Next Article in Journal
Distinct Effects of Beta-Amyloid, Its Isomerized and Phosphorylated Forms on the Redox Status and Mitochondrial Functioning of the Blood–Brain Barrier Endothelium
Next Article in Special Issue
New Frontiers on ER Stress Modulation: Are TRP Channels the Leading Actors?
Previous Article in Journal
IL-20 Activates ERK1/2 and Suppresses Splicing of X-Box Protein-1 in Intestinal Epithelial Cells but Does Not Improve Pathology in Acute or Chronic Models of Colitis
Previous Article in Special Issue
Modulation of Glia Activation by TRPA1 Antagonism in Preclinical Models of Migraine
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

TRPC Channels in the Physiology and Pathophysiology of the Renal Tubular System: What Do We Know?

by
Colya N. Englisch
1,
Friedrich Paulsen
2 and
Thomas Tschernig
1,*
1
Institute of Anatomy and Cell Biology, Saarland University, 66421 Homburg/Saar, Germany
2
Institute of Functional and Clinical Anatomy, Friedrich Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(1), 181; https://doi.org/10.3390/ijms24010181
Submission received: 29 October 2022 / Revised: 12 December 2022 / Accepted: 17 December 2022 / Published: 22 December 2022
(This article belongs to the Special Issue TRP Channels in Physiology and Pathophysiology)

Abstract

:
The study of transient receptor potential (TRP) channels has dramatically increased during the past few years. TRP channels function as sensors and effectors in the cellular adaptation to environmental changes. Here, we review literature investigating the physiological and pathophysiological roles of TRPC channels in the renal tubular system with a focus on TRPC3 and TRPC6. TRPC3 plays a key role in Ca2+ homeostasis and is involved in transcellular Ca2+ reabsorption in the proximal tubule and the collecting duct. TRPC3 also conveys the osmosensitivity of principal cells of the collecting duct and is implicated in vasopressin-induced membrane translocation of AQP-2. Autosomal dominant polycystic kidney disease (ADPKD) can often be attributed to mutations of the PKD2 gene. TRPC3 is supposed to have a detrimental role in ADPKD-like conditions. The tubule-specific physiological functions of TRPC6 have not yet been entirely elucidated. Its pathophysiological role in ischemia-reperfusion injuries is a subject of debate. However, TRPC6 seems to be involved in tumorigenesis of renal cell carcinoma. In summary, TRPC channels are relevant in multiples conditions of the renal tubular system. There is a need to further elucidate their pathophysiology to better understand certain renal disorders and ultimately create new therapeutic targets to improve patient care.

1. Introduction

The transient receptor potential (TRP) channels are key sensors and effectors in cellular adaptation to environmental stress [1]. The 29 channels are subdivided into seven subfamilies. Canonical TRPC1-7, vanilloid-receptor TRPV1-6, melastatin TRPM1-8, ankyrin TRPA1, polycystin TRPP2/3/5, mucolipin TRPML1-3, and no mechanoreceptor potential C TRPN1 subfamilies are defined [2]. TRPC channels are tetramers and belong to the nonselective cation channels. They are transiently permeable for both monovalent and divalent cations including Na+, K+, Ca2+, or Zn2+. Monomeric subunits assemble to form the tetrameric structure. The subunits can belong to the same or to different TRPC entities. Based on amino acid sequences and functional analogies, three subgroups can be distinguished in humans, which preferably heteropolymerize among themselves—TRPC1, TRPC4/5, and TRPC3/6/7 [3]. Six transmembrane segments (S1–S6) are linked to form a TRPC monomer. The cation-permeable pore is generated by the S5 and S6 segments of each subunit [4]. The regulation of the gating activity is subject to a diversity of mechanisms, often related to Gq/11- and receptor tyrosine kinase-associated phospholipase C pathways, Gi and Go proteins as well as intracellular Ca2+ stores [3,5]. The protein kinase C can for example directly activate TRPC channels through phosphorylation [6]. Both, mechanical and oxidative stress can also alter TRPC gating behavior [7,8,9]. DAG sensitivity (1,2-diacylglycerine) is a key feature enabling phospholipase C pathways to impact the activity of TRPC3, TRPC6, and TRPC7 [10]. Furthermore, low basal concentrations of DAG can potentially be empowered by pharmacological allosteric modulation of TRPC6 to activate the channel [11]. In contrast, TRPC1, TRPC4, and TRPC5 are insensitive to DAG [6]. TRPC4, however, is indirectly activated by phosphodiesterase-5-inhibitors as demonstrated using human embryonic kidney 293 and prostate smooth muscle cell lines. Cyclic guanosine-monophosphate (cGMP) is degraded by the phosphodiesterase-5. When the latter is inhibited, cGMP can stimulate the protein kinase G (PKG) which in turn phosphorylates and activates TRPC4 ultimately resulting in an increased cytosolic [Ca2+] [12]. In addition, TRPC channels play a critical role in inflammation. For instance, TRPC6 is upregulated in microglia by amyloid β-protein in a nuclear factor κ-light-chain-enhancer of activated B-cells (NF-κB)-dependent manner [13]. On the other hand, upregulated TRPC6 channels inhibit the signal transducers and activators of transcription (STAT) signaling and promote proliferative and inflammatory processes in tubular cells in diabetic nephropathy [14]. The role of TRPC6 in diabetic nephropathy is reviewed in [15,16]. In bronchial epithelial cells, TRPC6 is overexpressed after lipopolysaccharide (LPS) exposure and subsequent Toll-like receptor 4 (TLR-4)/phosphatidylinositol 3 kinase (PI3K)/ protein kinase B (Akt) signaling. The following TRPC6-dependent activation of ERK1/2, p38 and NF-κB triggers a cytokine-associated inflammatory response [17].
Several domains enrich the cytoplasmic TRPC-monomer-termini enabling interaction with a spectrum of molecular players. A coiled-coil domain and four ankyrin domains are localized at the NH2-terminus. They are involved in tetramerization of TRPC subunits and thus in the regulation of TRPC channel function. The TRP domain is localized at the COOH-terminus and is the linking site for other TRP-channel isoforms. The COOH-terminus further includes a coiled-coil domain and a calmodulin and IP3-R binding site, which regulates store-operated channel activation [7,18] (Figure 1).
The last domain is involved in the positive regulation of TRPC6 through Ca2+/calmodulin (CaM)-dependent kinase II in the cardiovascular system [21]. Elevated cytosolic [Ca2+] can activate CaM kinases which in turn may further enhance Ca2+ influx through activation of Ca2+-permeable channels including TRPC6 [21]. This is a mechanism that can potentially impact both physiological and pathophysiological conditions in different tissues including the renal tubular system. Indeed, several TRPC channels are deeply involved in Ca2+ signaling which can result in cell proliferation, cell migration, etc. [18]. Certain TRPC members are also involved in receptor operated Ca2+ entry (ROCE) and store operated Ca2+ entry (SOCE), with both mechanisms mediating regulated Ca2+ influx [18]. Phospholipase C cleaves PIP2 (phosphatidylinositol-4,5-bisphosphat) in DAG (1,2-diacylglycerine) and IP3 (inositol-1,4,5-triphosphate) which are critical in ROCE and SOCE, respectively. DAG can directly activate members of the TRPC subfamily as previously mentioned, whereas IP3 binds to the endoplasmic IP3-receptor inducing Ca2+ release from the endoplasmic reticulum (ER). The resulting ER depletion is sensed by stromal interaction protein 1 (STIM1) which interacts with Orai1 (calcium release-activated calcium channel protein 1) mediating store-operated Ca2+ entry. TRPC1 also plays an important role as store-operated channel while further TRPC channels regulate Orai1 and ultimately SOCE [6,18,22,23].
The nephron is the functional unit of renal physiology. The nephron is subdivided into a renal corpuscle—the compound of glomerulus and Bowman’s capsule—and a tubular system composed of proximal, intermediate, and distal tubules that drain into a collecting duct. The primary urine is gained at the glomerular filtration barrier and transformed into a secondary urine through many different reabsorbing tubular mechanisms [24] (Figure 2).
The renal tubular system underlies several hormonic loops such as the renin-angiotensin-aldosterone-system (RAAS) or the vasopressin system and is essential in acid–base and ion–water homeostasis [26,27]. From a pathophysiological point of view, substantial damage can arise from impaired tubular function. For instance, highly metabolic-active proximal tubule cells can be damaged by multiple conditions leading to acute kidney injury [28,29]. Additionally, a disturbed Ca2+ reabsorption can promote crystal precipitation with concomitant nephrolithiasis leading to inflammation and fibrosis and eventually resulting in chronic kidney disease [30,31,32]. Furthermore, malignant processes such as renal cell carcinoma originate from the tubular system [33]. In short, the tubular system is not only crucial for renal physiology, but also for a multitude of pathophysiological processes that remain challenging today, as most of them cannot yet be treated satisfactorily.
Ever since evidence suggested that mutations in the TRPC6 gene could lead to focal and segmental glomerulosclerosis (FSGS) [34], the research in renal TRPC channels has dramatically increased. The relationship between TRPC6 and glomerular permselectivity and its involvement in renal proteinuric disorders has been of special interest [35]. However, TRPC6 is not only found in the glomeruli, but also in the renal tubular system [36]. In the last few years, more and more studies have investigated TRPC channels—especially TRPC3 and TRPC6—in the renal tubules. However, the role of TRPC6 in tubular physiology remains insufficiently studied. It has been suggested that TRPC6 could promote flow-stimulated generation of endothelin-1—an autocrine inhibitor of sodium and water reabsorption—in cortical collecting ducts [37,38]. The role of TRPC channels in glomeruli and their involvement in proteinuric, diabetic, and chronic kidney diseases but also in renal fibrosis is summarized elsewhere [15,16,35,38,39]. We present here a summary of the implication of TRPC channels in both the specific physiology and the specific pathophysiology of the renal tubular system.

2. TRPC6 Is a Controversial Player in Tubular Cells Experiencing Ischemia-Reperfusion Injuries

Several conditions, such as acute hemorrhage or toxic shock, can cause renal ischemia-reperfusion injury (RIRI). RIRI is characterized by massive tissue damage and is a frequent cause of acute kidney injuries [40]. Acute kidney injuries can be defined as a 1.5-fold baseline increase in serum creatinine over the preceding seven days [41]. The generation of reactive oxygen species (ROS), Ca2+ overload, and immune responses are key factors in the promotion of tubular damage in acute kidney injury following RIRI [42]. Tubular injury is also considered as a driving force towards chronic kidney disease (CKD) [43]. Proximal tubular cells have an active metabolism and are therefore especially endangered by the oxidative stress that can occur during ischemia-reperfusion (I/R) [29,44,45,46,47,48]. Interrupted perfusion, for instance caused by thromboembolism, leads to a transition from aerobic to anaerobic cell metabolism and concomitant impaired ATP production (adenosine-triphosphate). This in turn is associated with acidification and an intracellular [Na+] and extracellular [K+] increase. Subsequently, membrane depolarization with compensatory Ca2+ influx induces the activation of proteases which contribute to the resulting cell death. Sudden reperfusion with concomitant reoxygenation creates a massive release of reactive oxygen species (ROS) [49]. ROS comprise free radicals, oxygen anions, and hydrogen peroxide [50]. The oxidative burst heavily damages tissues—cytoprotective ROS scavengers being disabled after ischemia-reperfusion—leading to different forms of cell death including apoptosis, necrosis, necroptosis, pyroptosis, ferroptosis, etc. [51]. Both ischemia and reperfusion contribute to the heavy tissue damage [51,52,53,54,55,56,57,58,59,60,61,62,63]. In 2013, a bioinformatic analysis of rat samples showed that TRPC6 was upregulated in RIRI-damaged tissues compared to the control [64]. Further investigations supported the upregulation of TRPC6 in RIRI [65]. The literature is not concordant, as the downregulation of TRPC6 has also been observed in I/R tissue compared to sham tissue [66,67]. ROS—released during RIRI—are involved in both the regulation of TRPC6-expression and -gating activity as well as the initiation of autophagy [8,50,68]. The altered Ca2+ signaling, which is mediated by the redox-sensitive TRPC6 channel, is involved in ROS-caused renal injury [8,9]. Autophagy is a dynamic recycling cellular process decomposing cell components in a lysosomal environment which is induced by the formation of autophagosomes in response to oxidative stress in ischemia-reperfusion injuries [69,70,71,72,73]. It is rapidly enhanced after reperfusion, ultimately deferring the increase of apoptotic activity in RIRI. The densitometric analysis of microtubule-associated proteins 1A/1B-light chain 3 (LC3-II/LC3-I), p62 and B-cell lymphoma 2 (Bcl-2)/Bax blots combined with hematoxylin and eosin, and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining revealed that the inhibition of autophagy using the PI3K-inhibitor 3-methyladenine (3-MA) aggravated tissue damage [40]. These results contribute to the concept of a cytoprotective function of autophagy in RIRI.
Hou et al. suggested that TRPC6-mediated Ca2+ influx regulates autophagic flux in proximal tubule cells undergoing oxidative stress through H2O2 exposure. Indeed, both basal as well as oxidative stress-associated autophagy can be decreased or increased by respective overexpression or silencing of TRPC6 [69]. Furthermore, apoptosis is attenuated in TRPC6 SAR7334-silenced proximal tubule cells after H2O2 exposure. Additionally, the mitochondrial permeability transition positive cells—hallmarks of ROS injury—were significantly diminished after SAR7334 silencing of TRPC6. Additional evidence was obtained suggesting that post-oxidative stress apoptosis is attenuated by increased autophagic flux through TRPC6 silencing [69]. Moreover, the involvement of the PI3K/Akt/mTOR and ERK1/2 pathways in TRPC6-driven autophagy inhibition after ROS exposure has been demonstrated. A mechanism of TRPC6-mediated Ca2+ signaling resulting in phosphorylation of Akt and ERK preventing autophagy but enhancing apoptosis in response to ROS, was proposed. However, contradicting evidence has appeared indicating that cell autophagy is promoted by I/R injury and accelerated in tubular epithelial HK-2 cells overexpressing TRPC6 [74]. The suggestion of Hou et al. was not refuted by Shen et al. who observed that apoptosis is not affected by TRPC6 [75]. In turn, they suggested that TRPC6 may inhibit necroptosis and thereby play a protective role in RIRI [75]. Restoration of TRPC6 expression—which was reduced after ischemic injury—was awarded a role in alleviating RIRI-induced acute tubular injury after erythropoietin premedication in collecting ducts [38,76]. Moreover, necrostatin-1, a receptor-interacting protein kinase-1-inhibitor (RIP1), was shown to alleviate RIRI; an effect which may be mediated by the hypoxia-inducible factor-1α (HIF-1α/miR-26a/TRPC6/poly (ADP-ribose) polymerases 1 (PARP1) pathway [67]. Shin et al. showed that L-ornithine-dependent activation of the calcium sensing receptor can decrease ROS generation and prevent H2O2-induced necrosis through TRPC-dependent ROCE in proximal tubular cells and thereby alleviate acute kidney injury [77]. Interestingly, another recent study showed no difference in renal function and tubular damage among TRPC6-/- mice, TRPC6-inhibitor treated mice, and wild type mice after RIRI [42]. TRPC6 inhibition did not impact the short-term outcome of acute kidney injury [42].
Aside from investigating the role of TRPC6, the function of Zn2+ in RIRI was also studied. Zinc is a trace element which is essentially involved in multiple physiological processes of the organism [78]. TRPC6 markedly contributes to transmembrane Zn2+-transportation and Zn2+ itself plays a key role in autophagy [66,79,80,81]. The Zn2+ content is increased in cells with ischemia and reperfusion. TRPC6 knockdown or TRPC6 overexpression in oxygen-glucose deprived and reoxygenated (OGD-R) human kidney-2 (HK-2) cells would respectively lead to a decrease or an increase in Zn2+-flow—the latter concomitantly with an augmented autophagic flux. Zn2+ significantly ameliorated the viability of OGD-R HK-2 cells and decreased the controlled necrosis rate [74]. A recent study revealed the role of TRPC6 and Zn2+ in inhibiting pyroptosis of tubular epithelial cells and thereby attenuating RIRI [66]. Pyroptosis is a form of programmed necrotic cell death which is induced by NLRP3 (nucleotide-binding and oligomerization domain-like receptors (NLR) family pyrin domain containing 3) associated caspase-1 activating gasdermin D to form pores in the plasma membrane resulting in proinflammatory cell lysis [82]. Interestingly, insufficient Zn2+ levels are thought to mediate activation of NLRP3 inflammasome after ROS exposure and subsequently induce pyroptosis [66,83]. Both animal RIRI models and OGD-R HK-2 cells were used to provide evidence that TRPC6 inhibition augmented pyropoptic activity and exacerbated renal injury in RIRI. Zn2+ influx and upregulation of the zinc finger protein A20 inhibiting the activation of NF-κB, which plays a key role in NLRP3 activation, seem to control the cytoprotective and antipyropoptic effects of TRPC6 in RIRI [66,84].
In summary, we can conclude that TRPC6 is involved in Ca2+ and Zn2+ signaling in RIRI. The relevant literature is not concordant on the function of tubular TRPC6 in RIRI. However, Ca2+ entry channels such as TRPC6 may have dual roles in renal epithelial cells [85]. Further studies are needed to clarify this discussion in the context of RIRI. A better understanding of the function of TRPC6 in RIRI is important as it may lead to targeted drug development.

3. TRPC6 Drives Tumorigenesis and the Progression of Renal Cell Carcinoma

Unsettled Ca2+ signaling is often involved in tumorigenesis [86,87]. Renal cell carcinoma (RCC) is a very common cancer affecting the kidney [88]. More than 300,000 new cases of RCC are reported worldwide yearly [89,90]. Different subtypes of renal cell carcinoma exist. The clear cell entity (70–80%) is the most common type of RCC, directly followed by the papillary type (15%) [91,92]. Both are originated from the proximal tubule [33]. From a biological and clinical point of view, clear cell renal cell carcinoma (ccRCC) and non-ccRCC are completely different pathologies and, therefore, so is the respective tumor-specific therapy. The single curative therapy in many localized cases is often surgical removal. However, studies indicate that 30% of patients who had been considered healed suffered a relapse [88]. Classical chemotherapy or radiotherapy treatments are not able to be used to treat RCC [90]. The focus needs to be placed on specific tumor biology, microenvironments, or vascularization [90]. Unfortunately, the exact pathogenesis of the different renal cell carcinoma types is not well understood. Nevertheless, the Ca2+ permeable TRPC6 channel is supposed to be implicated in receptor-operated Ca2+ entry of RCC cells. In addition, TRPC6 expression is by far the most increased compared to TRPC3, TRPC4 or TRPC5 expression in ccRCC tissue [93]. Immunohistochemistry has shown that TRPC6 reactivity was significantly stronger in RCC tissue compared to healthy tissue [94]. The tumor nuclear grading according to Fuhrmann was positively correlated with the amount of detected TRPC6, suggesting a significant function of TRPC6 in tumorigenesis and tumor progression [94]. Song et al. investigated the effects of inhibiting TRPC6 in ACNH cells—a cell line initiated in 1979 from the malignant pleural effusion of a 22-year-old man in the context of a metastatic renal cell carcinoma [95]—and revealed a significantly decreased hepatocyte growth factor-induced (HGF) cell proliferation. Additionally, TRPC6-mRNA inhibition via siRNA3 transfection led to an increased transition time through the G2/M phase of mitosis in ACHN cells, eventually providing sufficient time to ensure efficient DNA-repair machinery [94]. In general, TRPC6-gated Ca2+ influx has been shown to be critical in the transition of the G2/M phase in several different cell types [96]. HGF is a pleiotropic glycoprotein—able to increase TRPC6 expression in ACHN cells—that stimulates the c-met signaling which is a major player in tumorigenesis, progression, and vascularization of papillary RCC [92,97,98]. The MET signaling also mediates VEGF resistance in ccRCC [92]. Indeed, in the context of hereditary papillary RCC, a gain-of-function mutation of the HGF-tyrosine kinase membrane receptor MET (mesenchymal-epithelial transition factor) leads to uncontrolled cancer-promoting effects [99]. Subsequently, Kim et al. investigated the relevance of the lysine-deficient protein kinase 1-promoted (WNK1) TRPC6-NFAT (nuclear factor of activated T-cells) pathway in the development of ccRCC. WNK1 controls the tubular electrolyte homeostasis. Therefore, it regulates the distribution of several ion channels and transporters through various signaling cascades involving effectors such as the STE20-proline alanine rich kinase (SPAK) and oxidative stress responsive kinase 1 (OSR1) but also the mitogen-activated protein kinase (MAPK) [100]. Hence, the impairment of WNK1 function can lead to pseudo hypoaldosteronism type two [101,102]. Furthermore, evidence has emerged uncovering the key role of WNK1 in tumorigenesis [103]. In the context of ccRCC, upregulated WNK1 stimulates the phosphatidylinositol 4-kinase IIIα (PI4KIIIα) enzyme, which controls the phosphatidylinositol-4,5-diphosphate-dependent PLC-β signaling leading to DAG-mediated activation of TRPC6. The WNK1-TRPC6 pathway activates the NFATc1 signaling which in turn has been suggested to enhance WNK1 and TRPC6 expression in ccRCC cell lines. Consistent with these reports, the c-Myc gene, which is regulated by NFATc1 signaling is often overexpressed in RCC [104]. Furthermore, Kim et al. showed that the WNK1-activated TRPC6 is an important player in receptor-operated Ca2+ influx in ccRCC cell lines such as Caki1 and ACHN. Functional analysis has revealed that the colony-forming ability of ACHN and Caki1 cells was reduced in knockdown models of TRPC6, WNK1, and PI4KIIIα. The number of cells was also decreased. Inhibition of the TRPC6-NFATc1 pathway markedly diminished the survival as well as the proliferation of ccRCC cells. These results, among others, support the suggestion that the WNK1-driven TRPC6-NFATc1 pathway is a key component in the proliferation and migration of ccRCC cells [93]. In contrast, studies on human metastatic renal cell carcinoma cultures have demonstrated that TRPC6 is involved in SOCE and that SOCE inhibition did not impact cell proliferation [105]. In addition to TRPC6, TRPC1 deserves to be mentioned in the context of tubular players in RCC. TRPC1 is crucial for the polarity and the directionality of migrating cells including cancer cells [18]. A recent study investigated the TRPC1 expression in ccRCC tissue and demonstrated a positive correlation between the TRPC1-expression level and the tumor grade. The authors postulated that TRPC1 might enhance cell proliferation via Ca2+ entry and Ca2+-NFATc3 signaling pathways leading to ccRCC growth which is concomitant with higher tumor grading. However, the relevance of TRPC1 was limited to biomarking TNM stages and indicating long-term prognosis of RCC [106].
In summary, TRPC6 is a critical factor in tumorigenesis and tumor progression of different RCC entities [93]. Inhibitors of the downstream effector MAPK of WNK1 have been proposed for the treatment of RCC [107]. Similarly, WNK1 or TRPC6, but also MET in the papillary entity [92], may be new targets in the antiproliferative therapy of RCC.

4. TRPC3 Is a Cytoprotective Key Player in Ca2+ Reabsorption of the Proximal Tubule

TRPC3 is expressed in the proximal tubule and the collecting duct [31,108,109,110]. Approximatively 65–70% of the tubular calcium reabsorption is performed in the proximal tubule [111]. Paracellular mechanisms predominate. A difficulty to explain mechanisms resolving a sudden increase in luminal [Ca2+], has remained [109]. For this reason, an inducible transcellular pathway might be superior to simple paracellular osmotic and diffusion processes. The apical compound of calcium sensing receptors (CaSR)—a class three G-protein-coupled receptor—and TRPC3 has been proposed to play a critical role in transcellular Ca2+ reabsorption in proximal tubule cells [109,112]. The CaSR—expressed in the gut, kidneys, and parathyroid gland—is a main component of extracellular Ca2+ homeostasis and can activate both SOCE and ROCE pathways in the proximal tubule [109,113,114]. Nevertheless, ROCE remains in large parts responsible for Ca2+ entry in proximal tubule cells [109]. An alkaline hypercalciuric environment conditions a switch from ROCE to SOCE in TRPC3-deficient proximal tubule cells [31,32,115] (Figure 3).
The subsequent excess of intracellular [Ca2+] can lead to ER stress (endoplasmic reticulum) and ROS production [32,115,116]. NPS-2143, a CaSR inhibitor, reduced SOCE, ROS generation, and ER stress in TRPC3-deficient proximal tubule cells. This argues for a cytoprotective function of the CaSR-dependent TRPC3 activation as the SOCE-associated downstream cascade of injuring events following hypercalciuria is diminished. Excessive luminal [Ca2+] can activate the CaSR and initiate the phospholipase C signaling. The resulting DAG messaging can enhance TRPC3-gated Ca2+ influx in the proximal tubule. Hypercalciuria and subsequent calcium-phosphate crystal formation in the loop of Henle can thereby be limited [109,117]. Basolateral Ca2+-efflux mediators such as the plasma membrane Ca2+-ATPase 1 (PMCA1) or the Na+/Ca2+-exchanger 1 (NCX1) complete the concept of a transepithelial calbindin-mediated Ca2+ reabsorption process in the proximal tubule [109] (Figure 3). After oral calcium gluconate administration, TRPC3-gating augmented altering luminal [Ca2+] [32]. The critical role of TRPC3 in Ca2+ reabsorption is supported by the development of hypercalciuria after TRPC3 knockout [109]. Even though unlikely, CaSR-TRPC3 activation may also reduce luminal [Ca2+] by increasing the tight-junction associated paracellular Ca2+ permeability [109]. Hypercalciuria is the basement for calcium-phosphate crystal nucleation that displays the preliminary stage of both calcium phosphate and mixed stone formation [117,118,119,120,121]. This sequence—summarized as lithogenesis—is boosted in alkaline milieu. The CaSR can be sensitized by luminal alkalization eventually enhancing an increased Ca2+ reabsorption [31,122]. These circumstances were used to facilitate crystal nucleation in experimental designs after acetazolamide administration [31]. Acetazolamide inhibits proximal tubular carbonic anhydrases—essential components in the renal acid–base balance—provoking metabolic acidosis with concomitant tubular alkalization and facilitated crystal formation [123]. Subsequent tubular crystal uptake can activate the NF-κB—NLRP3—IL-1β pathway triggering IL-6 and TGF-β1 secretion which are key factors in advancing renal fibrosis and inflammation [32,124,125,126,127]. Tubular fibrosis and inflammation are exacerbated by TRPC3 knockdown as suggested by histology and increased fibrotic (TGF-β1, FN-1 and SMa) and inflammatory (IL-1β, IL-6, monocyte chemoattractant protein-1 (MCP1), NF-κB and NLRP3) markers [31,128]. The NF-κB pathway is also associated with ER-stress-induced apoptosis [129,130]. Hypercalciuric conditions obtained after the calcium gluconate treatment increased the expression of ER-stress-related genes such as C/EBP homologous protein and M18S [32]. Similarly, the apoptotic activity of proximal tubule cells was increased, especially when TRPC3 was silenced [32]. Disordered extracellular Ca2+ concentration can also evoke responsive ROS production driving oxidative cellular injury leading to apoptosis, fibrosis, inflammation, etc., which are concomitant with a decreasing renal function [31,32,131,132,133,134,135]. The resulting cellular debris promote lithogenesis creating a vicious circle [136,137]. TRPC3 may therefore contribute to a postponed and decelerated development of CKD in the context of nephrocalcinosis and -lithiasis [31].
In summary, TRPC3 is critically involved in Ca2+ reabsorption in the proximal tubule and its impaired expression can contribute to hypercalciuria and through crystal formation and calcification support both fibrosis and inflammation which can result in acute and chronic kidney disease [115]. Since proximal tubular injury and the crystal formation were exacerbated by TRPC3 deficiency, it is legitimate to attribute a preventive role to TRPC3 in hypercalciuria-induced crystal formation and tubular injury by reabsorption of excess luminal calcium.

5. TRPC3 Is Involved in Vasopressin-Dependent AQP-2 Trafficking, Osmosensation, and Ca2+ Reabsorption in the Collecting Duct

Since Khayyat et al. extensively reviewed the function of TRPC3 in the kidney in 2020 [110], there has been very little research performed on the topic. We will therefore only briefly report the most important findings for the sake of completeness but refer to Khayyat et al. for a detailed review [110]. The collecting duct (CD) is composed of principal cells (PC) and four types of intercalated cells (IC) [138]. While principal cells are important players in the ion–water balance, including Ca2+ reabsorption, intercalated cells play a critical role in acid–base homeostasis [138,139]. TRPC3 and TRPC6 are expressed in the principal cells of the collecting duct as aquaporin 2-colocalization (AQP-2) indicates [108,140]. Arginine vasopressin or antidiuretic hormone (ADH) is a major regulator of the ion–water balance in principal cells [141]. Binding to the basolateral V2 vasopressin receptor (V2R)—a G-protein-coupled receptor—results in the activation of the cyclic adenosine monophosphate and protein kinase A (cAMP/PKA) pathway enhancing the membrane trafficking of both AQP-2 and TRPC3 [140,142]. The long duration of the effects of arginine vasopressin is partly attributed to the “non-canonical” β-arrestin1/2-dependent V2R-internalization preserving cAMP-PKA signaling. The latter is instead supposed to be terminated by the endosomal retromer complex—a key component of the endosomal protein sorting machinery [143,144]. Sufficient evidence is presented, ascribing the anticalciuretic effects of arginine vasopressin on TRPC3-positive principal cells which translocate TRPC3 and AQP-2 to the apical membrane after V2R activation enabling an apicobasal Ca2+ flux ultimately counteracting calcium crystal formation in times of concentrating antidiuresis [140,145,146]. On the other hand, TRPC3 itself contributes to the translocation of AQP-2 to the apical membrane—as the AQP-2 membrane trafficking likely requires TRPC3-dependent [Ca2+]i raise—according TRPC3 an additional crucial role in water homeostasis [146]. Interestingly, apical CaSR activation occurs during antidiuresis—a state characterized by severe urine concentration—and has been proposed to trigger Ca2+ reabsorption to limit crystal precipitation. In addition, CaSR signaling reduces the vasopressin-induced AQP-2 membrane translocation within a negative feedback loop allowing formation of a not too severely concentrated urine and preventing nephrolithiasis [147]. Moreover, the collecting duct is not only sensitive to endocrine factors such as arginine vasopressin or aldosterone, but also to alterations of the luminal milieu including changes in osmotic gradients or in flow rate. An increase in [Ca2+]i often mediates the adaption of the cellular behavior in matters of water–ion balance, proliferation rates, etc. [148,149,150]. Evidence has been provided suggesting that hypotonicity induces TRPC3-gated Ca2+-entry and initiates the downstream osmosensitive signaling cascade which is reinforced by an additional Ca2+ release from intracellular stores resulting in cellular behavior adaptation [151] (Figure 4). Nevertheless, it is not exactly clear whether the channel itself is sensor of hypotonicity via its long S3 segment, for example [10], or whether it is only a second player in osmosensitive signaling [110]. In contrast, TRPV4 is a key player mediating the cellular response to alterations in tubular flow which is not affected by osmotic alterations such as TRPC3 [110,152,153]. On the basis of this example, we can retrace the diversity of TRP channels and their need in multiple different roles of sensors and effectors in the context of cellular adaptation to environmental changes.
In summary, TRPC3 is a critical player in the downstream signaling pathway that is triggered by arginine vasopressin stimulation in the collecting duct. Stimulation of TRPC3 could be employed to increase trafficking of AQP-2 mutants causing certain forms of nephrogenic diabetes insipidus. In contrast, TRPC3 inhibition might be critical in reversing excessive water retention which could have clinical benefits in certain conditions including congestive heart failure [151].

6. Mitochondrial TRPC3 Drives Detrimental Calcium Uptake and Mediates Cell Proliferation in Autosomal Dominant Polycystic Kidney Disease-like Conditions

ADPKD or autosomal dominant polycystic kidney disease is a genetic disorder that is characterized by multiple bilateral renal cysts resulting in progressive renal failure. It is a very common cause of end-stage kidney disease [154]. In most cases, ADPKD is induced by loss of function mutations affecting the nonselective calcium channels polycystin 1 or polycystin 2 (TRPP2, PKD2, PC2), which are physiologically involved in the regulation of various cellular functions including fluid transport, differentiation, proliferation, cell adhesion, and apoptosis [155,156]. Cellular Ca2+ is altered by a decreased channel function resulting in the activation of the adenylate cyclase with generation of cAMP. The latter stimulates the protein kinase A and Ras/Raf/extracellular-regulated signaling kinase (ERK) pathway that promotes cellular proliferation and cystogenesis [157,158,159]. Current understanding of ADPKD pathogenesis is summarized in [155]. Interestingly, oxidative stress and disordered mitochondrial metabolism were linked to the pathogenesis of ADPKD [159,160,161,162]. Both cellular and mitochondrial ROS and calcium mutually interact. Dysregulation of the one might heavily affect the other [163,164]. TRPC3, a critical player in Ca2+ signaling, is also found in the inner mitochondrial membrane and can directly interact with NADPH oxidase 2 thereby regulating generation of oxidative agents as shown in cardiomyocytes [159,165,166,167]. The involvement of TRPC3 and TRPC7 as components of TRPP2-mutant channel heteropolymers in receptor-operated Ca2+ influx leading to uncontrolled cell proliferation and cystogenesis in ADPKD, has been previously suggested [168]. Transfection of human conditionally immortalized proximal tubular epithelial cells (ciPTEC) and mouse collecting duct cells (IMCD3) with TRPP2-siRNA demonstrated TRPC3-upregulation in ADKPD-like conditions [159]. TRPC3 induced cell proliferation, ERK activation, and mitochondrial dysfunction in interplay with NCX1 upon TRPP2 knockdown. Mitochondrial TRPC3 was also upregulated after TRPP2 knockdown and involved in a mitochondrial Ca2+ influx promoting mitochondrial dysfunctions with impaired ROS generation driving cell proliferation [159]. Interestingly, polycystin-2 was shown to regulate calcium homeostasis players including IP3-receptors, STIM1, TRPV4, and TRPC1 [156]. However, expression levels of TRPC1, TRPC6, and TRPC7 remained the same upon TRPP2 knockdown [159].
In summary, TRPC3 is upregulated in TRPP2-knockdown cells and impairs mitochondrial calcium which is concomitant with mitochondrial dysfunctions thus driving cell proliferation. TRPC3 has already been proposed as medication strategy in various different diseases [169]. Similarly, TRPC3 may become a new focus in the treatment of the most common hereditary kidney disease—ADPKD.

7. Concluding Remarks

This literature review displays the current knowledge and understanding of the physiological and pathophysiological roles of transient receptor potential canonical channels in the renal tubules of the kidney. In this context, recent research mainly focusses on TRPC3 and TRPC6. Tubule-specific physiological functions of TRPC6 remain unclear [38], whereas TRPC3 is awarded a key role in Ca2+ homeostasis in the proximal tubule and collecting duct as illustrated by its Ca2+ reabsorbing function. From a pathophysiological point of view, evidence is provided showing the involvement of TRPC6 in renal cell carcinoma emergence and progression. However, its role in ischemia-reperfusion injuries with acute kidney injury is controversial. In contrast, TRPC3 is a protective player in hypercalciuria, while its upregulation and deleterious role have been demonstrated in autosomal dominant polycystic kidney disease-like conditions. All in all, TRPC channels fulfil diverse functions in the renal tubules of the kidney. Their involvement in severe diseases such as acute or chronic kidney damage, but also in renal cell carcinoma is well known. However, some reports need to be interpreted with caution as there have been only few works completed regarding certain topics. More research is needed to further elucidate and substantiate this underappreciated chapter and eventually achieve a better molecular understanding of severe pathophysiological conditions. Subsequent development of further targeted therapies could lead to a better clinical care.

Author Contributions

Writing—original draft preparation, C.N.E.; writing—review and editing, C.N.E., F.P. and T.T.; visualization, C.N.E. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We gratefully thank Jörg Pekarsky, who excellently supported us in terms of visualization.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Grayson, T.H.; Murphy, T.V.; Sandow, S.L. Transient receptor potential canonical type 3 channels: Interactions, role and relevance—A vascular focus. Pharmacol. Ther. 2017, 174, 79–96. [Google Scholar] [CrossRef] [PubMed]
  2. Nilius, B.; Owsianik, G. The transient receptor potential family of ion channels. Genome Biol. 2011, 12, 218. [Google Scholar] [CrossRef] [Green Version]
  3. Wang, H.; Cheng, X.; Tian, J.; Xiao, Y.; Tian, T.; Xu, F.; Hong, X.; Zhu, M.X. TRPC channels: Structure, function, regulation and recent advances in small molecular probes. Pharmacol. Ther. 2020, 209, 107497. [Google Scholar] [CrossRef]
  4. Tang, Q.; Guo, W.; Zheng, L.; Wu, J.X.; Liu, M.; Zhou, X.; Zhang, X.; Chen, L. Structure of the receptor-activated human TRPC6 and TRPC3 ion channels. Cell Res. 2018, 28, 746–755. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Gross, S.A.; Guzman, G.A.; Wissenbach, U.; Philipp, S.E.; Zhu, M.X.; Bruns, D.; Cavalie, A. TRPC5 is a Ca2+-activated channel functionally coupled to Ca2+-selective ion channels. J. Biol. Chem. 2009, 284, 34423–34432. [Google Scholar] [CrossRef] [Green Version]
  6. Gualdani, R.; Gailly, P. How TRPC Channels Modulate Hippocampal Function. Int. J. Mol. Sci. 2020, 21, 3915. [Google Scholar] [CrossRef] [PubMed]
  7. Canales Coutino, B.; Mayor, R. Mechanosensitive ion channels in cell migration. Cells Dev. 2021, 166, 203683. [Google Scholar] [CrossRef]
  8. Ma, R.; Chaudhari, S.; Li, W. Canonical Transient Receptor Potential 6 Channel: A New Target of Reactive Oxygen Species in Renal Physiology and Pathology. Antioxid Redox Signal. 2016, 25, 732–748. [Google Scholar] [CrossRef] [Green Version]
  9. Sakaguchi, R.; Mori, Y. Transient receptor potential (TRP) channels: Biosensors for redox environmental stimuli and cellular status. Free Radic. Biol. Med. 2020, 146, 36–44. [Google Scholar] [CrossRef]
  10. Fan, C.; Choi, W.; Sun, W.; Du, J.; Lu, W. Structure of the human lipid-gated cation channel TRPC3. Elife 2018, 7, e36852. [Google Scholar] [CrossRef]
  11. Hafner, S.; Urban, N.; Schaefer, M. Discovery and characterization of a positive allosteric modulator of transient receptor potential canonical 6 (TRPC6) channels. Cell Calcium 2019, 78, 26–34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Wie, J.; Jeong, S.; Kwak, M.; Myeong, J.; Chae, M.; Park, J.K.; Lee, S.W.; So, I. The regulation of transient receptor potential canonical 4 (TRPC4) channel by phosphodiesterase 5 inhibitor via the cyclic guanosine 3’5’-monophosphate. Pflugers Arch. 2017, 469, 693–702. [Google Scholar] [CrossRef] [PubMed]
  13. Liu, N.; Zhuang, Y.; Zhou, Z.; Zhao, J.; Chen, Q.; Zheng, J. NF-kappaB dependent up-regulation of TRPC6 by Abeta in BV-2 microglia cells increases COX-2 expression and contributes to hippocampus neuron damage. Neurosci. Lett. 2017, 651, 1–8. [Google Scholar] [CrossRef]
  14. Fu, Y.; Wang, C.; Zhang, D.; Xin, Y.; Li, J.; Zhang, Y.; Chu, X. Increased TRPC6 expression is associated with tubular epithelial cell proliferation and inflammation in diabetic nephropathy. Mol. Immunol. 2018, 94, 75–81. [Google Scholar] [CrossRef] [PubMed]
  15. Spires, D.; Manis, A.D.; Staruschenko, A. Ion channels and transporters in diabetic kidney disease. Curr. Top. Membr. 2019, 83, 353–396. [Google Scholar] [CrossRef] [PubMed]
  16. Staruschenko, A.; Spires, D.; Palygin, O. Role of TRPC6 in Progression of Diabetic Kidney Disease. Curr. Hypertens. Rep. 2019, 21, 48. [Google Scholar] [CrossRef]
  17. Zhou, L.F.; Chen, Q.Z.; Yang, C.T.; Fu, Z.D.; Zhao, S.T.; Chen, Y.; Li, S.N.; Liao, L.; Zhou, Y.B.; Huang, J.R.; et al. TRPC6 contributes to LPS-induced inflammation through ERK1/2 and p38 pathways in bronchial epithelial cells. Am. J. Physiol. Cell Physiol. 2018, 314, C278–C288. [Google Scholar] [CrossRef] [Green Version]
  18. Asghar, M.Y.; Tornquist, K. Transient Receptor Potential Canonical (TRPC) Channels as Modulators of Migration and Invasion. Int. J. Mol. Sci. 2020, 21, 1739. [Google Scholar] [CrossRef] [Green Version]
  19. Vazquez, G.; Wedel, B.J.; Aziz, O.; Trebak, M.; Putney, J.W., Jr. The mammalian TRPC cation channels. Biochim. Biophys. Acta 2004, 1742, 21–36. [Google Scholar] [CrossRef] [Green Version]
  20. Eder, P.; Molkentin, J.D. TRPC channels as effectors of cardiac hypertrophy. Circ. Res. 2011, 108, 265–272. [Google Scholar] [CrossRef]
  21. Shi, J.; Geshi, N.; Takahashi, S.; Kiyonaka, S.; Ichikawa, J.; Hu, Y.; Mori, Y.; Ito, Y.; Inoue, R. Molecular determinants for cardiovascular TRPC6 channel regulation by Ca2+/calmodulin-dependent kinase II. J. Physiol. 2013, 591, 2851–2866. [Google Scholar] [CrossRef]
  22. Lopez, J.J.; Jardin, I.; Sanchez-Collado, J.; Salido, G.M.; Smani, T.; Rosado, J.A. TRPC Channels in the SOCE Scenario. Cells 2020, 9, 126. [Google Scholar] [CrossRef] [Green Version]
  23. Chen, X.; Sooch, G.; Demaree, I.S.; White, F.A.; Obukhov, A.G. Transient Receptor Potential Canonical (TRPC) Channels: Then and Now. Cells 2020, 9, 1983. [Google Scholar] [CrossRef]
  24. Reilly, R.F.; Ellison, D.H. Mammalian distal tubule: Physiology, pathophysiology, and molecular anatomy. Physiol. Rev. 2000, 80, 277–313. [Google Scholar] [CrossRef] [Green Version]
  25. Moor, M.B.; Bonny, O. Ways of calcium reabsorption in the kidney. Am. J. Physiol. Renal Physiol. 2016, 310, F1337–F1350. [Google Scholar] [CrossRef] [Green Version]
  26. Vincent, J.L.; Su, F. Physiology and pathophysiology of the vasopressinergic system. Best Pract. Res. Clin. Anaesthesiol. 2008, 22, 243–252. [Google Scholar] [CrossRef]
  27. Ames, M.K.; Atkins, C.E.; Pitt, B. The renin-angiotensin-aldosterone system and its suppression. J. Vet. Intern. Med. 2019, 33, 363–382. [Google Scholar] [CrossRef] [Green Version]
  28. Gumbert, S.D.; Kork, F.; Jackson, M.L.; Vanga, N.; Ghebremichael, S.J.; Wang, C.Y.; Eltzschig, H.K. Perioperative Acute Kidney Injury. Anesthesiology 2020, 132, 180–204. [Google Scholar] [CrossRef] [Green Version]
  29. Bonventre, J.V.; Yang, L. Cellular pathophysiology of ischemic acute kidney injury. J. Clin. Investig. 2011, 121, 4210–4221. [Google Scholar] [CrossRef]
  30. Keddis, M.T.; Rule, A.D. Nephrolithiasis and loss of kidney function. Curr. Opin. Nephrol. Hypertens. 2013, 22, 390–396. [Google Scholar] [CrossRef]
  31. Awuah Boadi, E.; Shin, S.; Yeroushalmi, S.; Choi, B.E.; Li, P.; Bandyopadhyay, B.C. Modulation of Tubular pH by Acetazolamide in a Ca2+ Transport Deficient Mice Facilitates Calcium Nephrolithiasis. Int. J. Mol. Sci. 2021, 22, 3050. [Google Scholar] [CrossRef]
  32. Shin, S.; Ibeh, C.L.; Awuah Boadi, E.; Choi, B.E.; Roy, S.K.; Bandyopadhyay, B.C. Hypercalciuria switches Ca2+ signaling in proximal tubular cells, induces oxidative damage to promote calcium nephrolithiasis. Genes Dis. 2022, 9, 531–548. [Google Scholar] [CrossRef]
  33. Padala, S.A.; Kallam, A. Clear Cell Renal Carcinoma. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2022. [Google Scholar]
  34. Winn, M.P.; Conlon, P.J.; Lynn, K.L.; Farrington, M.K.; Creazzo, T.; Hawkins, A.F.; Daskalakis, N.; Kwan, S.Y.; Ebersviller, S.; Burchette, J.L.; et al. A mutation in the TRPC6 cation channel causes familial focal segmental glomerulosclerosis. Science 2005, 308, 1801–1804. [Google Scholar] [CrossRef] [Green Version]
  35. Hall, G.; Wang, L.; Spurney, R.F. TRPC Channels in Proteinuric Kidney Diseases. Cells 2019, 9, 44. [Google Scholar] [CrossRef] [Green Version]
  36. Englisch, C.N.; Röhricht, D.; Walz, M.; Junker, K.; Beckmann, A.; Meier, C.; Paulsen, F.; Jung, M.; Tschernig, T. TRPC6 Is Found in Distinct Compartments of the Human Kidney. Int. J. Transl. Med. 2022, 2, 156–163. [Google Scholar] [CrossRef]
  37. Ramkumar, N.; Gao, Y.; Kohan, D.E. Characterization of flow-regulated cortical collecting duct endothelin-1 production. Physiol. Rep. 2017, 5, e13126. [Google Scholar] [CrossRef]
  38. Dryer, S.E.; Roshanravan, H.; Kim, E.Y. TRPC channels: Regulation, dysregulation and contributions to chronic kidney disease. Biochim. Biophys. Acta Mol. Basis Dis. 2019, 1865, 1041–1066. [Google Scholar] [CrossRef]
  39. Staruschenko, A.; Ma, R.; Palygin, O.; Dryer, S.E. Ion channels and channelopathies in glomeruli. Physiol. Rev. 2023, 103, 787–854. [Google Scholar] [CrossRef]
  40. Guan, X.; Qian, Y.; Shen, Y.; Zhang, L.; Du, Y.; Dai, H.; Qian, J.; Yan, Y. Autophagy protects renal tubular cells against ischemia / reperfusion injury in a time-dependent manner. Cell. Physiol. Biochem. 2015, 36, 285–298. [Google Scholar] [CrossRef] [Green Version]
  41. Khwaja, A. KDIGO clinical practice guidelines for acute kidney injury. Nephron Clin. Pract. 2012, 120, c179–c184. [Google Scholar] [CrossRef]
  42. Zheng, Z.; Tsvetkov, D.; Bartolomaeus, T.U.P.; Erdogan, C.; Krugel, U.; Schleifenbaum, J.; Schaefer, M.; Nurnberg, B.; Chai, X.; Ludwig, F.A.; et al. Role of TRPC6 in kidney damage after acute ischemic kidney injury. Sci. Rep. 2022, 12, 3038. [Google Scholar] [CrossRef] [PubMed]
  43. Liu, B.C.; Tang, T.T.; Lv, L.L.; Lan, H.Y. Renal tubule injury: A driving force toward chronic kidney disease. Kidney Int. 2018, 93, 568–579. [Google Scholar] [CrossRef] [PubMed]
  44. Livingston, M.J.; Dong, Z. Autophagy in acute kidney injury. Semin. Nephrol. 2014, 34, 17–26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Sharfuddin, A.A.; Molitoris, B.A. Pathophysiology of ischemic acute kidney injury. Nat. Rev. Nephrol. 2011, 7, 189–200. [Google Scholar] [CrossRef] [PubMed]
  46. Skrypnyk, N.I.; Harris, R.C.; de Caestecker, M.P. Ischemia-reperfusion model of acute kidney injury and post injury fibrosis in mice. J. Vis. Exp. 2013, 9, e50495. [Google Scholar] [CrossRef]
  47. Ratliff, B.B.; Abdulmahdi, W.; Pawar, R.; Wolin, M.S. Oxidant Mechanisms in Renal Injury and Disease. Antioxid Redox Signal. 2016, 25, 119–146. [Google Scholar] [CrossRef] [Green Version]
  48. Bhargava, P.; Schnellmann, R.G. Mitochondrial energetics in the kidney. Nat. Rev. Nephrol. 2017, 13, 629–646. [Google Scholar] [CrossRef]
  49. Jurcau, A.; Ardelean, I.A. Molecular pathophysiological mechanisms of ischemia/reperfusion injuries after recanalization therapy for acute ischemic stroke. J. Integr. Neurosci. 2021, 20, 727–744. [Google Scholar] [CrossRef]
  50. Li, L.; Tan, J.; Miao, Y.; Lei, P.; Zhang, Q. ROS and Autophagy: Interactions and Molecular Regulatory Mechanisms. Cell. Mol. Neurobiol. 2015, 35, 615–621. [Google Scholar] [CrossRef]
  51. Yan, H.F.; Tuo, Q.Z.; Yin, Q.Z.; Lei, P. The pathological role of ferroptosis in ischemia/reperfusion-related injury. Zool. Res. 2020, 41, 220–230. [Google Scholar] [CrossRef]
  52. Gores, G.J.; Nieminen, A.L.; Fleishman, K.E.; Dawson, T.L.; Herman, B.; Lemasters, J.J. Extracellular acidosis delays onset of cell death in ATP-depleted hepatocytes. Am. J. Physiol. 1988, 255, C315–C322. [Google Scholar] [CrossRef] [PubMed]
  53. Raat, N.J.; Shiva, S.; Gladwin, M.T. Effects of nitrite on modulating ROS generation following ischemia and reperfusion. Adv. Drug Deliv. Rev. 2009, 61, 339–350. [Google Scholar] [CrossRef] [PubMed]
  54. Kimura, S.; Bassett, A.L.; Gaide, M.S.; Kozlovskis, P.L.; Myerburg, R.J. Regional changes in intracellular potassium and sodium activity after healing of experimental myocardial infarction in cats. Circ. Res. 1986, 58, 202–208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Halestrap, A.P. Calcium, mitochondria and reperfusion injury: A pore way to die. Biochem. Soc. Trans. 2006, 34, 232–237. [Google Scholar] [CrossRef] [PubMed]
  56. Nayler, W.G. The role of calcium in the ischemic myocardium. Am. J. Pathol. 1981, 102, 262–270. [Google Scholar] [PubMed]
  57. Weisfeldt, M.L.; Zweier, J.; Ambrosio, G.; Becker, L.C.; Flaherty, J.T. Evidence that free radicals result in reperfusion injury in heart muscle. Basic Life Sci. 1988, 49, 911–919. [Google Scholar] [CrossRef]
  58. Zweier, J.L.; Flaherty, J.T.; Weisfeldt, M.L. Direct measurement of free radical generation following reperfusion of ischemic myocardium. Proc. Natl. Acad. Sci. USA 1987, 84, 1404–1407. [Google Scholar] [CrossRef] [Green Version]
  59. Becker, L.C.; Ambrosio, G. Myocardial consequences of reperfusion. Prog. Cardiovasc. Dis. 1987, 30, 23–44. [Google Scholar] [CrossRef]
  60. Hess, M.L.; Manson, N.H. Molecular oxygen: Friend and foe. The role of the oxygen free radical system in the calcium paradox, the oxygen paradox and ischemia/reperfusion injury. J. Mol. Cell. Cardiol. 1984, 16, 969–985. [Google Scholar] [CrossRef]
  61. Dare, A.J.; Bolton, E.A.; Pettigrew, G.J.; Bradley, J.A.; Saeb-Parsy, K.; Murphy, M.P. Protection against renal ischemia-reperfusion injury in vivo by the mitochondria targeted antioxidant MitoQ. Redox Biol. 2015, 5, 163–168. [Google Scholar] [CrossRef]
  62. Tajima, T.; Yoshifuji, A.; Matsui, A.; Itoh, T.; Uchiyama, K.; Kanda, T.; Tokuyama, H.; Wakino, S.; Itoh, H. beta-hydroxybutyrate attenuates renal ischemia-reperfusion injury through its anti-pyroptotic effects. Kidney Int. 2019, 95, 1120–1137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Wu, M.Y.; Yiang, G.T.; Liao, W.T.; Tsai, A.P.; Cheng, Y.L.; Cheng, P.W.; Li, C.Y.; Li, C.J. Current Mechanistic Concepts in Ischemia and Reperfusion Injury. Cell. Physiol. Biochem. 2018, 46, 1650–1667. [Google Scholar] [CrossRef] [PubMed]
  64. Shen, B.; Zhou, S.; He, Y.; Zhao, H.; Mei, M.; Wu, X. Revealing the underlying mechanism of ischemia reperfusion injury using bioinformatics approach. Kidney Blood Press. Res. 2013, 38, 99–108. [Google Scholar] [CrossRef] [PubMed]
  65. Hou, X.; Huang, M.; Zeng, X.; Zhang, Y.; Sun, A.; Wu, Q.; Zhu, L.; Zhao, H.; Liao, Y. The Role of TRPC6 in Renal Ischemia/Reperfusion and Cellular Hypoxia/Reoxygenation Injuries. Front. Mol. Biosci 2021, 8, 698975. [Google Scholar] [CrossRef] [PubMed]
  66. Shen, B.; Mei, M.; Ai, S.; Liao, X.; Li, N.; Xiang, S.; Wen, C.; Tao, Y.; Dai, H. TRPC6 inhibits renal tubular epithelial cell pyroptosis through regulating zinc influx and alleviates renal ischemia-reperfusion injury. FASEB J. 2022, 36, e22527. [Google Scholar] [CrossRef]
  67. Shen, B.; Mei, M.; Pu, Y.; Zhang, H.; Liu, H.; Tang, M.; Pan, Q.; He, Y.; Wu, X.; Zhao, H. Necrostatin-1 Attenuates Renal Ischemia and Reperfusion Injury via Meditation of HIF-1alpha/mir-26a/TRPC6/PARP1 Signaling. Mol. Ther. Nucleic Acids 2019, 17, 701–713. [Google Scholar] [CrossRef] [Green Version]
  68. Ding, Y.; Winters, A.; Ding, M.; Graham, S.; Akopova, I.; Muallem, S.; Wang, Y.; Hong, J.H.; Gryczynski, Z.; Yang, S.H.; et al. Reactive oxygen species-mediated TRPC6 protein activation in vascular myocytes, a mechanism for vasoconstrictor-regulated vascular tone. J. Biol. Chem. 2011, 286, 31799–31809. [Google Scholar] [CrossRef] [Green Version]
  69. Hou, X.; Xiao, H.; Zhang, Y.; Zeng, X.; Huang, M.; Chen, X.; Birnbaumer, L.; Liao, Y. Transient receptor potential channel 6 knockdown prevents apoptosis of renal tubular epithelial cells upon oxidative stress via autophagy activation. Cell Death Dis. 2018, 9, 1015. [Google Scholar] [CrossRef] [Green Version]
  70. Liu, S.; Hartleben, B.; Kretz, O.; Wiech, T.; Igarashi, P.; Mizushima, N.; Walz, G.; Huber, T.B. Autophagy plays a critical role in kidney tubule maintenance, aging and ischemia-reperfusion injury. Autophagy 2012, 8, 826–837. [Google Scholar] [CrossRef] [Green Version]
  71. Kimura, T.; Takabatake, Y.; Takahashi, A.; Kaimori, J.Y.; Matsui, I.; Namba, T.; Kitamura, H.; Niimura, F.; Matsusaka, T.; Soga, T.; et al. Autophagy protects the proximal tubule from degeneration and acute ischemic injury. J. Am. Soc. Nephrol. 2011, 22, 902–913. [Google Scholar] [CrossRef] [Green Version]
  72. Havasi, A.; Dong, Z. Autophagy and Tubular Cell Death in the Kidney. Semin. Nephrol. 2016, 36, 174–188. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Lin, F. Autophagy in renal tubular injury and repair. Acta Physiol. 2017, 220, 229–237. [Google Scholar] [CrossRef] [PubMed]
  74. Pu, Y.; Zhao, H.; Shen, B.; Zhou, Q.; Xie, P.; Wu, X. TRPC6 ameliorates renal ischemic reperfusion injury by inducing Zn(2+) influx and activating autophagy to resist necrosis. Ann. Transl. Med. 2022, 10, 249. [Google Scholar] [CrossRef]
  75. Shen, B.; He, Y.; Zhou, S.; Zhao, H.; Mei, M.; Wu, X. TRPC6 May Protect Renal Ischemia-Reperfusion Injury Through Inhibiting Necroptosis of Renal Tubular Epithelial Cells. Med. Sci. Monit. 2016, 22, 633–641. [Google Scholar] [CrossRef]
  76. Shen, S.; Jin, Y.; Li, W.; Liu, X.; Zhang, T.; Xia, W.; Wang, Y.; Ma, K. Recombinant human erythropoietin pretreatment attenuates acute renal tubular injury against ischemia-reperfusion by restoring transient receptor potential channel-6 expression and function in collecting ducts. Crit. Care Med. 2014, 42, e663–e672. [Google Scholar] [CrossRef] [PubMed]
  77. Shin, S.; Gombedza, F.C.; Bandyopadhyay, B.C. l-ornithine activates Ca2+ signaling to exert its protective function on human proximal tubular cells. Cell. Signal. 2020, 67, 109484. [Google Scholar] [CrossRef]
  78. Livingstone, C. Zinc: Physiology, deficiency, and parenteral nutrition. Nutr. Clin. Pract. 2015, 30, 371–382. [Google Scholar] [CrossRef]
  79. Chevallet, M.; Jarvis, L.; Harel, A.; Luche, S.; Degot, S.; Chapuis, V.; Boulay, G.; Rabilloud, T.; Bouron, A. Functional consequences of the over-expression of TRPC6 channels in HEK cells: Impact on the homeostasis of zinc. Metallomics 2014, 6, 1269–1276. [Google Scholar] [CrossRef]
  80. Liu, J.; Kang, Y.; Yin, S.; Chen, A.; Wu, J.; Liang, H.; Shao, L. Key Role of Microtubule and Its Acetylation in a Zinc Oxide Nanoparticle-Mediated Lysosome-Autophagy System. Small 2019, 15, e1901073. [Google Scholar] [CrossRef]
  81. Gibon, J.; Tu, P.; Bohic, S.; Richaud, P.; Arnaud, J.; Zhu, M.; Boulay, G.; Bouron, A. The over-expression of TRPC6 channels in HEK-293 cells favours the intracellular accumulation of zinc. Biochim. Biophys. Acta 2011, 1808, 2807–2818. [Google Scholar] [CrossRef]
  82. Shi, J.; Gao, W.; Shao, F. Pyroptosis: Gasdermin-Mediated Programmed Necrotic Cell Death. Trends Biochem. Sci. 2017, 42, 245–254. [Google Scholar] [CrossRef] [PubMed]
  83. Li, C.; Chen, M.; He, X.; Ouyang, D. A mini-review on ion fluxes that regulate NLRP3 inflammasome activation. Acta Biochim. Biophys. Sin. 2021, 53, 131–139. [Google Scholar] [CrossRef] [PubMed]
  84. Chen, X.; Liu, G.; Yuan, Y.; Wu, G.; Wang, S.; Yuan, L. NEK7 interacts with NLRP3 to modulate the pyroptosis in inflammatory bowel disease via NF-kappaB signaling. Cell Death Dis. 2019, 10, 906. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Ning, B.; Guo, C.; Kong, A.; Li, K.; Xie, Y.; Shi, H.; Gu, J. Calcium Signaling Mediates Cell Death and Crosstalk with Autophagy in Kidney Disease. Cells 2021, 10, 3204. [Google Scholar] [CrossRef] [PubMed]
  86. Monteith, G.R.; Prevarskaya, N.; Roberts-Thomson, S.J. The calcium-cancer signalling nexus. Nat. Rev. Cancer 2017, 17, 367–380. [Google Scholar] [CrossRef] [Green Version]
  87. Monteith, G.R.; Davis, F.M.; Roberts-Thomson, S.J. Calcium channels and pumps in cancer: Changes and consequences. J. Biol. Chem. 2012, 287, 31666–31673. [Google Scholar] [CrossRef] [Green Version]
  88. Klatte, T.; Rossi, S.H.; Stewart, G.D. Prognostic factors and prognostic models for renal cell carcinoma: A literature review. World J. Urol. 2018, 36, 1943–1952. [Google Scholar] [CrossRef]
  89. Ferlay, J.; Soerjomataram, I.; Dikshit, R.; Eser, S.; Mathers, C.; Rebelo, M.; Parkin, D.M.; Forman, D.; Bray, F. Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int. J. Cancer 2015, 136, E359–E386. [Google Scholar] [CrossRef]
  90. Makhov, P.; Joshi, S.; Ghatalia, P.; Kutikov, A.; Uzzo, R.G.; Kolenko, V.M. Resistance to Systemic Therapies in Clear Cell Renal Cell Carcinoma: Mechanisms and Management Strategies. Mol. Cancer Ther. 2018, 17, 1355–1364. [Google Scholar] [CrossRef] [Green Version]
  91. Paner, G.P.; Chumbalkar, V.; Montironi, R.; Moch, H.; Amin, M.B. Updates in Grading of Renal Cell Carcinomas Beyond Clear Cell Renal Cell Carcinoma and Papillary Renal Cell Carcinoma. Adv. Anat. Pathol. 2022, 29, 117–130. [Google Scholar] [CrossRef]
  92. Nandagopal, L.; Sonpavde, G.P.; Agarwal, N. Investigational MET inhibitors to treat Renal cell carcinoma. Expert Opin. Investig. Drugs 2019, 28, 851–860. [Google Scholar] [CrossRef] [PubMed]
  93. Kim, J.H.; Hwang, K.H.; Eom, M.; Kim, M.; Park, E.Y.; Jeong, Y.; Park, K.S.; Cha, S.K. WNK1 promotes renal tumor progression by activating TRPC6-NFAT pathway. FASEB J. 2019, 33, 8588–8599. [Google Scholar] [CrossRef] [PubMed]
  94. Song, J.; Wang, Y.; Li, X.; Shen, Y.; Yin, M.; Guo, Y.; Diao, L.; Liu, Y.; Yue, D. Critical role of TRPC6 channels in the development of human renal cell carcinoma. Mol. Biol. Rep. 2013, 40, 5115–5122. [Google Scholar] [CrossRef] [PubMed]
  95. Kochevar, J. Blockage of autonomous growth of ACHN cells by anti-renal cell carcinoma monoclonal antibody 5F4. Cancer Res. 1990, 50, 2968–2972. [Google Scholar]
  96. Zhan, C.; Shi, Y. TRPC Channels and Cell Proliferation. Adv. Exp. Med. Biol. 2017, 976, 149–155. [Google Scholar] [CrossRef]
  97. Horie, S.; Aruga, S.; Kawamata, H.; Okui, N.; Kakizoe, T.; Kitamura, T. Biological role of HGF/MET pathway in renal cell carcinoma. J. Urol. 1999, 161, 990–997. [Google Scholar] [CrossRef]
  98. Yamauchi, M.; Kataoka, H.; Itoh, H.; Seguchi, T.; Hasui, Y.; Osada, Y. Hepatocyte growth factor activator inhibitor types 1 and 2 are expressed by tubular epithelium in kidney and down-regulated in renal cell carcinoma. J. Urol. 2004, 171, 890–896. [Google Scholar] [CrossRef]
  99. Finley, D.S.; Pantuck, A.J.; Belldegrun, A.S. Tumor biology and prognostic factors in renal cell carcinoma. Oncologist 2011, 16 (Suppl. 2), 4–13. [Google Scholar] [CrossRef] [Green Version]
  100. Bahena-Lopez, J.P.; Gamba, G.; Castaneda-Bueno, M. WNK1 in the kidney. Curr. Opin. Nephrol. Hypertens. 2022, 31, 471–478. [Google Scholar] [CrossRef]
  101. McCormick, J.A.; Ellison, D.H. The WNKs: Atypical protein kinases with pleiotropic actions. Physiol. Rev. 2011, 91, 177–219. [Google Scholar] [CrossRef] [Green Version]
  102. Shekarabi, M.; Zhang, J.; Khanna, A.R.; Ellison, D.H.; Delpire, E.; Kahle, K.T. WNK Kinase Signaling in Ion Homeostasis and Human Disease. Cell Metab. 2017, 25, 285–299. [Google Scholar] [CrossRef] [PubMed]
  103. Moniz, S.; Jordan, P. Emerging roles for WNK kinases in cancer. Cell Mol. Life Sci. 2010, 67, 1265–1276. [Google Scholar] [CrossRef] [PubMed]
  104. Shroff, E.H.; Eberlin, L.S.; Dang, V.M.; Gouw, A.M.; Gabay, M.; Adam, S.J.; Bellovin, D.I.; Tran, P.T.; Philbrick, W.M.; Garcia-Ocana, A.; et al. MYC oncogene overexpression drives renal cell carcinoma in a mouse model through glutamine metabolism. Proc. Natl. Acad. Sci. USA 2015, 112, 6539–6544. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Dragoni, S.; Turin, I.; Laforenza, U.; Potenza, D.M.; Bottino, C.; Glasnov, T.N.; Prestia, M.; Ferulli, F.; Saitta, A.; Mosca, A.; et al. Store-operated Ca2+ entry does not control. proliferation in primary cultures of human metastatic renal cellular carcinoma. Biomed. Res. Int. 2014, 2014, 739494. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Chen, L.; Shan, G.; Ge, M.; Qian, H.; Xia, Y. Transient Receptor Potential Channel 1 Potentially Serves as a Biomarker Indicating T/TNM Stages and Predicting Long-Term Prognosis in Patients With Renal Cell Carcinoma. Front. Surg. 2022, 9, 853310. [Google Scholar] [CrossRef] [PubMed]
  107. Chauhan, A.; Semwal, D.K.; Mishra, S.P.; Goyal, S.; Marathe, R.; Semwal, R.B. Combination of mTOR and MAPK Inhibitors-A Potential Way to Treat Renal Cell Carcinoma. Med. Sci. 2016, 4, 16. [Google Scholar] [CrossRef] [Green Version]
  108. Goel, M.; Sinkins, W.G.; Zuo, C.D.; Estacion, M.; Schilling, W.P. Identification and localization of TRPC channels in the rat kidney. Am. J. Physiol. Renal Physiol. 2006, 290, F1241–F1252. [Google Scholar] [CrossRef] [Green Version]
  109. Ibeh, C.L.; Yiu, A.J.; Kanaras, Y.L.; Paal, E.; Birnbaumer, L.; Jose, P.A.; Bandyopadhyay, B.C. Evidence for a regulated Ca2+ entry in proximal tubular cells and its implication in calcium stone formation. J. Cell Sci. 2019, 132, jcs225268. [Google Scholar] [CrossRef] [Green Version]
  110. Khayyat, N.H.; Tomilin, V.N.; Zaika, O.; Pochynyuk, O. Polymodal roles of TRPC3 channel in the kidney. Channels 2020, 14, 257–267. [Google Scholar] [CrossRef]
  111. Friedman, P.A. Mechanisms of renal calcium transport. Exp. Nephrol. 2000, 8, 343–350. [Google Scholar] [CrossRef]
  112. Bandyopadhyay, B.C.; Swaim, W.D.; Liu, X.; Redman, R.S.; Patterson, R.L.; Ambudkar, I.S. Apical localization of a functional TRPC3/TRPC6-Ca2+-signaling complex in polarized epithelial cells. Role in apical Ca2+ influx. J. Biol. Chem. 2005, 280, 12908–12916. [Google Scholar] [CrossRef] [PubMed]
  113. Brown, E.M.; MacLeod, R.J. Extracellular calcium sensing and extracellular calcium signaling. Physiol. Rev. 2001, 81, 239–297. [Google Scholar] [CrossRef] [PubMed]
  114. Chang, W.; Chen, T.H.; Pratt, S.; Shoback, D. Amino acids in the second and third intracellular loops of the parathyroid Ca2+-sensing receptor mediate efficient coupling to phospholipase C. J. Biol. Chem. 2000, 275, 19955–19963. [Google Scholar] [CrossRef] [Green Version]
  115. Gombedza, F.C.; Shin, S.; Kanaras, Y.L.; Bandyopadhyay, B.C. Abrogation of store-operated Ca2+ entry protects against crystal-induced ER stress in human proximal tubular cells. Cell Death Discov. 2019, 5, 124. [Google Scholar] [CrossRef] [Green Version]
  116. Yiu, A.J.; Ibeh, C.L.; Roy, S.K.; Bandyopadhyay, B.C. Melamine induces Ca2+-sensing receptor activation and elicits apoptosis in proximal tubular cells. Am. J. Physiol. Cell Physiol. 2017, 313, C27–C41. [Google Scholar] [CrossRef] [Green Version]
  117. Asplin, J.R.; Mandel, N.S.; Coe, F.L. Evidence of calcium phosphate supersaturation in the loop of Henle. Am. J. Physiol. 1996, 270, F604–F613. [Google Scholar] [CrossRef]
  118. Tiselius, H.G. A hypothesis of calcium stone formation: An interpretation of stone research during the past decades. Urol. Res. 2011, 39, 231–243. [Google Scholar] [CrossRef]
  119. Coe, F.L.; Worcester, E.M.; Evan, A.P. Idiopathic hypercalciuria and formation of calcium renal stones. Nat. Rev. Nephrol. 2016, 12, 519–533. [Google Scholar] [CrossRef] [Green Version]
  120. Bird, V.Y.; Khan, S.R. How do stones form? Is unification of theories on stone formation possible? Arch. Esp. Urol. 2017, 70, 12–27. [Google Scholar]
  121. Evan, A.P.; Lingeman, J.E.; Coe, F.L.; Parks, J.H.; Bledsoe, S.B.; Shao, Y.; Sommer, A.J.; Paterson, R.F.; Kuo, R.L.; Grynpas, M. Randall’s plaque of patients with nephrolithiasis begins in basement membranes of thin loops of Henle. J. Clin. Investig. 2003, 111, 607–616. [Google Scholar] [CrossRef] [Green Version]
  122. Doroszewicz, J.; Waldegger, P.; Jeck, N.; Seyberth, H.; Waldegger, S. pH dependence of extracellular calcium sensing receptor activity determined by a novel technique. Kidney Int. 2005, 67, 187–192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Matlaga, B.R.; Shah, O.D.; Assimos, D.G. Drug-induced urinary calculi. Rev. Urol. 2003, 5, 227–231. [Google Scholar] [PubMed]
  124. Luo, D.D.; Fielding, C.; Phillips, A.; Fraser, D. Interleukin-1 beta regulates proximal tubular cell transforming growth factor beta-1 signalling. Nephrol. Dial Transplant. 2009, 24, 2655–2665. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Mulay, S.R.; Evan, A.; Anders, H.J. Molecular mechanisms of crystal-related kidney inflammation and injury. Implications for cholesterol. embolism, crystalline nephropathies and kidney stone disease. Nephrol. Dial Transplant. 2014, 29, 507–514. [Google Scholar] [CrossRef] [Green Version]
  126. Vesey, D.A.; Cheung, C.W.; Cuttle, L.; Endre, Z.A.; Gobe, G.; Johnson, D.W. Interleukin-1beta induces human proximal tubule cell injury, alpha-smooth muscle actin expression and fibronectin production. Kidney Int. 2002, 62, 31–40. [Google Scholar] [CrossRef]
  127. Gewin, L.S. Renal fibrosis: Primacy of the proximal tubule. Matrix Biol. 2018, 68–69, 248–262. [Google Scholar] [CrossRef]
  128. Berg, C.; Tiselius, H.G. The effect of pH on the risk of calcium oxalate crystallization in urine. Eur. Urol. 1986, 12, 59–61. [Google Scholar] [CrossRef]
  129. Carra, G.; Avalle, L.; Secli, L.; Brancaccio, M.; Morotti, A. Shedding Light on NF-kappaB Functions in Cellular Organelles. Front. Cell Dev. Biol. 2022, 10, 841646. [Google Scholar] [CrossRef]
  130. Zhu, X.; Huang, L.; Gong, J.; Shi, C.; Wang, Z.; Ye, B.; Xuan, A.; He, X.; Long, D.; Zhu, X.; et al. NF-kappaB pathway link with ER stress-induced autophagy and apoptosis in cervical tumor cells. Cell Death Discov. 2017, 3, 17059. [Google Scholar] [CrossRef] [Green Version]
  131. Kannan, K.; Jain, S.K. Oxidative stress and apoptosis. Pathophysiology 2000, 7, 153–163. [Google Scholar] [CrossRef]
  132. Hitomi, J.; Katayama, T.; Taniguchi, M.; Honda, A.; Imaizumi, K.; Tohyama, M. Apoptosis induced by endoplasmic reticulum stress depends on activation of caspase-3 via caspase-12. Neurosci. Lett. 2004, 357, 127–130. [Google Scholar] [CrossRef] [Green Version]
  133. Rustom, R.; Wang, B.; McArdle, F.; Shalamanova, L.; Alexander, J.; McArdle, A.; Thomas, C.E.; Bone, J.M.; Shenkin, A.; Jackson, M.J. Oxidative stress in a novel model of chronic acidosis in LLC-PK1 cells. Nephron Exp. Nephrol. 2003, 95, e13–e23. [Google Scholar] [CrossRef]
  134. Zeeshan, H.M.; Lee, G.H.; Kim, H.R.; Chae, H.J. Endoplasmic Reticulum Stress and Associated ROS. Int. J. Mol. Sci. 2016, 17, 327. [Google Scholar] [CrossRef] [Green Version]
  135. Scholze, A.; Jankowski, J.; Pedraza-Chaverri, J.; Evenepoel, P. Oxidative Stress in Chronic Kidney Disease. Oxid Med. Cell. Longev. 2016, 2016, 8375186. [Google Scholar] [CrossRef] [Green Version]
  136. Khan, S.R. Reactive oxygen species, inflammation and calcium oxalate nephrolithiasis. Transl. Androl. Urol. 2014, 3, 256–276. [Google Scholar] [CrossRef]
  137. Gombedza, F.; Evans, S.; Shin, S.; Awuah Boadi, E.; Zhang, Q.; Nie, Z.; Bandyopadhyay, B.C. Melamine promotes calcium crystal formation in three-dimensional microfluidic device. Sci. Rep. 2019, 9, 875. [Google Scholar] [CrossRef] [Green Version]
  138. Lashhab, R.; Ullah, A.; Cordat, E. Renal collecting duct physiology and pathophysiology. Biochem. Cell Biol. 2019, 97, 234–242. [Google Scholar] [CrossRef]
  139. Magaldi, A.J.; van Baak, A.A.; Rocha, A.S. Calcium transport across rat inner medullary collecting duct perfused in vitro. Am. J. Physiol. 1989, 257, F738–F745. [Google Scholar] [CrossRef]
  140. Goel, M.; Sinkins, W.G.; Zuo, C.D.; Hopfer, U.; Schilling, W.P. Vasopressin-induced membrane trafficking of TRPC3 and AQP2 channels in cells of the rat renal collecting duct. Am. J. Physiol. Renal Physiol. 2007, 293, F1476–F1488. [Google Scholar] [CrossRef] [Green Version]
  141. Knepper, M.A.; Kwon, T.H.; Nielsen, S. Molecular physiology of water balance. N. Engl. J. Med. 2015, 372, 1349–1358. [Google Scholar] [CrossRef]
  142. Goel, M.; Zuo, C.D.; Schilling, W.P. Role of cAMP/PKA signaling cascade in vasopressin-induced trafficking of TRPC3 channels in principal cells of the collecting duct. Am. J. Physiol. Renal Physiol. 2010, 298, F988–F996. [Google Scholar] [CrossRef] [Green Version]
  143. Feinstein, T.N.; Yui, N.; Webber, M.J.; Wehbi, V.L.; Stevenson, H.P.; King, J.D., Jr.; Hallows, K.R.; Brown, D.; Bouley, R.; Vilardaga, J.P. Noncanonical control. of vasopressin receptor type 2 signaling by retromer and arrestin. J. Biol. Chem. 2013, 288, 27849–27860. [Google Scholar] [CrossRef]
  144. Seaman, M.N. The retromer complex—Endosomal protein recycling and beyond. J. Cell Sci. 2012, 125, 4693–4702. [Google Scholar] [CrossRef] [Green Version]
  145. Hanouna, G.; Haymann, J.P.; Baud, L.; Letavernier, E. Vasopressin regulates renal calcium excretion in humans. Physiol. Rep. 2015, 3, e12562. [Google Scholar] [CrossRef]
  146. Goel, M.; Schilling, W.P. Role of TRPC3 channels in ATP-induced Ca2+ signaling in principal cells of the inner medullary collecting duct. Am. J. Physiol. Renal Physiol. 2010, 299, F225–F233. [Google Scholar] [CrossRef] [Green Version]
  147. Ranieri, M. Renal Ca2+ and Water Handling in Response to Calcium Sensing Receptor Signaling: Physiopathological Aspects and Role of CaSR-Regulated microRNAs. Int. J. Mol. Sci. 2019, 20, 5341. [Google Scholar] [CrossRef] [Green Version]
  148. 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]
  149. Weinbaum, S.; Duan, Y.; Satlin, L.M.; Wang, T.; Weinstein, A.M. Mechanotransduction in the renal tubule. Am. J. Physiol. Renal Physiol. 2010, 299, F1220–F1236. [Google Scholar] [CrossRef] [Green Version]
  150. McCarty, N.A.; O’Neil, R.G. Calcium signaling in cell volume regulation. Physiol. Rev. 1992, 72, 1037–1061. [Google Scholar] [CrossRef]
  151. Tomilin, V.N.; Mamenko, M.; Zaika, O.; Ren, G.; Marrelli, S.P.; Birnbaumer, L.; Pochynyuk, O. TRPC3 determines osmosensitive [Ca2+]i signaling in the collecting duct and contributes to urinary concentration. PLoS ONE 2019, 14, e0226381. [Google Scholar] [CrossRef] [Green Version]
  152. Berrout, J.; Jin, M.; Mamenko, M.; Zaika, O.; Pochynyuk, O.; O’Neil, R.G. Function of transient receptor potential cation channel subfamily V member 4 (TRPV4) as a mechanical transducer in flow-sensitive segments of renal collecting duct system. J. Biol. Chem. 2012, 287, 8782–8791. [Google Scholar] [CrossRef] [Green Version]
  153. Mamenko, M.V.; Boukelmoune, N.; Tomilin, V.N.; Zaika, O.L.; Jensen, V.B.; O’Neil, R.G.; Pochynyuk, O.M. The renal TRPV4 channel is essential for adaptation to increased dietary potassium. Kidney Int. 2017, 91, 1398–1409. [Google Scholar] [CrossRef]
  154. Cornec-Le Gall, E.; Alam, A.; Perrone, R.D. Autosomal dominant polycystic kidney disease. Lancet 2019, 393, 919–935. [Google Scholar] [CrossRef]
  155. Reiterova, J.; Tesar, V. Autosomal Dominant Polycystic Kidney Disease: From Pathophysiology of Cystogenesis to Advances in the Treatment. Int. J. Mol. Sci. 2022, 23, 3317. [Google Scholar] [CrossRef]
  156. Chaudhari, S.; Mallet, R.T.; Shotorbani, P.Y.; Tao, Y.; Ma, R. Store-operated calcium entry: Pivotal roles in renal physiology and pathophysiology. Exp. Biol. Med. 2021, 246, 305–316. [Google Scholar] [CrossRef]
  157. Piazzon, N.; Maisonneuve, C.; Guilleret, I.; Rotman, S.; Constam, D.B. Bicc1 links the regulation of cAMP signaling in polycystic kidneys to microRNA-induced gene silencing. J. Mol. Cell Biol. 2012, 4, 398–408. [Google Scholar] [CrossRef] [Green Version]
  158. Kuo, I.Y.; DesRochers, T.M.; Kimmerling, E.P.; Nguyen, L.; Ehrlich, B.E.; Kaplan, D.L. Cyst formation following disruption of intracellular calcium signaling. Proc. Natl. Acad. Sci. USA 2014, 111, 14283–14288. [Google Scholar] [CrossRef] [Green Version]
  159. Li, Z.; Zhou, J.; Li, Y.; Yang, F.; Lian, X.; Liu, W. Mitochondrial TRPC3 promotes cell proliferation by regulating the mitochondrial calcium and metabolism in renal polycystin-2 knockdown cells. Int. Urol. Nephrol. 2019, 51, 1059–1070. [Google Scholar] [CrossRef]
  160. Klawitter, J.; Reed-Gitomer, B.Y.; McFann, K.; Pennington, A.; Klawitter, J.; Abebe, K.Z.; Klepacki, J.; Cadnapaphornchai, M.A.; Brosnahan, G.; Chonchol, M.; et al. Endothelial dysfunction and oxidative stress in polycystic kidney disease. Am. J. Physiol. Renal Physiol. 2014, 307, F1198–F1206. [Google Scholar] [CrossRef] [Green Version]
  161. Menon, V.; Rudym, D.; Chandra, P.; Miskulin, D.; Perrone, R.; Sarnak, M. Inflammation, oxidative stress, and insulin resistance in polycystic kidney disease. Clin. J. Am. Soc. Nephrol. 2011, 6, 7–13. [Google Scholar] [CrossRef] [Green Version]
  162. Ishimoto, Y.; Inagi, R.; Yoshihara, D.; Kugita, M.; Nagao, S.; Shimizu, A.; Takeda, N.; Wake, M.; Honda, K.; Zhou, J.; et al. Mitochondrial Abnormality Facilitates Cyst Formation in Autosomal Dominant Polycystic Kidney Disease. Mol. Cell Biol. 2017, 37, 00337-17. [Google Scholar] [CrossRef] [Green Version]
  163. Gorlach, A.; Bertram, K.; Hudecova, S.; Krizanova, O. Calcium and ROS: A mutual interplay. Redox Biol. 2015, 6, 260–271. [Google Scholar] [CrossRef]
  164. Bertero, E.; Maack, C. Calcium Signaling and Reactive Oxygen Species in Mitochondria. Circ. Res. 2018, 122, 1460–1478. [Google Scholar] [CrossRef]
  165. Feng, S.; Li, H.; Tai, Y.; Huang, J.; Su, Y.; Abramowitz, J.; Zhu, M.X.; Birnbaumer, L.; Wang, Y. Canonical transient receptor potential 3 channels regulate mitochondrial calcium uptake. Proc. Natl. Acad. Sci. USA 2013, 110, 11011–11016. [Google Scholar] [CrossRef] [Green Version]
  166. Wang, B.; Xiong, S.; Lin, S.; Xia, W.; Li, Q.; Zhao, Z.; Wei, X.; Lu, Z.; Wei, X.; Gao, P.; et al. Enhanced Mitochondrial Transient Receptor Potential Channel, Canonical Type 3-Mediated Calcium Handling in the Vasculature From Hypertensive Rats. J. Am. Heart Assoc 2017, 6, e005812. [Google Scholar] [CrossRef]
  167. Kitajima, N.; Numaga-Tomita, T.; Watanabe, M.; Kuroda, T.; Nishimura, A.; Miyano, K.; Yasuda, S.; Kuwahara, K.; Sato, Y.; Ide, T.; et al. TRPC3 positively regulates reactive oxygen species driving maladaptive cardiac remodeling. Sci. Rep. 2016, 6, 37001. [Google Scholar] [CrossRef] [Green Version]
  168. Miyagi, K.; Kiyonaka, S.; Yamada, K.; Miki, T.; Mori, E.; Kato, K.; Numata, T.; Sawaguchi, Y.; Numaga, T.; Kimura, T.; et al. A pathogenic C terminus-truncated polycystin-2 mutant enhances receptor-activated Ca2+ entry via association with TRPC3 and TRPC7. J. Biol. Chem. 2009, 284, 34400–34412. [Google Scholar] [CrossRef] [Green Version]
  169. Tiapko, O.; Groschner, K. TRPC3 as a Target of Novel Therapeutic Interventions. Cells 2018, 7, 83. [Google Scholar] [CrossRef]
Figure 1. A schematic drawing of the transient receptor potential canonical (TRPC) structure. Six transmembrane segments (numbered 1 to 6) contribute to the formation of a monomer. Both the COOH (C) and NH2 (N) terminus feature different domains enabling further channel interactions. These include coiled-coil domains (CC), ankyrin domains (ANK), a TRP box (TRP), a calmodulin, and IP3-R binding site (CIRB). Different outer potential glycosylation sites exist and differ among the different TRPC entities [19] ((A); inspired from [20]). Four monomers, from the same or different TRPC entities, assemble to form a homo- or heterotetramer. The loops between the transmembrane segments 5 and 6 contribute to the formation of the cation-permeable pore (P) ((B); view from above).
Figure 1. A schematic drawing of the transient receptor potential canonical (TRPC) structure. Six transmembrane segments (numbered 1 to 6) contribute to the formation of a monomer. Both the COOH (C) and NH2 (N) terminus feature different domains enabling further channel interactions. These include coiled-coil domains (CC), ankyrin domains (ANK), a TRP box (TRP), a calmodulin, and IP3-R binding site (CIRB). Different outer potential glycosylation sites exist and differ among the different TRPC entities [19] ((A); inspired from [20]). Four monomers, from the same or different TRPC entities, assemble to form a homo- or heterotetramer. The loops between the transmembrane segments 5 and 6 contribute to the formation of the cation-permeable pore (P) ((B); view from above).
Ijms 24 00181 g001
Figure 2. A schematic drawing of the renal tubular system. The glomerulus (brown) produces a primary urine that flows into the proximal convoluted tubule (red). Then, it reaches the proximal straight tubule before ending up in the loop of Henle, which is composed of a thin (dark blue) and a thick (green) part. The latter drains the urine into the distal convoluted tubule (purple) before reaching the collecting duct (light blue) via the connecting tubule, which is not specifically displayed here [24]. The magnifications detailed in (AD) reveal physiological mechanisms of the tubular Ca2+ reabsorption. Approximatively 98% of the filtered Ca2+ is reabsorbed. About 65% are reabsorbed in the proximal tubule, 25% in the thick ascending limb, and 8% in the distal and connecting tubule. The collecting duct plays a negligible role in this context. Most of the Ca2+ reabsorption in the proximal tubule is of paracellular nature and partly driven by the transcellular Na+ reabsorption mediated by the apical sodium/proton exchanger 3 (NHE3) and the basolateral Na+/K+-ATPase. Transcellular pathways and the roles of TRPC3, the plasma membrane calcium ATPase 1 (PMCA1), and the Na+/Ca2+-exchanger 1 (NCX1) are discussed below (A). The thick ascending limb features a regulated paracellular Ca2+ reabsorption which is driven by the Na+ reabsorption which is mediated by the Na+/K+/2Cl cotransporter (NKCC2) and the Na+/K+-ATPase. In terms of a negative feedback-loop, reabsorbed Ca2+ can activate basolateral calcium sensing receptors (CaSR) which in turn decrease the Na+ reabsorption and the paracellular claudin-mediated Ca2+ reabsorption (B). In the distal and connecting tubules, however, Ca2+ reabsorption is of a transcellular nature. Sodium enters the cell through the epithelial Na+ channel (ENaC) or the Na+/Cl cotransporter (NCC) and leaves it on the basolateral side using the Na+/K+-ATPase. The latter drives the Na+/Ca2+-exchanger 1 (NCX1) which shares the function of basal Ca2+ discharging with the plasma membrane calcium ATPase 4 (PMCA4). Apical Ca2+ entrance occurs using the vanilloid transient receptor potential 5 channel (TRPV5). Transcellular transport is mediated by calbindin D-28k (CB28) (C). Luminal Ca2+ concentration is sensed by calcium sensing receptors (CaSR) in the collecting duct. Their activation leads to the inhibition of aquaporin-2 (AQP-2)-mediated H2O reabsorption in the principal cells and activation of the H+-ATPase in the intercalated cells with subsequent urine acidification ultimately reducing the probability of crystal precipitation. Basolateral H2O transport occurs through aquaporins-3 and -4 (AQP-3/4) (D). Inspired by [25].
Figure 2. A schematic drawing of the renal tubular system. The glomerulus (brown) produces a primary urine that flows into the proximal convoluted tubule (red). Then, it reaches the proximal straight tubule before ending up in the loop of Henle, which is composed of a thin (dark blue) and a thick (green) part. The latter drains the urine into the distal convoluted tubule (purple) before reaching the collecting duct (light blue) via the connecting tubule, which is not specifically displayed here [24]. The magnifications detailed in (AD) reveal physiological mechanisms of the tubular Ca2+ reabsorption. Approximatively 98% of the filtered Ca2+ is reabsorbed. About 65% are reabsorbed in the proximal tubule, 25% in the thick ascending limb, and 8% in the distal and connecting tubule. The collecting duct plays a negligible role in this context. Most of the Ca2+ reabsorption in the proximal tubule is of paracellular nature and partly driven by the transcellular Na+ reabsorption mediated by the apical sodium/proton exchanger 3 (NHE3) and the basolateral Na+/K+-ATPase. Transcellular pathways and the roles of TRPC3, the plasma membrane calcium ATPase 1 (PMCA1), and the Na+/Ca2+-exchanger 1 (NCX1) are discussed below (A). The thick ascending limb features a regulated paracellular Ca2+ reabsorption which is driven by the Na+ reabsorption which is mediated by the Na+/K+/2Cl cotransporter (NKCC2) and the Na+/K+-ATPase. In terms of a negative feedback-loop, reabsorbed Ca2+ can activate basolateral calcium sensing receptors (CaSR) which in turn decrease the Na+ reabsorption and the paracellular claudin-mediated Ca2+ reabsorption (B). In the distal and connecting tubules, however, Ca2+ reabsorption is of a transcellular nature. Sodium enters the cell through the epithelial Na+ channel (ENaC) or the Na+/Cl cotransporter (NCC) and leaves it on the basolateral side using the Na+/K+-ATPase. The latter drives the Na+/Ca2+-exchanger 1 (NCX1) which shares the function of basal Ca2+ discharging with the plasma membrane calcium ATPase 4 (PMCA4). Apical Ca2+ entrance occurs using the vanilloid transient receptor potential 5 channel (TRPV5). Transcellular transport is mediated by calbindin D-28k (CB28) (C). Luminal Ca2+ concentration is sensed by calcium sensing receptors (CaSR) in the collecting duct. Their activation leads to the inhibition of aquaporin-2 (AQP-2)-mediated H2O reabsorption in the principal cells and activation of the H+-ATPase in the intercalated cells with subsequent urine acidification ultimately reducing the probability of crystal precipitation. Basolateral H2O transport occurs through aquaporins-3 and -4 (AQP-3/4) (D). Inspired by [25].
Ijms 24 00181 g002
Figure 3. The suggested role of TRPC3 in the proximal tubule cell. The cell is divided in two halves. The left half shows the proposed physiological function of TRPC3 in the proximal tubule. Luminal Ca2+ activates the calcium sensing receptor (CaSR), which triggers in turn the G-protein associated phospholipase C pathway. DAG is generated and increases TRPC3 activity. Receptor-operated Ca2+ entry (ROCE) results. Basolateral players including the plasma membrane Ca2+-ATPase 1 (PMCA1) and the Na+/Ca2+-exchanger 1 (NCX1) mediate the basal Ca2+ export. The right half shows a TRPC3-deficient proximal tubule cell which is exposed to alkaline hypercalciuric conditions. SOCE pathways outweigh ROCE pathways. Then, ER stress and ROS generation follow, along with subsequent calcification, inflammation, fibrosis, and apoptosis. Cellular debris accrue and promote stone formation which in turn aggravates the tubular damage. TRPC3 is suggested to contribute to prevention of such exacerbated luminal Ca2+ concentration and subsequently to attenuation of eventual cellular damage. Inspired by [31,109].
Figure 3. The suggested role of TRPC3 in the proximal tubule cell. The cell is divided in two halves. The left half shows the proposed physiological function of TRPC3 in the proximal tubule. Luminal Ca2+ activates the calcium sensing receptor (CaSR), which triggers in turn the G-protein associated phospholipase C pathway. DAG is generated and increases TRPC3 activity. Receptor-operated Ca2+ entry (ROCE) results. Basolateral players including the plasma membrane Ca2+-ATPase 1 (PMCA1) and the Na+/Ca2+-exchanger 1 (NCX1) mediate the basal Ca2+ export. The right half shows a TRPC3-deficient proximal tubule cell which is exposed to alkaline hypercalciuric conditions. SOCE pathways outweigh ROCE pathways. Then, ER stress and ROS generation follow, along with subsequent calcification, inflammation, fibrosis, and apoptosis. Cellular debris accrue and promote stone formation which in turn aggravates the tubular damage. TRPC3 is suggested to contribute to prevention of such exacerbated luminal Ca2+ concentration and subsequently to attenuation of eventual cellular damage. Inspired by [31,109].
Ijms 24 00181 g003
Figure 4. The postulated function of TRPC3 in the collecting duct. Arginine vasopressin stimulates the V2 vasopressin receptor (V2R). A signaling pathway is triggered leading to the generation of cyclic adenosine monophosphate (cAMP) and the activation of the protein kinase A (PKA). Membrane trafficking of aquaporin-2 (AQP-2) and TRPC3, as well as antidiuretic gene expression, is enhanced (CREB or cAMP response element-binding protein). TRPC3 is involved in Ca2+ reabsorption and sensing of hypotonicity by initiating Ca2+ signaling with direct and indirect stimulation of AQP-2 membrane trafficking. AQP-2 and AQP-3/4 mediate, respectively, apical and basolateral H2O flow along the osmotic gradient. The epithelial Na+ channel (ENaC) and renal outer medullary K+ channels (ROMK) mediate apical Na+ influx and K+ efflux.
Figure 4. The postulated function of TRPC3 in the collecting duct. Arginine vasopressin stimulates the V2 vasopressin receptor (V2R). A signaling pathway is triggered leading to the generation of cyclic adenosine monophosphate (cAMP) and the activation of the protein kinase A (PKA). Membrane trafficking of aquaporin-2 (AQP-2) and TRPC3, as well as antidiuretic gene expression, is enhanced (CREB or cAMP response element-binding protein). TRPC3 is involved in Ca2+ reabsorption and sensing of hypotonicity by initiating Ca2+ signaling with direct and indirect stimulation of AQP-2 membrane trafficking. AQP-2 and AQP-3/4 mediate, respectively, apical and basolateral H2O flow along the osmotic gradient. The epithelial Na+ channel (ENaC) and renal outer medullary K+ channels (ROMK) mediate apical Na+ influx and K+ efflux.
Ijms 24 00181 g004
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Englisch, C.N.; Paulsen, F.; Tschernig, T. TRPC Channels in the Physiology and Pathophysiology of the Renal Tubular System: What Do We Know? Int. J. Mol. Sci. 2023, 24, 181. https://doi.org/10.3390/ijms24010181

AMA Style

Englisch CN, Paulsen F, Tschernig T. TRPC Channels in the Physiology and Pathophysiology of the Renal Tubular System: What Do We Know? International Journal of Molecular Sciences. 2023; 24(1):181. https://doi.org/10.3390/ijms24010181

Chicago/Turabian Style

Englisch, Colya N., Friedrich Paulsen, and Thomas Tschernig. 2023. "TRPC Channels in the Physiology and Pathophysiology of the Renal Tubular System: What Do We Know?" International Journal of Molecular Sciences 24, no. 1: 181. https://doi.org/10.3390/ijms24010181

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