.; Dewitt, D.S. Cerebrovascular connexin expression: Effects of traumatic brain injury. J. Neurotrauma 2011, 28, 1803–1811. [Google Scholar] [CrossRef] [Green Version]
  149. Chen, W.; Guo, Y.; Yang, W.; Zheng, P.; Zeng, J.; Tong, W. Connexin40 correlates with oxidative stress in brains of traumatic brain injury rats. Restor. Neurol. Neurosci. 2017, 35, 217–224. [Google Scholar] [CrossRef]
  150. Le Gal, L.; Alonso, F.; Mazzolai, L.; Meda, P.; Haefliger, J.A. Interplay between Connexin40 and nitric oxide signaling during hypertension. Hypertension 2015, 65, 910–915. [Google Scholar] [CrossRef] [Green Version]
  151. De Wit, C.; Roos, F.; Bolz, S.S.; Pohl, U. Lack of vascular connexin 40 is associated with hypertension and irregular arteriolar vasomotion. Physiol. Genomics 2003, 13, 169–177. [Google Scholar] [CrossRef] [Green Version]
  152. Kameritsch, P.; Hoffmann, A.; Pohl, U. Opposing effects of nitric oxide on different connexins expressed in the vascular system. Cell Commun. Adhes. 2003, 10, 305–309. [Google Scholar] [CrossRef] [PubMed]
  153. Hoffmann, A.; Gloe, T.; Pohl, U.; Zahler, S. Nitric oxide enhances de novo formation of endothelial gap junctions. Cardiovasc. Res. 2003, 60, 421–430. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Yi, F.-X.; Boeldt, D.S.; Gifford, S.M.; Sullivan, J.A.; Grummer, M.A.; Magness, R.R.; Bird, I.M. Pregnancy Enhances Sustained Ca2+ Bursts and Endothelial Nitric Oxide Synthase Activation in Ovine Uterine Artery Endothelial Cells Through Increased Connexin 43 Function1. Biol. Reprod. 2010, 82, 66–75. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Dora, K.A.; Martin, P.E.M.; Chaytor, A.T.; Evans, W.H.; Garland, C.J.; Griffith, T.M. Role of heterocellular gap junctional communication in endothelium-dependent smooth muscle hyperpolarization: Inhibition by a connexin-mimetic peptide. Biochem. Biophys. Res. Commun. 1999, 254, 27–31. [Google Scholar] [CrossRef]
  156. Wang, N.; De Bock, M.; Antoons, G.; Gadicherla, A.K.; Bol, M.; Decrock, E.; Evans, W.H.; Sipido, K.R.; Bukauskas, F.F.; Leybaert, L. Connexin mimetic peptides inhibit Cx43 hemichannel opening triggered by voltage and intracellular Ca2+ elevation. Basic Res. Cardiol. 2012, 107, 304. [Google Scholar] [CrossRef] [Green Version]
  157. Liang, J.; Chen, P.; Li, C.; Li, D.; Wang, J.; Xue, R.; Zhang, S.; Ruan, J.; Zhang, X. IL-22 Down-Regulates Cx43 Expression and Decreases Gap Junctional Intercellular Communication by Activating the JNK Pathway in Psoriasis. J. Investig. Dermatol. 2019, 139, 400–411. [Google Scholar] [CrossRef]
  158. Morioka, N.; Zhang, F.F.; Nakamura, Y.; Kitamura, T.; Hisaoka-Nakashima, K.; Nakata, Y. Tumor necrosis factor-mediated downregulation of spinal astrocytic connexin43 leads to increased glutamatergic neurotransmission and neuropathic pain in mice. Brain. Behav. Immun. 2015, 49, 293–310. [Google Scholar] [CrossRef]
  159. Kim, J.; Kim, Y.J.; Lee, R.; Moon, J.H.; Jo, I. Serum levels of zinc, calcium, and iron are associated with the risk of preeclampsia in pregnant women. Nutr. Res. 2012, 32, 764–769. [Google Scholar] [CrossRef]
  160. Kisters, K.; Barenbrock, M.; Louwen, F.; Hausberg, M.; Rahn, K.H.; Kosch, M. Membrane, intracellular, and plasma magnesium and calcium concentrations in preeclampsia. Am. J. Hypertens. 2000, 13, 765–769. [Google Scholar] [CrossRef] [Green Version]
  161. López-Jaramillo, P. Calcium, nitric oxide, and preeclampsia. Semin. Perinatol. 2000, 24, 33–36. [Google Scholar] [CrossRef]
  162. Sun, X.; Li, H.; He, X.; Li, M.; Yan, P.; Xun, Y.; Lu, C.; Yang, K.; Zhang, X. The association between calcium supplement and preeclampsia and gestational hypertension: A systematic review and meta-analysis of randomized trials. Hypertens. Pregnancy 2019, 38, 129–139. [Google Scholar] [CrossRef] [PubMed]
  163. Ebihara, L.; Liu, X.; Pal, J.D. Effect of External Magnesium and Calcium on Human Connexin46 Hemichannels. Biophys. J. 2003, 84, 277–286. [Google Scholar] [CrossRef] [Green Version]
  164. Stout, C.E.; Costantin, J.L.; Naus, C.C.G.; Charles, A.C. Intercellular Calcium Signaling in Astrocytes via ATP Release through Connexin Hemichannels. J. Biol. Chem. 2002, 277, 10482–10488. [Google Scholar] [CrossRef] [Green Version]
  165. Burnstock, G. Overview. Purinergic mechanisms. Ann. N. Y. Acad. Sci. 1990, 603, 1–17. [Google Scholar] [CrossRef] [PubMed]
  166. Yi, F.X.; Boeldt, D.S.; Magness, R.R.; Bird, I.M. [Ca2+]i signaling vs. eNOS expression as determinants of NO output in uterine artery endothelium: Relative roles in pregnancy adaptation and reversal by VEGF165. Am. J. Physiol.-Hear. Circ. Physiol. 2011, 300, H1182–H1193. [Google Scholar] [CrossRef] [Green Version]
  167. Krupp, J.; Boeldt, D.S.; Yi, F.X.; Grummer, M.A.; Anaya, H.A.B.; Shah, D.M.; Bird, I.M. The loss of sustained Ca2+ signaling underlies suppressed endothelial nitric oxide production in preeclamptic pregnancies: Implications for new therapy. Am. J. Physiol.-Hear. Circ. Physiol. 2013, 305, H969–H979. [Google Scholar] [CrossRef] [Green Version]
  168. Ali, A.; Bouma, G.J.; Anthony, R.V.; Winger, Q.A. The Role of LIN28-let-7-ARID3B Pathway in Placental Development. Int. J. Mol. Sci. 2020, 21, 3637. [Google Scholar] [CrossRef]
  169. Peng, Y.; Wang, X.; Guo, Y.; Peng, F.; Zheng, N.; He, B.; Ge, H.; Tao, L.; Wang, Q. Pattern of cell-to-cell transfer of microRNA by gap junction and its effect on the proliferation of glioma cells. Cancer Sci. 2019, 110, 1947–1958. [Google Scholar]
  170. Fan, X.; Teng, Y.; Ye, Z.; Zhou, Y.; Tan, W.S. The effect of gap junction-mediated transfer of miR-200b on osteogenesis and angiogenesis in a co-culture of MSCs and HUVECs. J. Cell Sci. 2018, 131. [Google Scholar] [CrossRef] [Green Version]
  171. Thuringer, D.; Boucher, J.; Jego, G.; Pernet, N.; Cronier, L.; Hammann, A.; Solary, E.; Garrido, C. Transfer of functional microRNAs between glioblastoma and microvascular endothelial cells through gap junctions. Oncotarget 2016, 7, 73925–73934. [Google Scholar] [CrossRef] [Green Version]
Figure 1. General characteristics of Cx and Cx-based channels. Panel (A) illustrates a representative topography of Cx at the plasma membrane. Connexin proteins are formed by four transmembrane domains, two extracellular loops (EL1 and EL2), one intracellular loop (IL), and both the N- and C-terminus are facing the cytoplasm. Panel (B) displays a hexamer of Cxs for a connexon or hemichannel. The channel presents a central pore through which, bidirectionally flow of ions and molecules up to ~1.2 kDa occurs. Panel (C) shows the formation of a gap junction channel (GJC) by serial docking of two hemichannels at the cell-to-cell contact. GJC allow the exchange of ion and molecules between the cytoplasm of neighboring cells.
Figure 1. General characteristics of Cx and Cx-based channels. Panel (A) illustrates a representative topography of Cx at the plasma membrane. Connexin proteins are formed by four transmembrane domains, two extracellular loops (EL1 and EL2), one intracellular loop (IL), and both the N- and C-terminus are facing the cytoplasm. Panel (B) displays a hexamer of Cxs for a connexon or hemichannel. The channel presents a central pore through which, bidirectionally flow of ions and molecules up to ~1.2 kDa occurs. Panel (C) shows the formation of a gap junction channel (GJC) by serial docking of two hemichannels at the cell-to-cell contact. GJC allow the exchange of ion and molecules between the cytoplasm of neighboring cells.
Ijms 21 04698 g001
Figure 2. (A) In a normal pregnancy, Cx43 via GJCs y/o hemichannels controls the fusion of cytotrophoblast, to form syncytiotrophoblast. Cx40 controls the rate of cell division and differentiation of extravillous cytotrophoblast. Cx37 and Cx40 present in the endovascular cytotrophoblast could modulate smooth muscle relaxation via releasing vasoactive molecules such as NO. (B) in PE, the reduction in the expression and/or functionality (denoted by a black X) of the Cx43 GJC and hemichannels mediated by posttranslational modifications induced by ROS/RNS. Potentially, it could alter the formation and/or functionality of syncytiotrophoblast. Alterations of Cx40-based channels could reduce invasive cytotrophoblast cells, and finally, the reduction in expression/functionality of Cx37 and Cx40-based channels in the vasculature could increase smooth muscle basal contraction.
Figure 2. (A) In a normal pregnancy, Cx43 via GJCs y/o hemichannels controls the fusion of cytotrophoblast, to form syncytiotrophoblast. Cx40 controls the rate of cell division and differentiation of extravillous cytotrophoblast. Cx37 and Cx40 present in the endovascular cytotrophoblast could modulate smooth muscle relaxation via releasing vasoactive molecules such as NO. (B) in PE, the reduction in the expression and/or functionality (denoted by a black X) of the Cx43 GJC and hemichannels mediated by posttranslational modifications induced by ROS/RNS. Potentially, it could alter the formation and/or functionality of syncytiotrophoblast. Alterations of Cx40-based channels could reduce invasive cytotrophoblast cells, and finally, the reduction in expression/functionality of Cx37 and Cx40-based channels in the vasculature could increase smooth muscle basal contraction.
Ijms 21 04698 g002
Figure 3. Reduction of Cx-based channel activity in preeclampsia. Normal pregnancy displays physiological NO bioavailability, with physiological hemichannels (HC) and gap junction channels (GJCs) activity that supports the exchange of NO, endothelium-derived hyperpolarization factor (EDHF), and other molecules between endothelial and smooth muscle cells. Consequently, blood flow to the placenta is adequate to satisfy its metabolic needs. However, NO bioavailability is reduced in preeclampsia due to increased free radicals. Hemichannels (HC) and gap junction channels (GJC) decrease their open probability and/or expression, reducing the flux of vasodilator molecules between endothelial and smooth muscle cells.
Figure 3. Reduction of Cx-based channel activity in preeclampsia. Normal pregnancy displays physiological NO bioavailability, with physiological hemichannels (HC) and gap junction channels (GJCs) activity that supports the exchange of NO, endothelium-derived hyperpolarization factor (EDHF), and other molecules between endothelial and smooth muscle cells. Consequently, blood flow to the placenta is adequate to satisfy its metabolic needs. However, NO bioavailability is reduced in preeclampsia due to increased free radicals. Hemichannels (HC) and gap junction channels (GJC) decrease their open probability and/or expression, reducing the flux of vasodilator molecules between endothelial and smooth muscle cells.
Ijms 21 04698 g003
Table 1. Expression and role of Cxs in human placenta. The localization and attributed functions of the Cxs expressed in placenta are listed. The assay applied for detection is also listed.
Table 1. Expression and role of Cxs in human placenta. The localization and attributed functions of the Cxs expressed in placenta are listed. The assay applied for detection is also listed.
Cx TypeLocalizationAssayAttributed FunctionReference
Cx32Placenta tissuePCRunknown[135]
cytotrophoblastic cultureInmmunolocalization[136]
[141]
Cx37Endothelial cells fromPCR unknown [136]
chorionic arteries
Cx40Endothelial cells from PCRCytotrophoblast proliferation[135]
placenta arteriolesIn situ Hybridization [136]
Inmmunolocalization [137]
EVT cells in allNorthern blotEpithelial like
anchoring columns trophoblast marker
Cx43Placenta tissuePCRFusion of cytotrophoblast to[136]
Northern blotform syncytiotrophoblast[137]
In situ Hybridization [139]
Inmmunolocalization [141]
[142]
Cx45Placenta tissue PCRunknown[136]
Inmmunolocalization [143]
[144]

Share and Cite

MDPI and ACS Style

Rozas-Villanueva, M.F.; Casanello, P.; Retamal, M.A. Role of ROS/RNS in Preeclampsia: Are Connexins the Missing Piece? Int. J. Mol. Sci. 2020, 21, 4698. https://doi.org/10.3390/ijms21134698

AMA Style

Rozas-Villanueva MF, Casanello P, Retamal MA. Role of ROS/RNS in Preeclampsia: Are Connexins the Missing Piece? International Journal of Molecular Sciences. 2020; 21(13):4698. https://doi.org/10.3390/ijms21134698

Chicago/Turabian Style

Rozas-Villanueva, María F., Paola Casanello, and Mauricio A. Retamal. 2020. "Role of ROS/RNS in Preeclampsia: Are Connexins the Missing Piece?" International Journal of Molecular Sciences 21, no. 13: 4698. https://doi.org/10.3390/ijms21134698

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

Article Metrics

Back to TopTop