Next Article in Journal
Dental Composites with Magnesium Doped Zinc Oxide Nanoparticles Prevent Secondary Caries in the Alloxan-Induced Diabetic Model
Next Article in Special Issue
A Mouse Systems Genetics Approach Reveals Common and Uncommon Genetic Modifiers of Hepatic Lysosomal Enzyme Activities and Glycosphingolipids
Previous Article in Journal
Effect of Overexpression of γ-Tocopherol Methyltransferase on α-Tocopherol and Fatty Acid Accumulation and Tolerance to Salt Stress during Seed Germination in Brassica napus L.
Previous Article in Special Issue
Aquaporin Gating: A New Twist to Unravel Permeation through Water Channels
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Hypertensive Nephropathy: Unveiling the Possible Involvement of Hemichannels and Pannexons

1
Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, El Llano Subercaseaux #2801, Santiago 8910060, Chile
2
Departamento de Neurología, Escuela de Medicina y Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
3
Programa de Comunicación Celular en Cáncer, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago 7610658, Chile
4
Centro de Fisiología Celular e Integrativa, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago 7610658, Chile
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(24), 15936; https://doi.org/10.3390/ijms232415936
Submission received: 19 October 2022 / Revised: 9 December 2022 / Accepted: 12 December 2022 / Published: 14 December 2022
(This article belongs to the Special Issue State-of-the-Art Molecular Biology in Chile)

Abstract

:
Hypertension is one of the most common risk factors for developing chronic cardiovascular diseases, including hypertensive nephropathy. Within the glomerulus, hypertension causes damage and activation of mesangial cells (MCs), eliciting the production of large amounts of vasoactive and proinflammatory agents. Accordingly, the activation of AT1 receptors by the vasoactive molecule angiotensin II (AngII) contributes to the pathogenesis of renal damage, which is mediated mostly by the dysfunction of intracellular Ca2+ ([Ca2+]i) signaling. Similarly, inflammation entails complex processes, where [Ca2+]i also play crucial roles. Deregulation of this second messenger increases cell damage and promotes fibrosis, reduces renal blood flow, and impairs the glomerular filtration barrier. In vertebrates, [Ca2+]i signaling depends, in part, on the activity of two families of large-pore channels: hemichannels and pannexons. Interestingly, the opening of these channels depends on [Ca2+]i signaling. In this review, we propose that the opening of channels formed by connexins and/or pannexins mediated by AngII induces the ATP release to the extracellular media, with the subsequent activation of purinergic receptors. This process could elicit Ca2+ overload and constitute a feed-forward mechanism, leading to kidney damage.

1. Introduction

1.1. Chronic Kidney Disease, General Aspects

The kidney is commonly described as an excretory organ and is essential to maintaining systemic homeostasis [1,2]. Kidney-mediated blood filtering and modification of the ultrafiltration content by reabsorption and secretion, maintains pH, water, and electrolyte balance, contributing to regulating blood pressure and osmolality. The final product of the kidneys is urine, and its volume and composition can provide insights into body health status [2]. As mentioned, the kidneys’ central role is to eliminate waste materials ingested or produced by metabolism and to control the fluid body volume and electrolyte composition [3]. However, this organ can fail and when that happens, several deregulations in many organ functions can emerge. In this regard, chronic kidney disease (CKD) is a global health problem, the prevalence of which has increased [4]. Around 10% of adults suffer kidney damage in developed countries [5,6]. Currently, the estimated global prevalence of CKD is 697.5 million cases, and patients with end-stage renal disease (ESRD) who need renal replacement therapy range between 4.902 and 7.083 million [7,8].
CKD embodies different etiologies, including diabetic nephropathy, hypertensive nephrosclerosis, and glomerulonephritis. However, regardless of the causes, some morphological changes are common, including tubular necrosis and glomerular sclerosis [9,10,11] (Figure 1). The pathogenesis of CKD begins with a disturbance of the glomerular and tubulointerstitial compartments due to the release of cytokines from the glomerulus. This local inflammatory impairs tubular epithelial cell functions, causing excessive protein filtration at the injured glomeruli [11,12]. The above is then accompanied by tubulointerstitial ischemia at the glomerular lesion and the hyperfunction of the remaining tubules, promoting leukocyte, cytotoxicity, and fibrogenesis recruitment [11,12] (Figure 1). Altogether, these events could lead to acute renal failure, characterized by the abrupt and transitory decrease in renal function, inability to excrete waste products, and maintain the electrolyte balance, as well as generalized nephron dysfunction [13]. In contrast, patients with CKD usually develop progressive kidney damage typified by glomerular sclerosis or tubulointerstitial fibrosis, which eventually leads to end-stage renal disease (ESRD), and the last stage of this condition causes irreversible damage [14,15]. This phenomenon includes the progressive reduction in the glomerular filtration rate (GFR), due to increased nephron damage, which eventually triggers organ failure [6,16,17].

1.2. Role of Renin–Angiotensin System in CKD Development

Based on the renin localization, mainly in granular cells of the renal afferent arteriole, it is assumed that swelling or shrinking of the juxtaglomerular apparatus (JGA) is involved in the glomerular blood flow ascribed to the renin–angiotensin system (RAS). These cells make contact with the extraglomerular mesangium and macula densa, forming the JGA [18]. The unique juxtaposition of the macula densa to the afferent arterioles suggests the existence of mechanisms linking tubular function with the control of arteriolar contraction [18]. RAS is present not only in the juxtaglomerular apparatus and interstitium, but also in various segments of the nephron. Renal tubular RAS was recognized when the highest known angiotensin concentration in the body was measured in tubular fluid [18,19]. Subsequently, it was shown that all components of RAS are present in vascular smooth muscle cells of afferent arterioles and glomerular podocytes, suggesting the involvement of the intracellular renin–angiotensin system in local regulatory systems [18,20]. Hypertension is one of the most common risk factors for developing coronary artery disease, stroke, heart failure, peripheral arterial disease, vision loss, dementia, and CKD. The prevalence of hypertension among adults will escalate to more than 1.56 billion by 2025 [12,21]. Hypertensive nephropathy linked to hypertension is the second cause of terminal CKD, and, nowadays, the cost of its treatment exceeds the budget of health systems in many countries [18,19]. The inflammatory injury and tissue remodeling evoked by CDK have been attributed to angiotensin II (Ang II), the principal peptide in RAS [22].
AngII regulates blood pressure, intravascular volume, and electrolyte balance by acting on the kidneys and adrenal glands [22]. Treatment with high concentrations of AngII induces the expression of chemokines, adhesion molecules, and pro-inflammatory cytokines (interleukin- 1β [IL-1β] and tumor necrosis factor-α [TNF-α]) [15,23,24]. The latter is accompanied by infiltration of macrophages (positive ED-1) [25], and tubular overexpression of osteopontin, a macrophage chemoattractant and adhesion molecule [25]. All these events are intimately associated with AngII-induced kidney damage linked to redox imbalance [26,27]. AngII increases the expression of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX), one of the essential enzymes in the generation of reactive oxygen species (ROS) [28,29]. Indeed, the impairment of redox balance plays a decisive role in several chronic diseases, including the progression of CKD [6,30] (Figure 2). Given that intrarenal RAS is critical for the pathophysiology of hypertensive nephropathy [6,31], the administration of AngII has been used for modeling kidney damage. In addition to providing a clinically relevant model for systemic hypertension, exposure to AngII leads to progressive and chronic nephron damage that recapitulates events occurring in ESRD [27,31,32]. Due to the diverse effects of RAS, RAS inhibitors (including blockers of angiotensin converting enzyme, angiotensin receptors, renin/prorenin receptors, and renin release) reduce blood pressure and increase renal blood flow and GFR, decreasing the progression of renal tissue damage [17].

1.3. Role of Mesangial Cells in CKD

The renal corpuscle comprises the Bowman’s capsule and the glomerulus, both being fundamental structures for blood filtration. The Bowman’s capsule plays a vital role in the structural and functional stability of the glomerulus, allowing it to fulfill its filtering function successfully [20,33]. Structurally speaking, the mesangium consists of a network of mesangial cells (MCs) that constitute between 30–40% of the glomerular cell populations. They control the capillary dilation, the regulation of the GFR, and the synthesis and degradation of extracellular matrix proteins [33]. Moreover, they have contractile and phagocytic properties [34]. Interestingly, cells from the renin lineage repopulate the niche of damaged glomerular MCs that have undergone mesangiolysis via the experimental model of mesangial proliferative glomerulonephritis. Still, the mechanism and molecules involved in the above phenomenon remain to be defined [35]. On the other hand, MCs stimulated with AngII produce ROS [14,36,37], synthesize and release IL-1β, TNF-α, and chemokines, such as the macrophage chemoattractant protein (MCP-1) and the transforming growth factor β1 (TGF-β1). These inflammatory mediators favor fibrosis, reducing renal blood flow, permeability, and eventually glomerular filtration [14,20,27,33,37,38,39]. Transcription factors NF-κB and AP-1 command these alterations and increase the synthesis and release of extracellular matrix proteins, such as type IV collagen, laminin, and fibronectin (Figure 2). The latter result in the formation of mesangial nodules and lesions at the interstitium, hindering the adequate function of the glomerulus [27,33,37,38,39]. As MCs undergo progressive damage at the glomerulus, they may serve as a potential target for understanding the pathogenesis of chronic nephron lesions and subsequent CKD [14,33,36,38]. In this review, we discuss how MCs could contribute to glomerular dysfunction following hypertensive nephropathy, focusing on the signaling pathways mediated by two families of plasma membrane channels: hemichannels and pannexons.

2. Connexins and Pannexins

Gap junctions (GJs) are aggregates of ten to thousands of individual intercellular channels that permit the cytoplasmic exchange between adjacent cells. They allow the diffusion of ions, small metabolites (e.g., nicotinamide adenine dinucleotide: NAD+, glucose, lactate, and glutamate), and second messengers (e.g., cyclic adenosine monophosphate [cAMP] and inositol trisphosphate [IP3]) between adjacent cells [40,41,42]. These connections support several biological processes in the animal kingdom, such as the propagation of electrical and chemical signals, which are crucial for cell coordination and survival. GJ channels are formed by integral membrane proteins called connexins, whose family has 21 isoforms in humans that are named according to their molecular weight [42,43]. All connexins feature four transmembrane domains, two extracellular loops, each with three highly conserved cysteine residues, an intracellular loop, and the C- and N-terminal ends on the cytoplasmic domain [42]. Six connexin monomers assemble around a central pore to form a hemichannel (HC) or connexons, whereas the apposition of two connexons from adjacent cells creates an intercellular GJ channel [42]. Cells expressing more than one type of connexin could eventually form heteromeric connexons. Likewise, cells expressing different connexons could form heterotypic GJs [42].
HCs act as free channels in the plasma membrane, favoring the exchange of small molecules and ions between the cytosol and the extracellular space [43,44] (Figure 3). Al-though initially they were catalogued as non-selective channels, nowadays, the evidence seems to indicate the opposite [45,46,47,48]. For example, Cx32 HCs show a slight preference for anions, while Cx40 and Cx43 have a high selectivity for cations [49,50,51]. In the same way, selectivity for ATP is 300-fold higher in Cx43 than in Cx32 HCs; for ADP, glutamate, and glutathione, the selectivity is 20-fold higher in Cx43 than in Cx32 HCs, while adenosine is 10-fold more permeable in Cx32 than Cx43 HCs [42,52]. The physiological opening of these channels is critical for the proper function of multiple biological processes, such as neurite outgrowth [53], neutrophil migration [54], and regulation of acid–base homeostasis in the kidney [55], among others. The HC opening is tightly regulated and relies on changes in voltage, intracellular/extracellular Ca2+ concentration ([Ca2+]i) [56], pH [57], post-translational modifications (e.g., phosphorylation [58] and S-nitrosylation [59]), and protein interactions [42]. Nevertheless, persistent activation of HCs has been associated with the genesis and progression of diverse pathological stages [60,61,62]. At least three mechanisms have linked the exacerbated opening of HCs with cell dysfunction and damage [63]. Firstly, the uncontrolled entry of Na+ and Cl through HCs may result in osmotic and ionic imbalances coupled to further cell swelling and plasma membrane breakdown [63,64,65]. Second, some HCs are permeable to Ca2+ [66,67], which could permit its influx to the cytosol during pathological conditions. The direct or indirect increase in [Ca2+]i mediated by HCs could lead to Ca2+ overload and consequent induction of different proteases, phospholipases, and other hydrolytic enzymes, as well as oxidative stress and caspase activation [68,69]. Finally, exacerbated HC opening may trigger the release of high amounts of potentially toxic molecules to neighboring cells, such as glutamate, in the case of the brain [63,70].
Pannexins (Panxs) belong to a family of transmembrane proteins that bear structural homology with innexins, the GJ protein family of invertebrates [71]. Although connexins and pannexins do not share significant sequence homology, both families exhibit similar transmembrane topology: four transmembrane domains with two extracellular loops and intracellular N- and C-terminals. Unlike connexins, which have several extracellular cysteine residues, pannexins have only two cysteines and a highly glycosylated aspartate in the second extracellular loop, which would hinder GJs formation (see below) [72,73] (Figure 3). Three pannexin isoforms have been identified to date: pannexin-1 (Panx1) (~48 kDa), is the most ubiquitous member of its family, broadly expressed in the brain, muscle, and other tissues [74]; pannexin-2 (Panx2) (~45 kDa), predominantly expressed in the brain; and pannexin-3 (Panx3) (~73 kDa), found in skin, bone, cartilage, skeletal muscle, and blood vessels [72,73,75,76]. Pannexin monomers oligomerize around a central pore in heptamers (Panx1, Panx3) or octamers (Panx2) called pannexons, allowing the exchange of ions and small molecules between the cytoplasm and the extracellular milieu [71,72,77]. Panx1 channels have been historically described as non-junctional membrane channels; nevertheless, recent evidence suggests that Panx1 can form GJs channels due to a differential glycosylated state at its extracellular loop [78]. Sequence analysis of Panx1 reveals that the Asp254 residue can be gradually glycosylated, which yields three different Panx1 bands in Western blot analysis: Gly-0 (non-glycosylated), Gly-1 (several mannose glycosylation), and Gly-2 (complex sugar glycosylation) [78]. Mutagenesis studies show that these glycosylations facilitate Panx1 trafficking but are not essential for the arrival of Panx1 to the plasma membrane or the function of a GJ channel. However, the Gly-2 variant of Panx1 is predominant on the surface [72,78].
Panx1 channels are associated with the regulation of vascular tone and flow [79], mucociliary clearance in the airway epithelial [80], phagocytosis of apoptotic cells [81], activation of the inflammasome, and recruitment of immune cells [82,83]. Similar to HCs, the persistent opening of Panx1 channels has been proposed to be harmful to cells, contributing to cell death in episodes of ischemia [84] or facilitating viral infection [85]. Panx1 channels have been extensively studied as crucial pathways for releasing ATP and pivotal players for purinergic receptor-mediated intracellular Ca2+ wave propagation [86,87,88]. Indeed, Panx1 channels release ATP at resting membrane potential and under physiological extracellular concentrations of divalent cations (Ca2+ and Mg2+) [73]. Furthermore, multiple direct mechanisms activate these channels, including mechanical stimulation [89], caspases-dependent cleavage of the C-tail of Panx1 [81], hypotonicity, ischemia [73], or a secondary response to the activation of receptors ionotropic as P2X4, P2X7, and NMDAR, which are permeable to cations, including Na+, K+, and Ca2+ [83,88,90], or metabotropic receptors such as P2Y1, P2Y2, and P2Y6 [90], protease-activated receptor-1 (PAR-1), bradykinin B2 receptor (BDKRB2) [91], histamine H1 receptor (HRH1), and α1D-adrenergic receptors (α1D-AR) [92].

3. Connexins and Pannexin-Based Channels in the Kidney

The kidney expresses nine connexin isoforms, including Cx26, Cx30, Cx30.3, Cx32, Cx37, Cx40, Cx43, Cx45, and Cx46 [93,94,95,96] (Figure 4). The evidence indicates that these connexins could play a pivotal role in important aspects of renal function, such as the maintenance of acid–base homeostasis, hydroelectrolyte balance, regulation of blood pressure, and functional processes of the nephron: glomerular filtration, reabsorption and secretion of metabolites, water reabsorption, and renin secretion [93,94,95,96].

3.1. Glomerular Connexin and Pannexin-Based Channels

Cx43 is found in the endothelium of the pre-glomerular and post-glomerular regions. In that place, Cx43 builds GJs between endothelial cells, SMCs, and among endothelial cells and SMCs. Moreover, the myoendothelial GJ coupling occurs more frequently in the juxtaglomerular segment of afferent arterioles than in the proximal region and interlobular arteries [94]. The secretory renin-producing cells mainly express Cx40, Cx37, and Cx45 [55,93,97,98]. The role of Cx40 in renin secretion and blood pressure regulation has been established [99,100]. Cx40-deficient mice exhibit hyperreninemia and hypertension because of overactivation of the renin–angiotensin–aldosterone system [101,102,103]. The evidence suggests a decreased coupling between renin-producing cells and neighboring cells. The latter could impair the increase in [Ca2+]i to renin-secreting cells [102,103,104]. Within the glomeruli, the intraglomerular MCs express mainly Cx40 [105], Cx45 [98], Cx43 [106], and Cx37 in the vascular pole [98], while Cx43 is expressed by podocytes [106]. Ultrastructural studies have shown the presence of GJs between different cell populations at the glomerulus [107] with strong levels of Cx40 in the entire intraglomerular mesangium. Cx37 shows expression only in MCs from the vascular pole of the glomerulus [97,108] and GJs composed of Cx43 have been detected between the podocytes [109] (Figure 4).
Although Panx1 represents the most ubiquitous isoform in mammal tissues, it is poorly detected in the kidney tissues [110]. Panx1 expression has been shown in renal vasculature, juxtaglomerular cells [111,112,113], and α-actin-positive SMCs in the afferent arteriole [113]. No data on Panx2 expression have been described in the kidney, whereas Panx3 has been observed in juxtaglomerular cells [114]. Panx1 channel opening reduces renin secretion from renin-expressing cells by increasing [Ca2+]i, which further inactivates Ca2+-sensitive adenylate cyclase. However, the mechanisms associated with cAMP and the subsequent inhibition of renin secretion are poorly understood [111]. Furthermore, P1 and P2X (1-2-4-7) and P2Y (1-2) receptors show expression in the renal vasculature [115], where the P2X1 receptor increases vascular resistance in the afferent arteriole [115] and promotes renin secretion from juxtaglomerular cells [116]. Conversely, P1 receptors mediate tubuloglomerular feedback and suppresses Ca2+-dependent renin secretion [117]. Furthermore, it has been seen that extracellular ATP activates P2X7Rs, which are permeable to cations, including Na+, K+, and Ca2+ [88]. In turn, the activation of P2X7R leads to Panx1 channel opening [15] (Figure 4).

3.2. Tubular Connexin and Pannexin-Based Channels

Under physiological conditions, epithelial cells of the proximal tubule express Cx26 and Cx32, where they colocalize with megalin throughout the proximal tubule segment [98,118]. Additionally, in this renal structure, Cx37 [119] and Cx43 [118] have also been detected in the above cells. Interestingly, in diabetic animals, both Cx26 and Cx43 increase their expression in epithelial cells of the proximal tubule [120]. On the other hand, in the thin ascending limb of the Henle loop and intercalated cells of the collecting duct, the presence of functional HCs formed by Cx30.3 has been confirmed [55]. Purinergic receptors are expressed in this segment and their activation could rely on ATP released through Cx30.3 HCs [55]. On the other hand, Cx37 has been observed in the thick ascending limb of the Henle loop, with its expression being regulated by salt intake, indicating that it likely contributes to Na+ reabsorption [119]. Furthermore, the expression of Cx30 in distal and collecting tubules has also been observed [121]. The opening of Cx30 HCs allows the release of ATP, which reduces the reabsorption of NaCl, thereby increasing natriuresis. Relevant to the above, animals deficient in Cx30 excrete low levels of Na+, causing an increase in blood pressure due to salt retention [122] (Figure 4).
Immunodetection studies have shown the presence of Panx1 in different kidney structures, such as polarized epithelial cells, specifically in apical membranes of proximal tubules, thick descending limbs, and collecting ducts [111,112,113]. This strategic localization suggests that Panx1 regulates the release of ATP from epithelial cells towards the tubular lumen, contributing to renal hemodynamics and tubular salt and water transport [113,123]. Supporting this idea, high levels of extracellular ATP have been found in proximal tubule cells, which correlates with the increased expression of Panx1 and P2Y1 receptors [124,125]. Interestingly, the activation of P2Y1 receptors in the proximal tubules inhibits bicarbonate reabsorption [124,125]. Here, Panx1 could contribute to the release of ATP, similar to the way Cx30.3 HCs contribute in the proximal tubules [55,126]. Panx1 is also expressed in the principal and intercalated cells in the collecting tubule, which regulates water reabsorption and acid–base balance, respectively [127]. ATP released by Panx1 channels in principal cells inhibits the epithelial Na+ channel-mediated Na+ reabsorption linked to Ca2+ signaling [127,128,129] (Figure 4).

4. Cx43 and Panx 1 in Hypertensive Nephropathy

In pathological conditions such as hypertension, levels of renal connexins change. For example, in the two kidneys-one clip model (2K1C), an increase in Cx43 mRNA and protein levels in the glomerulus was observed [130]. A recent study by Abed and colleagues found that Cx43 augments its expression at the early stages of hypertension induced by obstructive nephropathy in the renal cortex of diseased mice [131]. Likewise, ablation of Cx43 improves the renal function of CKD induced by hypertension. The latter suggests that Cx43 contributes to renal cortex damage, and, therefore, its inhibition could be considered a therapeutic target in CDK models [131]. Also, Cx43 is selectively regulated in the vascular small muscle cells of renin-dependent models of hypertension, the AngII activation of the ERK and NF-κB pathways being crucial in this phenomenon [132]. On the other hand, after silencing Cx43 expression, aldosterone-induced apoptosis in cultured podocytes was partially attenuated, suggesting that Cx43 was involved in podocyte apoptosis, induced by aldosterone [131,133]. Toubas and collaborators observed that three different CKD models boosted the immunodetection of Cx43 in glomerulus and tubules, postulating that inflammation could be pivotal in the rise in Cx43 levels in the damaged kidney [134]. Similarly, using a model of renal fibrosis developed in HK2 cells, Potter and coworkers demonstrate that collagen I (an early marker of kidney disease) and TGF-β1 elevate the release of ATP via Cx43 HCs [135]. Furthermore, Yu and coworkers employed another HK2 cell model, and observed that silencing Cx43 expression prevented cisplatin-induced ferroptosis, as well as cell apoptosis [136] (Table 1).
TNF-α and IL-1β reduce intercellular communication mediated by GJs and increase the activity of Cx43 HCs in astrocytes [137]. Interestingly, cultures of proximal tubule cells treated with metabolic inhibitors or pro-inflammatory cytokines display an increased activity of HCs [118,138]. Moreover, in vivo administration of AngII for three weeks or less causes a transitory increase in inflammation, ROS, and Cx43 in the renal cortex, which is associated with renal damage. However, these alterations become irreversible and irreparable if AngII is administered for extended periods (four or more weeks), amplifying inflammatory and oxidative responses along with kidney cell damage [139]. These data support Cx43 as a potential new mediator of renal disease involved in central processes of inflammation and fibrosis, while its inhibition even after the initiation of the disease attenuates renal damage and preserves renal function in animal models of vascular, tubular, and glomerular CKD [140]. The inhibition of Cx43 likely represents a promising future therapeutic option against nephropathy. In summary, the available data highlight the promising beneficial effect of Cx43 inhibition on inflammation, tissue integrity, and fibrosis [141,142] (Table 1).
Table 1. Connexin 43 in hypertensive nephropathy.
Table 1. Connexin 43 in hypertensive nephropathy.
Renal Hypertension ModelExperimental ModelEffect on Cx43Renal SiteTechniqueReference
2K1CKI32 miceWithout changesG, aa, ilaIF, WBHaefliger et al., 2006 [130]
Hypertension-induced CKDRenTg miceRcqPCR, IFAbed et al., 2014 [131]
Uninephrectomized, 1%NaCl and aldosterone infusionSD ratsPoIF, WB, DCFDA fluorescenceYang et al., 2014 [133]
RenTg, anti-glomerular basement membrane, unilateral ureteral obstruction MiceG, Trt-PCR, IFToubas et al., 2011 [134]
Collagen I and TGFβ1 treatmentHK2 cellsPT (cells)Carboxyfluorescein dye uptake Potter et al., 2021 [135]
The cisplatin-induced kidney injury modelHK2 cells, miceRcIF, WB, IHCYu et al., 2021 [136]
ATP depletionhPTCPT (cells)IF, WB, fluorescein dextran dye uptakeVergara et al., 2003 [118]
High glucosehCDCCDIF, WB, Lucifer yellow dye uptakeHills et al., 2006 [138]
Angiotensin II infusionSD ratsRcWBGómez et al., 2019 [139]
TGFβ1 treatmenthPTECs, HK2 cellsPT (cells)Carboxyfluorescein dye uptake Price et al., 2020 [142]
Different study models of hypertensive nephropathy and its effects on connexin 43. Abbreviations: 2K1C, two kidney-one clip model; aa, afferent arteriole; CD, collecting duct; G, glomerulus; hCDC, human collecting duct cells; HK2, clonal tubular epithelial cells; hPTC, human proximal tubule cells; ila, interlobular arteriole; Po, podocytes; PT, proximal tubules; Rc, renal cortex; SD, Sprague–Dawley; T, tubules. ↑ indicates that there is greater expression or protein levels; ↓ less expression or protein levels.
The AngII-mediated activation of AT1Rs is critical for the progressive deterioration of glomerular function, which contributes to the inflammatory and oxidative damage observed in renal diseases [143,144]. In this context, the impairment of Ca2+ buffering leads to podocyte cytoskeletal disorganization, foot process effacement, disruption of the slit diaphragm, and proteinuria, which are critical for the development of diabetic kidney diseases [145]. The AngII half-life lasts about a few hours [15,146]; however, soon after AngII binds to the AT1R [Ca2+]i increases [147,148]. In this sense, those responses might be enough to increase the open probability of Panx1 channels [15,149], and the further release of ATP to the extracellular milieu [15,150]. As previously demonstrated in the nervous system, the ATP released as a result of the [Ca2+]i–induced Panx1 channel opening likely contributes to the progression of cell death. Critical in perpetuating this phenomenon could be the P2X receptor activation, as it leads to persistent Ca2+ entry and subsequent stimulation of intracellular toxic cascades [151].
In the last decade, various studies have linked the uncontrolled opening of Panx1 channels with some diseases [72]. Moreover, although not yet demonstrated, it is possible that Panx1 channels could be permeable to Ca2+, which possibly result in [Ca2+]i overload and the consequent production of free radicals, lipid peroxidation, and plasma membrane damage [152]. Alternatively, uncontrolled Panx1 channel activity could also trigger the release of molecules that, at high concentrations, may be toxic to surrounding cells, such as ATP [152]. In other cell types, the increase in [Ca2+]i raised the activity of Panx1 channels allowing the release of ATP and further activation of P2X7 and P2Y1 receptors [90], where cytokines could also perpetuate Panx1 channel opening [153,154].
On the other hand, more recent evidence has proposed that Panx1 channels increase [Ca2+]i upon stimulation with TNF-α [155], which could lead to the production of free radicals, lipid peroxidation, and plasma membrane damage [152]. Panx1 has been detected in different areas of the kidney [113,156], and palmitic acid stimulates ATP release through the Panx1 channels in human renal tubule epithelial cells (HK-2) by a mechanism involving the activation of caspase-3/7 induced by mitochondrial ROS [155]. Indeed, another study shows that danger signals from necrotic tubular epithelial cells could activate the NOD-like receptor protein 3 (NLRP3) inflammasome in macrophages through the TLR2/caspase 5/Panx1 axis during acute kidney injury (AKI). Relevantly, Panx1 cleavage evoked by caspase 11 was crucial for facilitating the ATP release and posterior activation of the NLRP3 inflammasome during ischemia/reperfusion-induced by AKI [157,158]. Recent work of our group revealed that in AngII-stimulated mesangial MES-13 cells, a similar feed-forward mechanism occurs, similarly between Panx1 channels, Cx43 HCs, and/or P2X7Rs [15]. Accordingly, the blockade of the above protein channels dramatically prevented the AngII-induced increase in oxidative stress and release of cytokines [15].
It is known that high extracellular ATP levels contribute to the transformation of MCs and promote renal injury in AngII-dependent hypertension [159,160] (Figure 5). The latter may induce a self-perpetuating mechanism characterized by the influx of extracellular Ca2+ and IP3 receptor-dependent release of Ca2+ stored in the endoplasmic reticulum. This could occur in the companion of the Panx1 channels-dependent release of cytokines, all these processes being simultaneous, as previously demonstrated in other cell types [161,162] (Figure 5). However, it remains unknown whether Panx1 is present in native MCs and if Panx-1 channels and/or Cx-43 HCs mediate the release of ATP associated with glomerular inflammation or fibrosis, as seen in the example for podocytes and other cells [163,164].

5. Conclusions and Perspectives

For years, hypertensive kidney disease has been considered a disorder limited to afferent arterioles and glomeruli in which the mechanical stresses induced by hypertension, RAS stimulation, and activation of resident fibroblasts were thought to be crucial in underlying kidney damage [21]. Findings discussed in this review support activation of MCs-Cx43 HCs and Panx1 channels as potential contributors to glomerular abnormalities observed following hypertensive nephropathy. The opening of these channels could be the hidden link between mesangial dysfunction and impaired kidney function. Recent development of specific Cx43 HCs and Panx1 channel-blocking peptides will enable future study of molecular mechanisms underlying Cx43 and Panx1 signaling in renal functions.
Despite existing tools, such as connexin KO mice and connexin-blocking peptides, knowledge about connexin implications in renal physiology is still limited [165]. Moreover, whether required communication for renal homeostasis occurs via GJs or HCs is currently poorly understood, and the role of the pannexin family in renal physiology is still unclear. However, recent studies reported their involvement in several diseases in humans and rodents [165]. The use of Panx1-deficient mice would be of significant interest to increase knowledge of Panx1 channel biology in the kidney [165]. Future studies will clarify whether MCs connexin HCs and pannexon opening evoked by AngII occurs with glomerular damage. Understanding how HCs and pannexons participate in the impairment of glomerular cell cross-talk during hypertensive nephropathy is potentially critical for developing pharmacological therapies.

Author Contributions

Conceptualization, C.M.L., J.B., J.P.-V., T.F.A., L.M.-R., M.V., P.B., M.A.R., J.A.O. and G.I.G.; writing—review and editing, C.M.L., J.B., J.P.-V., T.F.A., L.M.-R., M.V., P.B., M.A.R., J.A.O. and G.I.G.; visualization, C.M.L., J.B., J.P.-V., T.F.A., J.A.O. and G.I.G. (figures were created with BioRender.com (accessed~on 15 October 2022)); supervision, M.A.R., J.A.O. and G.I.G.; project administration, G.I.G.; funding acquisition, G.I.G. All authors have read and agreed to the published version of the manuscript.

Funding

FONDECYT INICIACIÓN grant number 11200584 (to G.I.G.). FONDECYT grant number 1210375 (to J.A.O.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

This research was funded by FONDECYT INICIACIÓN grant number 11200584 (to G.I.G.) and FONDECYT grant number 1210375 (to J.A.O.).

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

2K1CTwo kidneys-one clip model
AngIIAngiotensin II
AP-1Apoliprotein-1
AT1RAngiotensin membrane G-protein-coupled receptors type I
AT2RAngiotensin membrane G-protein-coupled receptors type II
ATPAdenosine triphosphate
BMMCBone marrow mononuclear cell
cAMPCyclic adenosine monophosphate
CKDChronic kidney disease
Cx43Connexin 43
eNOSEndothelial nitric oxide synthase
ESRDEnd-stage renal disease
GFRGlomerular filtration rate
IL-1βInterleukin-1β
IP3Inositol trisphosphate
MCP-1Macrophage chemoattractant protein-1
MCsMesangial cells
MES-13Mesangial cells line
NFκBNuclear factor kappa-light-chain-enhancer of activated B cells
NADPHNicotinamide adenine dinucleotide phosphate
NONitric oxide
NOXNADPH oxidase
OSOxidative stress
P2X7RsP2X7 receptors
Panx1Pannexin-1
PGE2Prostaglandin E2
RASRenin–angiotensin system
ROSReactive oxidative species
TGF-β1Transforming growth factor β1
TNF-αTumor necrosis factor-α

References

  1. Atherton, J.C. Renal physiology. Br. J. Anaesth. 1972, 44, 236–245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Mohandas, R.; Douma, L.G.; Scindia, Y.; Gumz, M.L. Circadian Rhythms and Renal Pathophysiology. J. Clin. Investig. 2022, 132, e148277. [Google Scholar] [CrossRef] [PubMed]
  3. Brown, D.; Bouley, R.; Păunescu, T.; Breton, S.; Lu, H. New Insights into the Dynamic Regulation of Water and Acid-Base Balance by Renal Epithelial Cells.o Title. Am. J. Physiol. Cell Physiol. 2012, 302, C1421–C1433. [Google Scholar] [CrossRef] [Green Version]
  4. Lv, J.-C.; Zhang, L.-X. Prevalence and Disease Burden of Chronic Kidney Disease. In Renal Fibrosis: Mechanisms and Therapies; Springer: Berlin, Germany, 2019; pp. 3–15. [Google Scholar]
  5. de Zeeuw, D.; Hillege, H.L.; de Jong, P.E. The Kidney, a Cardiovascular Risk Marker, and a New Target for Therapy. Kidney Int. Suppl. 2005, 68, S25–S29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Gómez, G.I.; Velarde, V. Boldine Improves Kidney Damage in the Goldblatt 2K1C Model Avoiding the Increase in TGF-β. Int. J. Mol. Sci. 2018, 19, 1864. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Lv, J.C.; Zhang, L.X. Prevalence and Disease Burden of Chronic Kidney Disease; Springer: Singapore, 2019; Volume 1165, ISBN 9789811388712. [Google Scholar]
  8. Bikbov, B.; Purcell, C.A.; Levey, A.S.; Smith, M.; Abdoli, A.; Abebe, M.; Adebayo, O.M.; Afarideh, M.; Agarwal, S.K.; Agudelo-Botero, M.; et al. Global, Regional, and National Burden of Chronic Kidney Disease, 1990–2017: A Systematic Analysis for the Global Burden of Disease Study 2017. Lancet 2020, 395, 709–733. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Coresh, J.; Byrd-Holt, D.; Astor, B.C.; Briggs, J.P.; Eggers, P.W.; Lacher, D.A.; Hostetter, T.H. Chronic Kidney Disease Awareness, Prevalence, and Trends among U.S. Adults, 1999 to 2000. J. Am. Soc. Nephrol. 2004, 16, 180–188. [Google Scholar] [CrossRef] [Green Version]
  10. Nangaku, M. Chronic Hypoxia and Tubulointerstitial Injury: A Final Common Pathway to End-Stage Renal Failure. J. Am. Soc. Nephrol. 2005, 17, 17–25. [Google Scholar] [CrossRef] [Green Version]
  11. Gómez, G.I.; Velarde, V.; Sáez, J.C. Connexin-Based Channels and RhoA/ROCK Pathway in Angiotensin II-Induced Kidney Damage. In Selected Chapters from the Renin-Angiotensin System; IntechOpen: London, UK, 2020. [Google Scholar]
  12. Fleck, C.; Appenroth, D.; Jonas, P.; Koch, M.; Kundt, G.; Nizze, H.; Stein, G. Suitability of 5/6 Nephrectomy (5/6NX) for the Induction of Interstitial Renal Fibrosis in Rats--Influence of Sex, Strain, and Surgical Procedure. Exp. Toxicol. Pathol. 2006, 57, 195–205. [Google Scholar] [CrossRef]
  13. Singri, N.; Ahya, S.N.; Levin, M.L. Acute Renal Failure. JAMA 2003, 289, 747–751. [Google Scholar] [CrossRef]
  14. Levey, A.S.; Coresh, J.; Balk, E.; Kausz, A.T.; Levin, A.; Steffes, M.W.; Hogg, R.J.; Perrone, R.D.; Lau, J.; Eknoyan, G.; et al. National Kidney Foundation Practice Guidelines for Chronic Kidney Disease: Evaluation, Classification, and Stratification. Ann. Intern. Med. 2003, 139, 137–147. [Google Scholar] [CrossRef] [PubMed]
  15. Gómez, G.; Fernández, P.; Velarde, V.; Sáez, J. Angiotensin II-Induced Mesangial Cell Damage Is Preceded by Cell Membrane Permeabilization Due to Upregulation of Non-Selective Channels. Int. J. Mol. Sci. 2018, 19, 957. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. López-Novoa, J.M.; Martínez-Salgado, C.; Rodríguez-Peña, A.B.; López-Hernández, F.J. Common Pathophysiological Mechanisms of Chronic Kidney Disease: Therapeutic Perspectives. Pharmacol. Ther. 2010, 128, 61–81. [Google Scholar] [CrossRef] [PubMed]
  17. Petreski, T.; Piko, N.; Ekart, R.; Hojs, R.; Bevc, S. Review on Inflammation Markers in Chronic Kidney Disease. Biomedicines 2021, 9, 182. [Google Scholar] [CrossRef] [PubMed]
  18. Rosivall, L. Intrarenal Renin–Angiotensin System. Mol. Cell Endocrinol. 2009, 302, 185–192. [Google Scholar] [CrossRef] [Green Version]
  19. Seikaly, M.G.; Arant, B.S.; Seney, F.D. Endogenous Angiotensin Concentrations in Specific Intrarenal Fluid Compartments of the Rat. J. Clin. Investig. 1990, 86, 1352–1357. [Google Scholar] [CrossRef] [Green Version]
  20. Rightsel, W.A.; Okamura, T.; Inagami, T.; Pitcock, J.A.; Takii, Y.; Brooks, B.; Brown, P.; Muirhead, E.E. Juxtaglomerular Cells Grown as Monolayer Cell Culture Contain Renin, Angiotensin I-Converting Enzyme, and Angiotensin I and II/III. Circ. Res. 1982, 50, 822–829. [Google Scholar] [CrossRef] [Green Version]
  21. Costantino, V.V.; Gil Lorenzo, A.F.; Bocanegra, V.; Vallés, P.G. Molecular Mechanisms of Hypertensive Nephropathy: Renoprotective Effect of Losartan through Hsp70. Cells 2021, 10, 3146. [Google Scholar] [CrossRef]
  22. Border, W.A.; Noble, N.A. Interactions of Transforming Growth Factor-Beta and Angiotensin II in Renal Fibrosis. Hypertension 1998, 31, 181–188. [Google Scholar] [CrossRef] [Green Version]
  23. Sanchez-Lopez, E.; Rodriguez-Vita, J.; Cartier, C.; Rupérez, M.; Esteban, V.; Carvajal, G.; Díez, R.R.; Plaza, J.J.; Egido, J.; Ruiz-Ortega, M. Inhibitory effect of interleukin-1β on angiotensin II-induced connective tissue growth factor and type IV collagen production in cultured mesangial cells. Am. J. Physiol. Physiol. 2008, 294, F149–F160. [Google Scholar] [CrossRef]
  24. Singh, P.; Bahrami, L.; Castillo, A.; Majid, D.S.A. TNF-α Type 2 Receptor Mediates Renal Inflammatory Response to Chronic Angiotensin II Administration with High Salt Intake in Mice. Am. J. Physiol. Ren. Physiol. 2013, 304, F991–F999. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Mezzano, S.A.; Ruiz-Ortega, M.; Egido, J. Angiotensin II and Renal Fibrosis. Hypertension 2001, 38, 635–638. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Mezzano, S.A.; Aros, C.A.; Droguett, A.; Burgos, M.E.; Ardiles, L.G.; Flores, C.A.; Carpio, D.; Vío, C.P.; Ruiz-Ortega, M. Jes Renal angiotensin II up-regulation and myofibroblast activation in human membranous nephropathy. Kidney Int. 2003, 64, S39–S45. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Ozawa, Y.; Kobori, H.; Suzaki, Y.; Navar, L.G. Sustained renal interstitial macrophage infiltration following chronic angiotensin II infusions. Am. J. Physiol. Physiol. 2007, 292, F330–F339. [Google Scholar] [CrossRef] [PubMed]
  28. Rivera, P.; Ocaranza, M.P.; Lavandero, S.; Jalil, J.E. Rho Kinase Activation and Gene Expression Related to Vascular Remodeling in Normotensive Rats with High Angiotensin I Converting Enzyme Levels. Hypertension 2007, 50, 792–798. [Google Scholar] [CrossRef] [Green Version]
  29. Zhao, W.; Chen, S.S.; Chen, Y.; Ahokas, R.A.; Sun, Y. Kidney Fibrosis in Hypertensive Rats: Role of Oxidative Stress. Am. J. Nephrol. 2008, 28, 548–554. [Google Scholar] [CrossRef] [Green Version]
  30. Clermont, G.; Lecour, S.; Lahet, J.-J.; Siohan, P.; Vergely, C.; Chevet, D.; Rifle, G.; Rochette, L. Alteration in plasma antioxidant capacities in chronic renal failure and hemodialysis patients: A possible explanation for the increased cardiovascular risk in these patients. Cardiovasc. Res. 2000, 47, 618–623. [Google Scholar] [CrossRef]
  31. Johnson, R.J.; Alpers, C.E.; Yoshimura, A.; Lombardi, D.; Pritzl, P.; Floege, J.; Schwartz, S.M. Renal Injury from Angiotensin II-Mediated Hypertension. Hypertension 1992, 19, 464–474. [Google Scholar] [CrossRef] [Green Version]
  32. Lohmeier, T.E. Angiotensin II Infusion Model of Hypertension. Hypertension 2012, 59, 539–541. [Google Scholar] [CrossRef] [Green Version]
  33. Hernández-Salinas, R.; Vielma, A.Z.; Arismendi, M.N.; Boric, M.P.; Sáez, J.C.; Velarde, V. Boldine Prevents Renal Alterations in Diabetic Rats. J. Diabetes Res. 2013, 2013, 593672. [Google Scholar] [CrossRef]
  34. Herrera, G.A. Plasticity of Mesangial Cells: A Basis for Understanding Pathological Alterations. Ultrastruct. Pathol. 2006, 30, 471–479. [Google Scholar] [CrossRef] [PubMed]
  35. Sequeira-Lopez, M.L.S.; Gomez, R.A. Renin Cells, the Kidney, and Hypertension. Circ. Res. 2021, 128, 887–907. [Google Scholar] [CrossRef] [PubMed]
  36. Moon, J.-Y.; Tanimoto, M.; Gohda, T.; Hagiwara, S.; Yamazaki, T.; Ohara, I.; Murakoshi, M.; Aoki, T.; Ishikawa, Y.; Lee, S.-H.; et al. Attenuating effect of angiotensin-(1–7) on angiotensin II-mediated NAD(P)H oxidase activation in type 2 diabetic nephropathy of KK-Ay/Ta mice. Am. J. Physiol. Physiol. 2011, 300, F1271–F1282. [Google Scholar] [CrossRef] [PubMed]
  37. Yue, L.; Wang, W.; Wang, Y.; Du, T.; Shen, W.; Tang, H.; Wang, Y.; Yin, H. Bletilla Striata Polysaccharide Inhibits Angiotensin II-Induced ROS and Inflammation via NOX4 and TLR2 Pathways. Int. J. Biol. Macromol. 2016, 89, 376–388. [Google Scholar] [CrossRef]
  38. Ding, G.-X.; Zhang, A.-H.; Huang, S.-M.; Pan, X.-Q.; Chen, R.-H. SP600125, an inhibitor of c-Jun NH2-terminal kinase, blocks expression of angiotensin II-induced monocyte chemoattractant protein-1 in human mesangial cells. World J. Pediatr. 2010, 6, 169–176. [Google Scholar] [CrossRef]
  39. Bolick, D.T.; Hatley, M.E.; Srinivasan, S.; Hedrick, C.C.; Nadler, J.L. Lisofylline, a Novel Antiinflammatory Compound, Protects Mesangial Cells from Hyperglycemia- and Angiotensin II-Mediated Extracellular Matrix Deposition. Endocrinology 2003, 144, 5227–5231. [Google Scholar] [CrossRef] [Green Version]
  40. Söhl, G.; Maxeiner, S.; Willecke, K. Expression and functions of neuronal gap junctions. Nat. Rev. Neurosci. 2005, 6, 191–200. [Google Scholar] [CrossRef]
  41. Goodenough, D.A.; Paul, D.L. Gap Junctions. Cold Spring Harb. Perspect. Biol. 2009, 1, a002576. [Google Scholar] [CrossRef] [Green Version]
  42. Nielsen, M.S.; Nygaard Axelsen, L.; Sorgen, P.L.; Verma, V.; Delmar, M.; Holstein-Rathlou, N. Gap Junctions. In Comprehensive Physiology; Wiley: New York, NY, USA, 2012; pp. 1981–2035. [Google Scholar]
  43. Scemes, E.; Spray, D.C.; Meda, P. Connexins, Pannexins, Innexins: Novel Roles of “Hemi-Channels”. Pflug. Arch. 2009, 457, 1207–1226. [Google Scholar] [CrossRef] [Green Version]
  44. Leybaert, L.; Lampe, P.D.; Dhein, S.; Kwak, B.R.; Ferdinandy, P.; Beyer, E.C.; Laird, D.W.; Naus, C.C.; Green, C.R.; Schulz, R. Connexins in Cardiovascular and Neurovascular Health and Disease: Pharmacological Implications. Pharm. Rev. 2017, 69, 396–478. [Google Scholar] [CrossRef] [Green Version]
  45. Nielsen, B.S.; Alstrom, J.S.; Nicholson, B.J.; Nielsen, M.S.; MacAulay, N. Permeant-Specific Gating of Connexin 30 Hemichannels. J. Biol. Chem. 2017, 292, 19999–20009. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Hansen, D.B.; Ye, Z.-C.; Calloe, K.; Braunstein, T.H.; Hofgaard, J.P.; Ransom, B.R.; Nielsen, M.S.; MacAulay, N. Activation, Permeability, and Inhibition of Astrocytic and Neuronal Large Pore (Hemi)Channels. J. Biol. Chem. 2014, 289, 26058–26073. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. 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] [PubMed]
  48. Nielsen, B.S.; Zonta, F.; Farkas, T.; Litman, T.; Nielsen, M.S.; MacAulay, N. Structural Determinants Underlying Permeant Discrimination of the Cx43 Hemichannel. J. Biol. Chem. 2019, 294, 16789–16803. [Google Scholar] [CrossRef]
  49. Beblo, D.A.; Veenstra, R.D. Monovalent Cation Permeation through the Connexin40 Gap Junction Channel. J. Gen. Physiol. 1997, 109, 509–522. [Google Scholar] [CrossRef] [Green Version]
  50. 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]
  51. Wang, H.-Z.; Veenstra, R.D. Monovalent Ion Selectivity Sequences of the Rat Connexin43 Gap Junction Channel. J. Gen. Physiol. 1997, 109, 491–507. [Google Scholar] [CrossRef] [Green Version]
  52. Goldberg, G.S.; Moreno, A.P.; Lampe, P.D. Gap Junctions between Cells Expressing Connexin 43 or 32 Show Inverse Permselectivity to Adenosine and ATP. J. Biol. Chem. 2002, 277, 36725–36730. [Google Scholar] [CrossRef] [Green Version]
  53. Belliveau, D.J.; Bani-Yaghoub, M.; McGirr, B.; Naus, C.C.G.; Rushlow, W.J. Enhanced Neurite Outgrowth in PC12 Cells Mediated by Connexin Hemichannels and ATP. J. Biol. Chem. 2006, 281, 20920–20931. [Google Scholar] [CrossRef] [Green Version]
  54. Sarieddine, M.Z.R.; Scheckenbach, K.E.L.; Foglia, B.; Maass, K.; Garcia, I.; Kwak, B.R.; Chanson, M. Connexin43 Modulates Neutrophil Recruitment to the Lung. J. Cell Mol. Med. 2009, 13, 4560–4570. [Google Scholar] [CrossRef]
  55. Hanner, F.; Schnichels, M.; Zheng-Fischhöfer, Q.; Yang, L.E.; Toma, I.; Willecke, K.; McDonough, A.A.; Peti-Peterdi, J. Connexin 30.3 Is Expressed in the Kidney But Not Regulated by Dietary Salt or High Blood Pressure. Cell Commun. Adhes. 2008, 15, 219–230. [Google Scholar] [CrossRef] [PubMed]
  56. Peracchia, C. Chemical gating of gap junction channels: Roles of calcium, pH and calmodulin. Biochim. Biophys. Acta (BBA) Biomembr. 2004, 1662, 61–80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Liu, S.; Taffet, S.; Stoner, L.; Delmar, M.; Vallano, M.L.; Jalife, J. A Structural Basis for the Unequal Sensitivity of the Major Cardiac and Liver Gap Junctions to Intracellular Acidification: The Carboxyl Tail Length. Biophys. J. 1993, 64, 1422–1433. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Saez, J.C.; Spray, D.C.; Nairn, A.C.; Hertzberg, E.; Greengard, P.; Bennett, M. v CAMP Increases Junctional Conductance and Stimulates Phosphorylation of the 27-KDa Principal Gap Junction Polypeptide. Proc. Natl. Acad. Sci. USA 1986, 83, 2473–2477. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Retamal, M.A.; Cortes, C.J.; Reuss, L.; Bennett, M.V.L.; Saez, 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]
  60. Chandrasekhar, A.; Bera, A.K. Hemichannels: Permeants and Their Effect on Development, Physiology and Death. Cell Biochem. Funct. 2012, 30, 89–100. [Google Scholar] [CrossRef]
  61. Sáez, J.C.; Schalper, K.A.; Retamal, M.A.; Orellana, J.A.; Shoji, K.F.; Bennett, M.V.L. Cell Membrane Permeabilization via Connexin Hemichannels in Living and Dying Cells. Exp. Cell Res. 2010, 316, 2377–2389. [Google Scholar] [CrossRef]
  62. 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]
  63. Prieto-Villalobos, J.; Alvear, T.F.; Liberona, A.; Lucero, C.M.; Martínez-Araya, C.J.; Balmazabal, J.; Inostroza, C.A.; Ramírez, G.; Gómez, G.I.; Orellana, J.A. Astroglial Hemichannels and Pannexons: The Hidden Link between Maternal Inflammation and Neurological Disorders. Int. J. Mol. Sci. 2021, 22, 9503. [Google Scholar] [CrossRef]
  64. O’Carroll, S.J.; Alkadhi, M.; Nicholson, L.F.B.; Green, C.R. Connexin43 Mimetic Peptides Reduce Swelling, Astrogliosis, and Neuronal Cell Death after Spinal Cord Injury. Cell Commun. Adhes. 2008, 15, 27–42. [Google Scholar] [CrossRef]
  65. Paul, D.L.; Ebihara, L.; Takemoto, L.J.; Swenson, K.I.; Goodenough, D.A. Connexin46, a Novel Lens Gap Junction Protein, Induces Voltage-Gated Currents in Nonjunctional Plasma Membrane of Xenopus Oocytes. J. Cell Biol. 1991, 115, 1077–1089. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Fiori, M.C.; Figueroa, V.; Zoghbi, M.E.; Saéz, J.C.; Reuss, L.; Altenberg, G.A. Permeation of Calcium through Purified Connexin 26 Hemichannels. J. Biol. Chem. 2012, 287, 40826–40834. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. 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. Cell Physiol. 2010, 299, C1504–C1515. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Kim, J.-C.; Pérez-Hernández, M.; Alvarado, F.J.; Maurya, S.R.; Montnach, J.; Yin, Y.; Zhang, M.; Lin, X.; Vasquez, C.; Heguy, A.; et al. Disruption of Ca 2+ i Homeostasis and Connexin 43 Hemichannel Function in the Right Ventricle Precedes Overt Arrhythmogenic Cardiomyopathy in Plakophilin-2–Deficient Mice. Circulation 2019, 140, 1015–1030. [Google Scholar] [CrossRef]
  69. Yi, C.; Mei, X.; Ezan, P.; Mato, S.; Matias, I.; Giaume, C.; Koulakoff, A. Astroglial connexin43 contributes to neuronal suffering in a mouse model of Alzheimer’s disease. Cell Death Differ. 2016, 23, 1691–1701. [Google Scholar] [CrossRef] [Green Version]
  70. Orellana, J.A.; Shoji, K.F.; Abudara, V.; Ezan, P.; Amigou, E.; Saez, P.J.; Jiang, J.X.; Naus, C.C.; Saez, J.C.; Giaume, C. Amyloid -Induced Death in Neurons Involves Glial and Neuronal Hemichannels. J. Neurosci. 2011, 31, 4962–4977. [Google Scholar] [CrossRef] [Green Version]
  71. Barbe, M.T.; Monyer, H.; Bruzzone, R. Cell-Cell Communication Beyond Connexins: The Pannexin Channels. Physiology 2006, 21, 103–114. [Google Scholar] [CrossRef]
  72. Penuela, S.; Harland, L.; Simek, J.; Laird, D.W. Pannexin Channels and Their Links to Human Disease. Biochem. J. 2014, 461, 371–381. [Google Scholar] [CrossRef]
  73. Lohman, A.W.; Isakson, B.E. Differentiating Connexin Hemichannels and Pannexin Channels in Cellular ATP Release. FEBS Lett. 2014, 588, 1379–1388. [Google Scholar] [CrossRef] [Green Version]
  74. Gajardo-Gómez, R.; Labra, V.C.; Orellana, J.A. Connexins and Pannexins: New Insights into Microglial Functions and Dysfunctions. Front. Mol. Neurosci. 2016, 9, 86. [Google Scholar] [CrossRef]
  75. O’Donnell, B.L.; Penuela, S. Pannexin 3 Channels in Health and Disease. Purinergic Signal. 2021, 17, 577–589. [Google Scholar] [CrossRef] [PubMed]
  76. Taylor, K.A.; Wright, J.R.; Mahaut-Smith, M.P. Regulation of Pannexin-1 Channel Activity. Biochem. Soc. Trans. 2015, 43, 502–507. [Google Scholar] [CrossRef] [PubMed]
  77. Michalski, K.; Syrjanen, J.L.; Henze, E.; Kumpf, J.; Furukawa, H.; Kawate, T. The Cryo-EM Structure of Pannexin 1 Reveals Unique Motifs for Ion Selection and Inhibition. Elife 2020, 9, e54670. [Google Scholar] [CrossRef] [Green Version]
  78. Palacios-Prado, N.; Soto, P.A.; López, X.; Choi, E.J.; Marquez-Miranda, V.; Rojas, M.; Duarte, Y.; Lee, J.; González-Nilo, F.D.; Sáez, J.C. Endogenous Pannexin1 Channels Form Functional Intercellular Cell–Cell Channels with Characteristic Voltage-Dependent Properties. Proc. Natl. Acad. Sci. USA 2022, 119, e2202104119. [Google Scholar] [CrossRef] [PubMed]
  79. Billaud, M.; Sandilos, J.K.; Isakson, B.E. Pannexin 1 in the Regulation of Vascular Tone. Trends Cardiovasc. Med. 2012, 22, 68–72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Krick, S.; Wang, J.; St-Pierre, M.; Gonzalez, C.; Dahl, G.; Salathe, M. Dual Oxidase 2 (Duox2) Regulates Pannexin 1-Mediated ATP Release in Primary Human Airway Epithelial Cells via Changes in Intracellular PH and Not H2O2 Production. J. Biol. Chem. 2016, 291, 6423–6432. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Chekeni, F.B.; Elliott, M.R.; Sandilos, J.K.; Walk, S.F.; Kinchen, J.M.; Lazarowski, E.R.; Armstrong, A.J.; Penuela, S.; Laird, D.W.; Salvesen, G.S.; et al. Pannexin 1 Channels Mediate “find-Me” Signal Release and Membrane Permeability during Apoptosis. Nature 2010, 467, 863–867. [Google Scholar] [CrossRef] [Green Version]
  82. Adamson, S.E.; Leitinger, N. The Role of Pannexin1 in the Induction and Resolution of Inflammation. FEBS Lett. 2014, 588, 1416–1422. [Google Scholar] [CrossRef] [Green Version]
  83. Silverman, W.R.; de Rivero Vaccari, J.P.; Locovei, S.; Qiu, F.; Carlsson, S.K.; Scemes, E.; Keane, R.W.; Dahl, G. The Pannexin 1 Channel Activates the Inflammasome in Neurons and Astrocytes. J. Biol. Chem. 2009, 284, 18143–18151. [Google Scholar] [CrossRef] [Green Version]
  84. Su, L.; Jiang, X.; Yang, C.; Zhang, J.; Chen, B.; Li, Y.; Yao, S.; Xie, Q.; Gomez, H.; Murugan, R.; et al. Pannexin 1 Mediates Ferroptosis That Contributes to Renal Ischemia/Reperfusion Injury. J. Biol. Chem. 2019, 294, 19395–19404. [Google Scholar] [CrossRef]
  85. Malik, S.; Eugenin, E.A. Role of Connexin and Pannexin Containing Channels in HIV Infection and NeuroAIDS. Neurosci. Lett. 2019, 695, 86–90. [Google Scholar] [CrossRef] [PubMed]
  86. Penuela, S.; Gehi, R.; Laird, D.W. The Biochemistry and Function of Pannexin Channels. Biochim. Biophys. Acta (BBA) Biomembr. 2013, 1828, 15–22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Whyte-Fagundes, P.; Zoidl, G. Mechanisms of Pannexin1 Channel Gating and Regulation. Biochim. Biophys. Acta (BBA) Biomembr. 2018, 1860, 65–71. [Google Scholar] [CrossRef]
  88. Volonté, C.; Apolloni, S.; Skaper, S.D.; Burnstock, G. P2X7 Receptors: Channels, Pores and More. CNS Neurol. Disord. Drug Targets 2012, 11, 705–721. [Google Scholar] [CrossRef] [PubMed]
  89. Bao, L.; Locovei, S.; Dahl, G. Pannexin Membrane Channels Are Mechanosensitive Conduits for ATP. FEBS Lett. 2004, 572, 65–68. [Google Scholar] [CrossRef] [Green Version]
  90. Locovei, S.; Wang, J.; Dahl, G. Activation of Pannexin 1 Channels by ATP through P2Y Receptors and by Cytoplasmic Calcium. FEBS Lett. 2006, 580, 239–244. [Google Scholar] [CrossRef] [Green Version]
  91. Pinheiro, A.R.; Paramos-de-Carvalho, D.; Certal, M.; Costa, C.; Magalhães-Cardoso, M.T.; Ferreirinha, F.; Costa, M.A.; Correia-de-Sá, P. Bradykinin-Induced Ca2+ Signaling in Human Subcutaneous Fibroblasts Involves ATP Release via Hemichannels Leading to P2Y12 Receptors Activation. Cell Commun. Signal. 2013, 11, 70. [Google Scholar] [CrossRef] [Green Version]
  92. Billaud, M.; Lohman, A.W.; Straub, A.C.; Looft-Wilson, R.; Johnstone, S.R.; Araj, C.A.; Best, A.K.; Chekeni, F.B.; Ravichandran, K.S.; Penuela, S.; et al. Pannexin1 Regulates A1-Adrenergic Receptor–Mediated Vasoconstriction. Circ. Res. 2011, 109, 80–85. [Google Scholar] [CrossRef] [Green Version]
  93. Kurtz, A. Renal Connexins and Blood Pressure. Biochim. Biophys. Acta 2012, 1818, 1903–1908. [Google Scholar] [CrossRef] [Green Version]
  94. Hanner, F.; Sorensen, C.M.; Holstein-Rathlou, N.-H.; Peti-Peterdi, J. Connexins and the kidney. Am. J. Physiol. Integr. Comp. Physiol. 2010, 298, R1143–R1155. [Google Scholar] [CrossRef]
  95. Zhang, Q.; Cao, C.; Mangano, M.; Zhang, Z.; Silldorff, E.P.; Lee-Kwon, W.; Payne, K.; Pallone, T.L. Descending vasa recta endothelium is an electrical syncytium. Am. J. Physiol. Integr. Comp. Physiol. 2006, 291, R1688–R1699. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Zehra, T.; Cupples, W.A.; Braam, B. Tubuloglomerular Feedback Synchronization in Nephrovascular Networks. J. Am. Soc. Nephrol. 2021, 32, 1293–1304. [Google Scholar] [CrossRef] [PubMed]
  97. Hwan Seul, K.; Beyer, E.C. Heterogeneous Localization of Connexin40 in the Renal Vasculature. Microvasc. Res. 2000, 59, 140–148. [Google Scholar] [CrossRef] [PubMed]
  98. Geis, L.; Boudriot, F.-F.; Wagner, C. Connexin MRNA Distribution in Adult Mouse Kidneys. Pflug. Arch. 2021, 473, 1737–1747. [Google Scholar] [CrossRef]
  99. de Vriese, A.S.; van de Voorde, J.; Lameire, N.H. Effects of Connexin-Mimetic Peptides on Nitric Oxide Synthase- and Cyclooxygenase-Independent Renal Vasodilation. Kidney Int. 2002, 61, 177–185. [Google Scholar] [CrossRef] [Green Version]
  100. Takenaka, T.; Inoue, T.; Kanno, Y.; Okada, H.; Meaney, K.R.; Hill, C.E.; Suzuki, H. Expression and Role of Connexins in the Rat Renal Vasculature. Kidney Int. 2008, 73, 415–422. [Google Scholar] [CrossRef] [Green Version]
  101. Krattinger, N.; Capponi, A.; Mazzolai, L.; Aubert, J.-F.; Caille, D.; Nicod, P.; Waeber, G.; Meda, P.; Haefliger, J.-A. Connexin40 Regulates Renin Production and Blood Pressure. Kidney Int. 2007, 72, 814–822. [Google Scholar] [CrossRef] [Green Version]
  102. Wagner, C.; Jobs, A.; Schweda, F.; Kurtz, L.; Kurt, B.; Lopez, M.L.S.; Gomez, R.A.; van Veen, T.A.B.; de Wit, C.; Kurtz, A. Selective Deletion of Connexin 40 in Renin-Producing Cells Impairs Renal Baroreceptor Function and Is Associated with Arterial Hypertension. Kidney Int. 2010, 78, 762–768. [Google Scholar] [CrossRef] [Green Version]
  103. Wagner, C.; de Wit, C.; Kurtz, L.; Grünberger, C.; Kurtz, A.; Schweda, F. Connexin40 Is Essential for the Pressure Control of Renin Synthesis and Secretion. Circ. Res. 2007, 100, 556–563. [Google Scholar] [CrossRef]
  104. Schweda, F.; Kurtz, A. Cellular mechanism of renin release. Acta Physiol. Scand. 2004, 181, 383–390. [Google Scholar] [CrossRef]
  105. Just, A.; Kurtz, L.; de Wit, C.; Wagner, C.; Kurtz, A.; Arendshorst, W.J. Connexin 40 Mediates the Tubuloglomerular Feedback Contribution to Renal Blood Flow Autoregulation. J. Am. Soc. Nephrol. 2009, 20, 1577–1585. [Google Scholar] [CrossRef] [PubMed]
  106. Morioka, T.; Okada, S.; Nameta, M.; Kamal, F.; Yanakieva-Georgieva, N.T.; Yao, J.; Sato, A.; Piao, H.; Oite, T. Glomerular Expression of Connexin 40 and Connexin 43 in Rat Experimental Glomerulonephritis. Clin. Exp. Nephrol. 2013, 17, 191–204. [Google Scholar] [CrossRef] [PubMed]
  107. Kar, R.; Batra, N.; Riquelme, M.A.; Jiang, J.X. Biological Role of Connexin Intercellular Channels and Hemichannels. Arch. Biochem. Biophys. 2012, 524, 2–15. [Google Scholar] [CrossRef] [Green Version]
  108. Zhang, J.; Hill, C.E. Differential Connexin Expression in Preglomerular and Postglomerular Vasculature: Accentuation during Diabetes. Kidney Int. 2005, 68, 1171–1185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Sawai, K.; Mukoyama, M.; Mori, K.; Yokoi, H.; Koshikawa, M.; Yoshioka, T.; Takeda, R.; Sugawara, A.; Kuwahara, T.; Saleem, M.A.; et al. Redistribution of Connexin43 Expression in Glomerular Podocytes Predicts Poor Renal Prognosis in Patients with Type 2 Diabetes and Overt Nephropathy. Nephrol. Dial. Transplant. 2006, 21, 2472–2477. [Google Scholar] [CrossRef] [Green Version]
  110. Baranova, A.; Ivanov, D.; Petrash, N.; Pestova, A.; Skoblov, M.; Kelmanson, I.; Shagin, D.; Nazarenko, S.; Geraymovych, E.; Litvin, O.; et al. The Mammalian Pannexin Family Is Homologous to the Invertebrate Innexin Gap Junction Proteins. Genomics 2004, 83, 706–716. [Google Scholar] [CrossRef]
  111. DeLalio, L.J.; Masati, E.; Mendu, S.; Ruddiman, C.A.; Yang, Y.; Johnstone, S.R.; Milstein, J.A.; Keller, T.C.S.; Weaver, R.B.; Guagliardo, N.A.; et al. Pannexin 1 Channels in Renin-Expressing Cells Influence Renin Secretion and Blood Pressure Homeostasis. Kidney Int. 2020, 98, 630–644. [Google Scholar] [CrossRef]
  112. Jankowski, J.; Perry, H.M.; Medina, C.B.; Huang, L.; Yao, J.; Bajwa, A.; Lorenz, U.M.; Rosin, D.L.; Ravichandran, K.S.; Isakson, B.E.; et al. Epithelial and Endothelial Pannexin1 Channels Mediate AKI. J. Am. Soc. Nephrol. 2018, 29, 1887–1899. [Google Scholar] [CrossRef] [Green Version]
  113. Hanner, F.; Lam, L.; Nguyen, M.T.X.; Yu, A.; Peti-Peterdi, J. Intrarenal localization of the plasma membrane ATP channel pannexin1. Am. J. Physiol. Physiol. 2012, 303, F1454–F1459. [Google Scholar] [CrossRef] [Green Version]
  114. Lohman, A.W.; Billaud, M.; Straub, A.C.; Johnstone, S.R.; Best, A.K.; Lee, M.; Barr, K.; Penuela, S.; Laird, D.W.; Isakson, B.E. Expression of Pannexin Isoforms in the Systemic Murine Arterial Network. J. Vasc. Res. 2012, 49, 405–416. [Google Scholar] [CrossRef]
  115. Guan, Z.; Inscho, E.W. Role of Adenosine 5′-Triphosphate in Regulating Renal Microvascular Function and in Hypertension. Hypertension 2011, 58, 333–340. [Google Scholar] [CrossRef] [PubMed]
  116. Yao, J.; Suwa, M.; Li, B.; Kawamura, K.; Morioka, T.; Oite, T. ATP-Dependent Mechanism for Coordination of Intercellular Ca 2+ Signaling and Renin Secretion in Rat Juxtaglomerular Cells. Circ. Res. 2003, 93, 338–345. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Ortiz-Capisano, M.C.; Atchison, D.K.; Harding, P.; Lasley, R.D.; Beierwaltes, W.H. Adenosine inhibits renin release from juxtaglomerular cells via an A1 receptor-TRPC-mediated pathway. Am. J. Physiol. Physiol. 2013, 305, F1209–F1219. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Vergara, L.; Bao, X.; Cooper, M.; Bello-Reuss, E.; Reuss, L. Gap-Junctional Hemichannels Are Activated by ATP Depletion in Human Renal Proximal Tubule Cells. J. Membr. Biol. 2003, 196, 173–184. [Google Scholar] [CrossRef]
  119. Stoessel, A.; Himmerkus, N.; Bleich, M.; Bachmann, S.; Theilig, F. Connexin 37 is localized in renal epithelia and responds to changes in dietary salt intake. Am. J. Physiol. Physiol. 2010, 298, F216–F223. [Google Scholar] [CrossRef]
  120. Hills, C.; Price, G.W.; Wall, M.J.; Kaufmann, T.J.; Tang, C.-W.; Yiu, W.H.; Squires, P.E. Transforming Growth Factor Beta 1 Drives a Switch in Connexin Mediated Cell-to-Cell Communication in Tubular Cells of the Diabetic Kidney. Cell. Physiol. Biochem. 2018, 45, 2369–2388. [Google Scholar] [CrossRef] [Green Version]
  121. McCulloch, F.; Chambrey, R.; Eladari, D.; Peti-Peterdi, J. Localization of connexin 30 in the luminal membrane of cells in the distal nephron. Am. J. Physiol. Physiol. 2005, 289, F1304–F1312. [Google Scholar] [CrossRef]
  122. Sipos, A.; Vargas, S.L.; Toma, I.; Hanner, F.; Willecke, K.; Peti-Peterdi, J. Connexin 30 Deficiency Impairs Renal Tubular ATP Release and Pressure Natriuresis. J. Am. Soc. Nephrol. 2009, 20, 1724–1732. [Google Scholar] [CrossRef] [Green Version]
  123. Praetorius, H.A.; Leipziger, J. Intrarenal Purinergic Signaling in the Control of Renal Tubular Transport. Annu. Rev. Physiol. 2010, 72, 377–393. [Google Scholar] [CrossRef]
  124. Vekaria, R.M.; Unwin, R.J.; Shirley, D.G. Intraluminal ATP Concentrations in Rat Renal Tubules. J. Am. Soc. Nephrol. 2006, 17, 1841–1847. [Google Scholar] [CrossRef]
  125. Bailey, M.A. Inhibition of Bicarbonate Reabsorption in the Rat Proximal Tubule by Activation of Luminal P2Y1 Receptors. Am. J. Physiol. Ren. Physiol. 2004, 287, F789–F796. [Google Scholar] [CrossRef] [Green Version]
  126. Kordowitzki, P.; Kranc, W.; Bryl, R.; Kempisty, B.; Skowronska, A.; Skowronski, M.T. The Relevance of Aquaporins for the Physiology, Pathology, and Aging of the Female Reproductive System in Mammals. Cells 2020, 9, 2570. [Google Scholar] [CrossRef] [PubMed]
  127. Almomani, E.; Kaur, S.; Alexander, R.; Cordat, E. Intercalated Cells: More than PH Regulation. Diseases 2014, 2, 71–92. [Google Scholar] [CrossRef] [Green Version]
  128. Svenningsen, P.; Burford, J.; Peti-Peterdi, J. ATP Releasing Connexin 30 Hemichannels Mediate Flow-Induced Calcium Signaling in the Collecting Duct. Front. Physiol. 2013, 4, 292. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Leipziger, J. Control of Epithelial Transport via Luminal P2 Receptors. Am. J. Physiol. Ren. Physiol. 2003, 284, F419–F432. [Google Scholar] [CrossRef] [Green Version]
  130. Haefliger, J.-A.; Krattinger, N.; Martin, D.; Pedrazzini, T.; Capponi, A.; Döring, B.; Plum, A.; Charollais, A.; Willecke, K.; Meda, P. Connexin43-Dependent Mechanism Modulates Renin Secretion and Hypertension. J. Clin. Investig. 2006, 116, 405–413. [Google Scholar] [CrossRef] [Green Version]
  131. Abed, A.; Toubas, J.; Kavvadas, P.; Authier, F.; Cathelin, D.; Alfieri, C.; Boffa, J.-J.; Dussaule, J.-C.; Chatziantoniou, C.; Chadjichristos, C.E. Targeting Connexin 43 Protects against the Progression of Experimental Chronic Kidney Disease in Mice. Kidney Int. 2014, 86, 768–779. [Google Scholar] [CrossRef] [Green Version]
  132. Alonso, F.; Krattinger, N.; Mazzolai, L.; Simon, A.; Waeber, G.; Meda, P.; Haefliger, J.-A. An Angiotensin II- and NF-ΚB-Dependent Mechanism Increases Connexin 43 in Murine Arteries Targeted by Renin-Dependent Hypertension. Cardiovasc. Res. 2010, 87, 166–176. [Google Scholar] [CrossRef] [Green Version]
  133. Yang, M.; Wang, B.; Li, M.; Jiang, B. Connexin 43 Is Involved in Aldosterone-Induced Podocyte Injury. Cell. Physiol. Biochem. 2014, 34, 1652–1662. [Google Scholar] [CrossRef]
  134. Toubas, J.; Beck, S.; Pageaud, A.-L.; Huby, A.-C.; Mael-Ainin, M.; Dussaule, J.-C.; Chatziantoniou, C.; Chadjichristos, C.E. Alteration of Connexin Expression Is an Early Signal for Chronic Kidney Disease. Am. J. Physiol. Ren. Physiol. 2011, 301, F24–F32. [Google Scholar] [CrossRef]
  135. Potter, J.A.; Price, G.W.; Cliff, C.L.; Green, C.R.; Squires, P.E.; Hills, C.E. Collagen I Modifies Connexin-43 Hemichannel Activity via Integrin A2β1 Binding in TGFβ1-Evoked Renal Tubular Epithelial Cells. Int. J. Mol. Sci. 2021, 22, 3644. [Google Scholar] [CrossRef] [PubMed]
  136. Yu, M.; Lin, Z.; Tian, X.; Chen, S.; Liang, X.; Qin, M.; Zhu, Q.; Wu, Y.; Zhong, S. Downregulation of Cx43 Reduces Cisplatin-Induced Acute Renal Injury by Inhibiting Ferroptosis. Food Chem. Toxicol. 2021, 158, 112672. [Google Scholar] [CrossRef] [PubMed]
  137. 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] [Green Version]
  138. Hills, C.E.; Bland, R.; Wheelans, D.C.; Bennett, J.; Ronco, P.M.; Squires, P.E. Glucose-Evoked Alterations in Connexin43-Mediated Cell-to-Cell Communication in Human Collecting Duct: A Possible Role in Diabetic Nephropathy. Am. J. Physiol. Ren. Physiol. 2006, 291, F1045–F1051. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Gómez, G.I.; Velarde, V.; Sáez, J.C. Role of a RhoA/ROCK-Dependent Pathway on Renal Connexin43 Regulation in the Angiotensin II-Induced Renal Damage. Int. J. Mol. Sci. 2019, 20, 4408. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  140. Prakoura, N.; Kavvadas, P.; Chadjichristos, C.E. Connexin 43: A New Therapeutic Target Against Chronic Kidney Disease. Cell. Physiol. Biochem. 2018, 49, 985. [Google Scholar] [CrossRef]
  141. Cliff, C.L.; Williams, B.M.; Chadjichristos, C.E.; Mouritzen, U.; Squires, P.E.; Hills, C.E. Connexin 43: A Target for the Treatment of Inflammation in Secondary Complications of the Kidney and Eye in Diabetes. Int. J. Mol. Sci. 2022, 23, 600. [Google Scholar] [CrossRef]
  142. Price, G.W.; Chadjichristos, C.E.; Kavvadas, P.; Tang, S.C.W.; Yiu, W.H.; Green, C.R.; Potter, J.A.; Siamantouras, E.; Squires, P.E.; Hills, C.E. Blocking Connexin-43 Mediated Hemichannel Activity Protects against Early Tubular Injury in Experimental Chronic Kidney Disease. Cell Commun. Signal. 2020, 18, 79. [Google Scholar] [CrossRef] [PubMed]
  143. Kolavennu, V.; Zeng, L.; Peng, H.; Wang, Y.; Danesh, F.R. Targeting of RhoA/ROCK Signaling Ameliorates Progression of Diabetic Nephropathy Independent of Glucose Control. Diabetes 2008, 57, 714–723. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Rupérez, M.; Sánchez-López, E.; Blanco-Colio, L.M.; Esteban, V.; Rodríguez-Vita, J.; Plaza, J.J.; Egido, J.; Ruiz-Ortega, M. The Rho-Kinase Pathway Regulates Angiotensin II-Induced Renal Damage. Kidney Int. Suppl. 2005, 68, S39–S45. [Google Scholar] [CrossRef]
  145. Ilatovskaya, D.V.; Blass, G.; Palygin, O.; Levchenko, V.; Pavlov, T.S.; Grzybowski, M.N.; Winsor, K.; Shuyskiy, L.S.; Geurts, A.M.; Cowley, A.W.; et al. A NOX4/TRPC6 Pathway in Podocyte Calcium Regulation and Renal Damage in Diabetic Kidney Disease. J. Am. Soc. Nephrol. 2018, 29, 1917–1927. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. van Kats, J.P.; de Lannoy, L.M.; Jan Danser, A.H.; van Meegen, J.R.; Verdouw, P.D.; Schalekamp, M.A. Angiotensin II Type 1 (AT1) Receptor-Mediated Accumulation of Angiotensin II in Tissues and Its Intracellular Half-Life in Vivo. Hypertension 1997, 30, 42–49. [Google Scholar] [CrossRef] [PubMed]
  147. Feng, Z.; Wei, C.; Chen, X.; Wang, J.; Cheng, H.; Zhang, X.; Hong, Q.; Shi, S.; Fu, B.; Wei, R. Essential Role of Ca2+ Release Channels in Angiotensin II-Induced Ca2+ Oscillations and Mesangial Cell Contraction. Kidney Int. 2006, 70, 130–138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Qiu, G.; Ji, Z. AngII-Induced Glomerular Mesangial Cell Proliferation Inhibited by Losartan via Changes in Intracellular Calcium Ion Concentration. Clin. Exp. Med. 2014, 14, 169–176. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Zhang, M.; Piskuric, N.A.; Vollmer, C.; Nurse, C.A. P2Y2 Receptor Activation Opens Pannexin-1 Channels in Rat Carotid Body Type II Cells: Potential Role in Amplifying the Neurotransmitter ATP. J. Physiol. 2012, 590, 4335–4350. [Google Scholar] [CrossRef]
  150. Dahl, G.; Qiu, F.; Wang, J. The Bizarre Pharmacology of the ATP Release Channel Pannexin1. Neuropharmacology 2013, 75, 583–593. [Google Scholar] [CrossRef] [Green Version]
  151. Orellana, J.A.; von Bernhardi, R.; Giaume, C.; Sáez, J.C. Glial Hemichannels and Their Involvement in Aging and Neurodegenerative Diseases. Rev. Neurosci. 2012, 23, 163–177. [Google Scholar] [CrossRef]
  152. Abudara, V.; Retamal, M.A.; Del Rio, R.; Orellana, J. Synaptic Functions of Hemichannels and Pannexons: A Double-Edged Sword. Front. Mol. Neurosci. 2018, 11, 435. [Google Scholar] [CrossRef]
  153. Sáez, P.J.; Orellana, J.A.; Vega-Riveros, N.; Figueroa, V.A.; Hernández, D.E.; Castro, J.F.; Klein, A.D.; Jiang, J.X.; Zanlungo, S.; Sáez, J.C. Disruption in Connexin-Based Communication Is Associated with Intracellular Ca2+ Signal Alterations in Astrocytes from Niemann-Pick Type C Mice. PLoS ONE 2013, 8, e71361. [Google Scholar] [CrossRef] [Green Version]
  154. Garre, J.M.; Yang, G.; Bukauskas, F.F.; Bennett, M.V.L. FGF-1 Triggers Pannexin-1 Hemichannel Opening in Spinal Astrocytes of Rodents and Promotes Inflammatory Responses in Acute Spinal Cord Slices. J. Neurosci. 2016, 36, 4785–4801. [Google Scholar] [CrossRef] [Green Version]
  155. Sun, H.; Sun, Z.; Varghese, Z.; Guo, Y.; Moorhead, J.F.; Unwin, R.J.; Ruan, X.Z. Nonesterified Free Fatty Acids Enhance the Inflammatory Response in Renal Tubules by Inducing Extracellular ATP Release. Am. J. Physiol. Ren. Physiol. 2020, 319, F292–F303. [Google Scholar] [CrossRef] [PubMed]
  156. Menzies, R.I.; Howarth, A.R.; Unwin, R.J.; Tam, F.W.K.; Mullins, J.J.; Bailey, M.A. Inhibition of the Purinergic P2X7 Receptor Improves Renal Perfusion in Angiotensin-II-Infused Rats. Kidney Int. 2015, 88, 1079–1087. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Yin, F.; Zheng, P.; Zhao, L.; Wang, Y.; Miao, N.; Zhou, Z.; Cheng, Q.; Chen, P.; Xie, H.; Li, J.; et al. Caspase-11 Promotes NLRP3 Inflammasome Activation via the Cleavage of Pannexin1 in Acute Kidney Disease. Acta Pharm. Sin. 2022, 43, 86–95. [Google Scholar] [CrossRef] [PubMed]
  158. Liu, C.; Shen, Y.; Huang, L.; Wang, J. TLR2/Caspase-5/Panx1 Pathway Mediates Necrosis-Induced NLRP3 Inflammasome Activation in Macrophages during Acute Kidney Injury. Cell Death Discov. 2022, 8, 232. [Google Scholar] [CrossRef] [PubMed]
  159. Graciano, M.L.; Nishiyama, A.; Jackson, K.; Seth, D.M.; Ortiz, R.M.; Prieto-Carrasquero, M.C.; Kobori, H.; Navar, L.G. Purinergic Receptors Contribute to Early Mesangial Cell Transformation and Renal Vessel Hypertrophy during Angiotensin II-Induced Hypertension. Am. J. Physiol. Ren. Physiol. 2008, 294, F161–F169. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  160. Vonend, O.; Turner, C.M.; Chan, C.M.; Loesch, A.; Dell’Anna, G.C.; Srai, K.S.; Burnstock, G.; Unwin, R.J. Glomerular Expression of the ATP-Sensitive P2X Receptor in Diabetic and Hypertensive Rat Models. Kidney Int. 2004, 66, 157–166. [Google Scholar] [CrossRef] [Green Version]
  161. Díaz, E.F.; Labra, V.C.; Alvear, T.F.; Mellado, L.A.; Inostroza, C.A.; Oyarzún, J.E.; Salgado, N.; Quintanilla, R.A.; Orellana, J.A. Connexin 43 hemichannels and pannexin-1 channels contribute to the α-synuclein-induced dysfunction and death of astrocytes. Glia 2019, 67, 1598–1619. [Google Scholar] [CrossRef]
  162. Pelegrin, P.; Surprenant, A. Pannexin-1 Couples to Maitotoxin- and Nigericin-Induced Interleukin-1β Release through a Dye Uptake-Independent Pathway. J. Biol. Chem. 2007, 282, 2386–2394. [Google Scholar] [CrossRef] [Green Version]
  163. Li, G.; Zhang, Q.; Hong, J.; Ritter, J.K.; Li, P.-L. Inhibition of Pannexin-1 Channel Activity by Adiponectin in Podocytes: Role of Acid Ceramidase Activation. Biochim. Biophys. Acta (BBA) Mol. Cell Biol. Lipids 2018, 1863, 1246–1256. [Google Scholar] [CrossRef]
  164. Kang, J.; Kang, N.; Lovatt, D.; Torres, A.; Zhao, Z.; Lin, J.; Nedergaard, M. Connexin 43 Hemichannels Are Permeable to ATP. J. Neurosci. 2008, 28, 4702–4711. [Google Scholar] [CrossRef] [Green Version]
  165. Abed, A.B.; Kavvadas, P.; Chadjichristos, C.E. Functional Roles of Connexins and Pannexins in the Kidney. Cell Mol. Life Sci. 2015, 72, 2869–2877. [Google Scholar] [CrossRef]
Figure 1. Primary causes and morphological outcomes of chronic kidney disease. Although the leading causes of chronic kidney disease include diabetes and hypertension, other disorders such as glomerulonephritis, cystic kidney disease, and diverse urologic diseases also contribute in a minor proportion. Regardless of the etiology, the progressive reduction in glomerular filtration rate occurs accompanied by two common histological changes: glomerular sclerosis and tubular necrosis. Elaborated in BioRender.com (accessed on 15 October 2022).
Figure 1. Primary causes and morphological outcomes of chronic kidney disease. Although the leading causes of chronic kidney disease include diabetes and hypertension, other disorders such as glomerulonephritis, cystic kidney disease, and diverse urologic diseases also contribute in a minor proportion. Regardless of the etiology, the progressive reduction in glomerular filtration rate occurs accompanied by two common histological changes: glomerular sclerosis and tubular necrosis. Elaborated in BioRender.com (accessed on 15 October 2022).
Ijms 23 15936 g001
Figure 2. Schematics showing general aspects of the generation of chronic kidney disease mediated by hypertensive nephropathy. At the renal level, arterial hypertension increases intra-glomerular pressure, leading to reductions in renal blood flow and alterations in the inflammatory profile of mesangial cells, podocytes, and epithelial cells. The epithelial cells boost the production of vasoactive and pro-inflammatory mediators, triggering a reduction in glomerular permeability and filtration. Consequently, hypertensive nephropathy can contribute to CKD. Renal impairment would then be associated mainly with the persistent production of AngII, which could augment the expression of proinflammatory cytokines [IL-1β and TNF-α] and the NOX-mediated generation of ROS. These processes likely result in macrophage infiltration and tubular overexpression of osteopontin, favoring the production of TGF- β1 and activation of NF-κB and AP-1. Elaborated in BioRender.com (accessed on 15 October 2022).
Figure 2. Schematics showing general aspects of the generation of chronic kidney disease mediated by hypertensive nephropathy. At the renal level, arterial hypertension increases intra-glomerular pressure, leading to reductions in renal blood flow and alterations in the inflammatory profile of mesangial cells, podocytes, and epithelial cells. The epithelial cells boost the production of vasoactive and pro-inflammatory mediators, triggering a reduction in glomerular permeability and filtration. Consequently, hypertensive nephropathy can contribute to CKD. Renal impairment would then be associated mainly with the persistent production of AngII, which could augment the expression of proinflammatory cytokines [IL-1β and TNF-α] and the NOX-mediated generation of ROS. These processes likely result in macrophage infiltration and tubular overexpression of osteopontin, favoring the production of TGF- β1 and activation of NF-κB and AP-1. Elaborated in BioRender.com (accessed on 15 October 2022).
Ijms 23 15936 g002
Figure 3. Structural organization of connexin and pannexin-based channels. Connexins and pannexins have four transmembrane domains (TM), two extracellular loops (EL), one cytoplasmatic loop (CL), and both the N- and C-terminus are localized on the intracellular side. The glycosylated extracellular loop of pannexins is also shown. Hemichannels or connexons are formed by the oligomerization of six subunits of connexins around a central pore, while pannexons are composed of seven pannexin subunits in the case of Panx1. Under normal conditions, the opening of hemichannels and pannexons is tightly regulated to fulfill several biological processes. Hemichannels and likely pannexons can also dock each other in junctional membrane interfaces to form cell-to-cell intercellular channels, termed gap junction channels. Elaborated in BioRender.com (accessed on 15 October 2022).
Figure 3. Structural organization of connexin and pannexin-based channels. Connexins and pannexins have four transmembrane domains (TM), two extracellular loops (EL), one cytoplasmatic loop (CL), and both the N- and C-terminus are localized on the intracellular side. The glycosylated extracellular loop of pannexins is also shown. Hemichannels or connexons are formed by the oligomerization of six subunits of connexins around a central pore, while pannexons are composed of seven pannexin subunits in the case of Panx1. Under normal conditions, the opening of hemichannels and pannexons is tightly regulated to fulfill several biological processes. Hemichannels and likely pannexons can also dock each other in junctional membrane interfaces to form cell-to-cell intercellular channels, termed gap junction channels. Elaborated in BioRender.com (accessed on 15 October 2022).
Ijms 23 15936 g003
Figure 4. Schematic drawing of the localization of connexin and pannexin isoforms in the kidney. AA, afferent arteriole; EA, efferent arteriole; JGC, juxtaglomerular cells; Po, podocytes; Mes, mesangial cells; PT, proximal tubules; DLH, descending limb of loop of Henle; ALH, ascending limb of loop of Henle; DT, distal tubules; CT, collecting tubules; and Vr, Vasa recta. Elaborated in BioRender.com (accessed on 15 October 2022).
Figure 4. Schematic drawing of the localization of connexin and pannexin isoforms in the kidney. AA, afferent arteriole; EA, efferent arteriole; JGC, juxtaglomerular cells; Po, podocytes; Mes, mesangial cells; PT, proximal tubules; DLH, descending limb of loop of Henle; ALH, ascending limb of loop of Henle; DT, distal tubules; CT, collecting tubules; and Vr, Vasa recta. Elaborated in BioRender.com (accessed on 15 October 2022).
Ijms 23 15936 g004
Figure 5. Scheme of possible signaling pathways involved in regulating of Cx43 hemichannels and Panx1 channels in mesangial cells stimulated with AngII. High AngII concentrations (10−7 M) activate angiotensin type 1 receptors (AT1R) in MCs, causing the PLC-mediated stimulation of IP3 receptors and further release of Ca2+ stored in the endoplasmic reticulum, NF-κB-dependent release of proinflammatory cytokines such as TNF-α and IL-1β, as well as the formation of reactive oxygen species (ROS). The resulting increase in [Ca2+]i can also stimulate Panx1 channels and Cx43 hemichannels, eliciting the subsequent release of ATP to the extracellular compartment. Then, ATP activates P2X7 and P2Y1 receptors that, together with Cx43 hemichannels, permit a drastic increase in Ca2+ influx. As already seen in other systems, the increase in [Ca2+]i promotes the expression and release of pro-inflammatory cytokines such as TNF-α, IL-1β via the activation of the inflammasome, and the generation of ROS. The release of ATP and influx of Ca2+ establish a positive feedback loop. Of note, alterations in [Ca2+]i homeostasis mediated by Cx43 hemichannels or Panx1 channels could affect diverse aspects of MCs function (e.g., morphology and pro-inflammatory profile). Solid lines depict fluxes of molecules through channels, whereas dashed lines indicate activation or induction. Elaborated in BioRender.com (accessed on 15 October 2022).
Figure 5. Scheme of possible signaling pathways involved in regulating of Cx43 hemichannels and Panx1 channels in mesangial cells stimulated with AngII. High AngII concentrations (10−7 M) activate angiotensin type 1 receptors (AT1R) in MCs, causing the PLC-mediated stimulation of IP3 receptors and further release of Ca2+ stored in the endoplasmic reticulum, NF-κB-dependent release of proinflammatory cytokines such as TNF-α and IL-1β, as well as the formation of reactive oxygen species (ROS). The resulting increase in [Ca2+]i can also stimulate Panx1 channels and Cx43 hemichannels, eliciting the subsequent release of ATP to the extracellular compartment. Then, ATP activates P2X7 and P2Y1 receptors that, together with Cx43 hemichannels, permit a drastic increase in Ca2+ influx. As already seen in other systems, the increase in [Ca2+]i promotes the expression and release of pro-inflammatory cytokines such as TNF-α, IL-1β via the activation of the inflammasome, and the generation of ROS. The release of ATP and influx of Ca2+ establish a positive feedback loop. Of note, alterations in [Ca2+]i homeostasis mediated by Cx43 hemichannels or Panx1 channels could affect diverse aspects of MCs function (e.g., morphology and pro-inflammatory profile). Solid lines depict fluxes of molecules through channels, whereas dashed lines indicate activation or induction. Elaborated in BioRender.com (accessed on 15 October 2022).
Ijms 23 15936 g005
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Lucero, C.M.; Prieto-Villalobos, J.; Marambio-Ruiz, L.; Balmazabal, J.; Alvear, T.F.; Vega, M.; Barra, P.; Retamal, M.A.; Orellana, J.A.; Gómez, G.I. Hypertensive Nephropathy: Unveiling the Possible Involvement of Hemichannels and Pannexons. Int. J. Mol. Sci. 2022, 23, 15936. https://doi.org/10.3390/ijms232415936

AMA Style

Lucero CM, Prieto-Villalobos J, Marambio-Ruiz L, Balmazabal J, Alvear TF, Vega M, Barra P, Retamal MA, Orellana JA, Gómez GI. Hypertensive Nephropathy: Unveiling the Possible Involvement of Hemichannels and Pannexons. International Journal of Molecular Sciences. 2022; 23(24):15936. https://doi.org/10.3390/ijms232415936

Chicago/Turabian Style

Lucero, Claudia M., Juan Prieto-Villalobos, Lucas Marambio-Ruiz, Javiera Balmazabal, Tanhia F. Alvear, Matías Vega, Paola Barra, Mauricio A. Retamal, Juan A. Orellana, and Gonzalo I. Gómez. 2022. "Hypertensive Nephropathy: Unveiling the Possible Involvement of Hemichannels and Pannexons" International Journal of Molecular Sciences 23, no. 24: 15936. https://doi.org/10.3390/ijms232415936

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