Next Article in Journal
Vitamins in the Prevention and Support Therapy of Neurodegenerative Diseases
Previous Article in Journal
The Role of Molecular Profiling in De-Escalation of Toxic Therapy in Breast Cancer
Previous Article in Special Issue
Aquaporins in Biliary Function: Pathophysiological Implications and Therapeutic Targeting
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Expanding Role of Aquaporin-1, Aquaporin-3 and Aquaporin-5 as Transceptors: Involvement in Cancer Development and Potential Druggability

by
Catarina Pimpão
1,2,
Inês V. da Silva
1,2 and
Graça Soveral
1,2,*
1
Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisboa, Portugal
2
Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisboa, Portugal
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(3), 1330; https://doi.org/10.3390/ijms26031330
Submission received: 29 December 2024 / Revised: 27 January 2025 / Accepted: 3 February 2025 / Published: 4 February 2025
(This article belongs to the Special Issue New Insights into Aquaporins: 2nd Edition)

Abstract

:
Aquaporins (AQPs) are transmembrane proteins that facilitate the transport of water and small solutes, including glycerol, hydrogen peroxide and ions, across cell membranes. Beyond their established physiological roles in water regulation and metabolic processes, AQPs also exhibit receptor-like signaling activities in cancer-associated signaling pathways, integrating the dual roles of transporters and receptors, hence functioning as transceptors. This dual functionality underpins their critical involvement in cancer biology, where AQPs play key roles in promoting cell proliferation, migration, and invasion, contributing significantly to carcinogenesis. Among the AQPs, AQP1, AQP3 and AQP5 have been consistently identified as being aberrantly expressed in various tumor types. Their overexpression is strongly associated with tumor progression, metastasis, and poor patient prognosis. This review explores the pivotal roles of AQP1, AQP3 and AQP5 as transceptors in cancer biology, underscoring their importance as pharmacological targets. It highlights the urgent need for the development of effective modulators to target these AQPs, offering a promising avenue to enhance current therapeutic approaches for cancer treatment.

1. Introduction

Aquaporins (AQPs) are transmembrane protein channels that facilitate the bidirectional transport of water, glycerol and other small non-charged solutes across cell membranes, being involved in water and energy homeostasis [1]. These proteins can be found in all living organisms, including archaea, eubacteria, fungi, plants and animals [2]. In humans, 13 AQP isoforms (AQP0-12) identified are widely distributed in the body and differentially expressed in each tissue and cell type [3]. According to their primary structure and pore selectivity, AQPs are classified into three subgroups: (1) orthodox or classical aquaporins (AQP0, AQP1, AQP2, AQP4, AQP5, AQP6 and AQP8), that are considered primarily selective to water; (2) aquaglyceroporins (AQP3, AQP7, AQP9 and AQP10) that, besides water, can also transport glycerol and other small neutral solutes; and (3) subcellular or unorthodox aquaporins (AQP11 and AQP12) [4] with a distinct evolutionary pathway, localized in intracellular membranes and with their permeability still uncertain [5], although AQP11 was found to permeate both water and glycerol [6]. A few of the above-mentioned isoforms can also transport ammonia (ammoniaporins) [7] and gases such as CO2 [8], O2 [9] and NO [10]. An overlapping subgroup, the peroxiporins (AQP0, AQP1, AQP3, AQP5, AQP8, AQP9, AQP11), comprises isoforms that facilitate the diffusion of hydrogen peroxide (H2O2) across membranes [11,12].
AQPs are assembled as homotetramers in cell membranes, where each monomer acts as an independent pore (Figure 1A,B) [13]. Each AQP monomer has approximately 28 kDa and is formed by six transmembrane helices and five connecting loops, with loop B and E containing the conserved Asn-Pro-Ala (NPA) motifs that form short α-helices that fold into the membrane to form the pore, leading to an hourglass structure (Figure 1C,D) [13,14]. AQP monomers have a cytoplasmic N- and C-terminal and present two main constriction sites responsible for pore selectivity and proton exclusion. The first one corresponds to the aromatic/arginine (ar/R) selectivity filter (SF), near the extracellular pore entrance, which determines the size of molecules allowed to be permeated, being responsible for the selectivity to water and/or small neutral solutes and forming the narrowest region of the pore (2.8 Å diameter pore for orthodox AQPs and >3.4 Å for aquaglyceroporins). The second constriction site is located at the center of the channel and consists of two half-helices containing the conserved NPA motifs that generate a dipole moment, creating a positive electrostatic barrier that prevents the passage of positively charged ions through the pore [15,16,17,18].

2. Physiological Roles of AQPs and Their Implication in Cancer

The investigation of aquaporins as therapeutic targets is emerging as a hot topic within the scientific community. Studies on AQP-knockout (KO) mice have provided information about the importance of AQPs in physiology such as renal water reabsorption, brain water homeostasis and metabolism but also in their implication in a broad range of disorders such as metabolic disorders, inflammation and cancer [19,20,21,22].
Numerous studies have reported that AQPs play key roles in tumor biology, facilitating cancer cell migration, proliferation and angiogenesis [23]. In particular, AQP1, AQP3 and AQP5, in addition to their physiological role in transepithelial water transport, skin hydration and fluid secretion [1,19,24,25], were found to be overexpressed in several types of cancer [26,27,28,29], such as colon cancer [30], hepatocellular carcinoma [31] and pancreatic ductal adenocarcinoma [32], being correlated with tumor progression, metastasis and poor patient prognosis, and implying their importance as prognostic markers and therapeutic targets for anticancer drug discovery. AQP4 has been consistently linked to glioblastoma progression contributing not only to cancer migration and invasion [33,34] but also influencing the glioma tumor microenvironment [35]. Similarly, AQP9 regulates the tumor microenvironment in kidney cancer [36], hepatocellular carcinoma [37] and colon cancer, where it modulates the polarization of tumor-associated macrophages [38]. Although less studied, AQP2, AQP6, AQP7 and AQP8 are also implicated in tumorigenesis. AQP2 is overexpressed in endometrial carcinoma by estrogen, promoting cancer migration, invasion and adhesion [39]. AQP6 and AQP7 can increase the resistance to oxidative stress in mesothelioma cells [40] and breast cancer cells, respectively, with AQP7 also altering cancer cell metabolism [41]. AQP8 overexpression reduces colorectal cancer growth and spread [42] but enhances cancer cell proliferation, migration and invasion in gliomas and cervical cancer [43,44].
Considering the number of studies reporting the consistent implication of AQP1, AQP3 and AQP5 in the development of multiple cancer types and their strong association with signaling pathways, this review is focused on the critical roles of these AQPs as transceptors involved in cancer progression.

2.1. AQP1, AQP3 and AQP5: Key Players in Cancer Progression

AQP1-null mice implanted with melanoma cells showed impaired tumor growth, reduced angiogenesis and decreased cell migration. These findings underline the crucial role of endothelial AQP1 in facilitating cancer cell migration [45].
A mechanism for the involvement of AQPs in cell migration has been proposed based on their role as membrane channels and their polarization at the leading edge of the lamellipodium in migrating cells. Local osmotic gradients created by the cleavage of actin and ion uptake at the lamellipodium tip induce fast water fluxes mediated by AQPs, required for the rapid changes in cell shape. These events cause an increase in the local hydrostatic pressure, leading to the expansion of the lamellipodium and creating spaces for actin polymerization and consequent cell migration (Figure 2) [46].
Cancer cell migration can contribute to tumor cell infiltration in the surrounding tissue and consequently lead to metastasis [23]. Since AQP1 is overexpressed in tumor endothelial cells, this protein is crucial for angiogenesis, endothelial cell migration and tumor growth, enhancing tumor infiltration and spread. This correlation was verified in studies with AQP1-null mice with melanoma [45,47] and breast cancer [48,49] as well as in colon and lung cancer cell lines [26,50]. A possible interaction between AQP1 and the vascular endothelial growth factor (VEGF) signaling pathway was reported, implicating AQP1 in tumor angiogenesis and endometrial adenocarcinoma progression [51,52]. In addition, AQP1 was shown to contribute to hypoxia-inducible angiogenesis in retinal vascular endothelial cells through a VEGF-independent mechanism [53]. Interestingly, hypoxia-induced AQP1 upregulation was correlated with the p38 mitogen activated protein kinase (MAPK) signaling pathway in prostate cancer cells [54].
AQP1 has also been related to epithelial–mesenchymal transition (EMT), a cellular process during which epithelial cells undergo phenotypic changes, losing their cell–cell adhesion and cell polarity while acquiring mesenchymal features that include invasiveness and cell motility [52]. AQP1 overexpression in lung adenocarcinoma was associated with EMT markers: loss of E-cadherin (epithelial marker) and increased expression of vimentin (mesenchymal marker), indicating the involvement of AQP1 in EMT and consequent invasive potential [55].
AQP3 is the aquaglyceroporin most associated with tumor progression, being overexpressed in different types of cancer such as skin [56,57], lung [58], colon [59], pancreatic [32] and liver cancer [60]. AQP3-null mice were found to be resistant to the development of skin tumors after exposure to a skin tumor initiator and phorbol ester promoter, exhibiting a reduction in epidermal cell proliferation. In addition, a notable decrease in glycerol and its metabolite glycerol-3-phosphate was detected, correlating with a reduction in ATP levels. After oral glycerol administration, ATP levels and epidermal cell proliferation were reestablished, suggesting that AQP3-facilitated glycerol permeability contributes to the generation of ATP and is implicated in tumor growth [1,56]. In several tumors, AQP3 expression was correlated with the activation of signaling pathways that promote cancer cell proliferation, migration and invasion [61]. For instance, human epidermal growth factor (EGF) upregulates AQP3 expression, thereby enhancing colorectal cancer cell migration via the phosphoinositide 3-kinase (PI3K)/Akt signaling pathway, being also associated with metastasis in colorectal cancer patients [62]. Likewise, AQP3 was reported to promote EMT through the activation of the PI3K/Akt/SNAIL signaling pathway in gastric cancer cells [63] and was found to contribute to stem-like properties, facilitating tumor growth via the Wnt/glycogen synthase kinase-3 beta (GSK-3β)/β-catenin signaling pathway [64]. In pancreatic cancer cells, AQP3 was found to promote tumor proliferation through the activation of the mammalian target of rapamycin (mTOR) signaling pathway [28]. Additionally, AQP3 overexpression activates the extracellular signal-regulated kinase (ERK)1/2 pathway in prostate cancer cells, leading to increased expression of matrix metalloproteinase-3 (MMP-3), which stimulates cell motility and invasion, further reinforcing AQP3’s contribution to cancer aggressiveness [65]. Furthermore, AQP3 suppression in esophageal and oral squamous cell carcinoma cells led to reduced cell adhesion and increased cell death through the inhibition of focal adhesion kinase (FAK) phosphorylation, decreasing the phosphorylation of ERK and MAPK pathways [66]. These findings highlight AQP3 as a critical mediator of EMT, tumor growth and metastasis, through its role in glycerol permeation, ATP production and the activation of key signaling pathways that contribute to tumor progression across various cancer types.
AQP5 involvement in tumor initiation and progression has been strongly linked with the activation of signaling pathways that can contribute to cancer cell migration, invasion and proliferation [67]. For example, AQP5 overexpression in colon cancer cells promotes cell proliferation via the Ras signaling pathway, probably mediated by the phosphorylation of the PKA consensus site of AQP5, and contributes to EMT by increasing phosphorylated Smad2/3 levels [68,69,70]. AQP5 silencing decreases cell invasion by modulating EMT-related proteins in colon cancer and hepatocellular carcinoma (HCC), but also through the downregulation of Wnt/β-catenin and the nuclear factor-kappa B (NF-ĸB) signaling pathways, respectively [71,72]. Additionally, AQP5 stimulates lung cancer cell proliferation and migration through the modulation of the epidermal growth factor receptor (EGFR)/ERK/p38 MAPK signaling pathway, while also upregulating the proliferation marker c-myc and increasing MUC5AC mucin production, revealing that AQP5 contributes to a higher metastatic and invasive potential [29]. In addition, the incubation of rat astrocytes in hypotonic media increased AQP5 expression, potentially regulated by the p38 MAPK pathway [73]. Thus, these findings indicate that AQP5 overexpression in cancer cells boosts their metastatic and invasive potential by modulating key signal transduction pathways, contributing to tumor progression.
In colorectal cancer progression, AQP1 and AQP5 expression was found to be induced in the early stage of the disease and maintained stability through the late stage of cancer development [74]. Interestingly, AQP3 and AQP5 expression patterns vary during the progression of pancreatic ductal adenocarcinoma. AQP3 expression levels are increased with cancer development from the early to late stages of the tumor, while AQP5 expression is augmented in the early stage but almost undetectable in later stages, showing its potential as a biomarker of this disease. In the same study, AQP3 and AQP5 were associated with increased levels of EGFR, proliferation marker Ki-67, cytokeratin 7 and vimentin, while E-cadherin was found to be decreased, indicating the contribution of these AQP isoforms to EMT, tumor growth and invasion [32]. The correlation of AQP3 and AQP5 with EMT and tumorigenesis has also been reported in triple-negative breast cancer and hepatocellular carcinoma patients, suggesting the potential of AQP3 and AQP5 as prognostic and therapeutic markers [31,75]. Recently, AQP1, AQP3 and AQP5 overexpression was reported to disrupt cell polarity in breast cancer by interacting with the protein Scribble, leading to its downregulation [76,77].
Furthermore, our group reported that both AQP3 and AQP5 play a critical role in cancer cell migration in pancreatic cancer where cells silenced for AQP3 and AQP5 and those double-silenced (AQP3/AQP5) exhibited a significant impairment in cell migration [78,79]. Interestingly, AQP3-, AQP5- and double-silenced human pancreatic ductal adenocarcinoma cells exhibited morphological alterations and reduced cell–cell adhesion, with AQP5 additionally affecting cell stiffness and membrane fluidity. These findings suggest that AQP3 and AQP5 may influence tumor initiation and progression by modulating cellular morphological and biomechanical properties [79].

2.2. Peroxiporin Activity of AQP1, AQP3 and AQP5

H2O2 is the main reactive oxygen species (ROS) acting as a signaling molecule in redox signaling [80]. At low concentrations, H2O2 can regulate several physiological processes such as cell differentiation, proliferation, survival and immune response via the reversible oxidation of specific protein targets and consequent alteration of their activity [81]. However, at high concentrations, H2O2 promotes the damage of biomolecules such as proteins, lipids and nucleic acids, and leads to abnormal induction of signaling pathways due to oxidative stress, suggesting its key role in tumorigenesis [82,83,84]. Therefore, the role of AQP1, AQP3 and AQP5 in tumor development is closely associated with their peroxiporin function [12,85]. For instance, AQP3-mediated H2O2 uptake was correlated with the activation of the EGFR signaling pathway in human squamous cell carcinoma and lung cancer cell lines, contributing to cancer progression [86]. In human breast cancer cell lines, AQP3 peroxiporin activity was found to be implicated in CXCL12/CXCR4-dependent breast cancer cell migration. Extracellular H2O2 produced by the CXCL12-activated membrane NADPH oxidase 2 (NOX2) is taken up by breast cancer cells through AQP3 with consequent activation of H2O2-mediated signaling pathways and stimulation of cancer cell migration [86]. Similarly, AQP3 contributes to cervical cancer infiltration and metastasis by mediating NOX4-derived H2O2 transport and activating the Syk/PI3K/Akt signaling pathway [87]. Moreover, H2O2 transport via AQP3 can promote tissue inflammation through the NF-ĸB signaling pathway and macrophage activation [88], contributing to the inflammatory response [20,89]. The inhibition of AQP3 peroxiporin activity in melanoma cell lines was recently reported, impairing cancer cell migration, proliferation and adhesion and highlighting the impact of AQP3 on melanoma progression [57].
The involvement of AQP5 in intracellular signaling pathways implicated in tumorigenesis may be attributed to its ability to facilitate H2O2 transport across cell membranes [67]. Interestingly, AQP5 is often found phosphorylated in cancer as opposed to normal cells, indicating that AQP5 phosphorylation could be related to its role in tumor initiation and development [90]. Moreover, yeast cells transformed to express AQP5 showed increased sensitivity to oxidative damage at long-term exposure to H2O2, while in the short term, AQP5-mediated H2O2 permeability was correlated with cell growth and survival. In addition, AQP5 overexpression in pancreatic cancer cells was found to promote cancer cell migration due to its peroxiporin activity [78].
The contribution of AQP1, AQP3 and AQP5 to oxidative stress resistance has been investigated in colon and breast cancer cell lines by assessing their expression before and after treatment with H2O2. Regarding colorectal cancer, the HT-29 cell line exhibited the highest expression of the three aquaporin isoforms, with no significant changes following H2O2 exposure, similar to the SW620 cell line. In contrast, H2O2 treatment upregulated AQP5 in Caco-2 cells and increased AQP1 and AQP3 expression in HCT116 cells [91]. In breast cancer, H2O2 treatment increased AQP3 expression in MCF7 (estrogen- and progesterone-receptor-positive) and SkBr3 (HER2-positive) cells but downregulated AQP3 in SUM159 (triple-negative) cells, indicating that these cells are less responsive to oxidative stress which aligns with their more aggressive and metastatic phenotype. AQP1 and AQP5 expression decreased in MCF7 cells and was upregulated in SkBr3 and SUM159 cells after the addition of H2O2 [92]. These findings suggest an adaptive role of aquaporins to stress, which can contribute to the malignancy of tumors and resistance to therapy.

3. Role of AQP1, AQP3 and AQP5 as Transceptors in Cancer

In addition to their transport activity, the overexpression of AQP1, AQP3 and AQP5 in cancer has been associated with signal transduction pathways and cytoskeleton reorganization, resembling mechanisms typically mediated by receptors [93]. In fact, AQP1 has been reported to interact with focal adhesion kinases, promoting cell migration in chick neural crest cells and bone marrow mesenchymal stem cells [94,95], and to modulate actin cytoskeleton reorganization by interacting with Lin-7/β-catenin in human endothelial and melanoma cells [96]. Additionally, in estrogen receptor (ER)-positive breast cancer cells, upregulation of AQP3 contributes to cell migration and invasion through the modulation of EMT-related markers and reorganization of the actin cytoskeleton [97]. AQP3 co-localizes with ezrin, influencing both ezrin and actin organization, as well as the formation of lamellipodia and filopodia, with further impact on cell migration and invasion in endometrial epithelial cells [98]. Similarly, AQP5 interacts with ezrin, forming complexes that are mislocated in the salivary glands of Sjögren’s syndrome patients [99]. Interestingly, the trafficking of AQP5 to the plasma membrane depends on the interaction between AQP5-containing vesicles and the cytoskeleton [100,101], a mechanism also observed for AQP1 [102]. Furthermore, AQP5 was found to promote microtubule assembly and stabilization, with a potential impact on paracellular permeability in human airway epithelial cells [103].
The transceptor activity of AQP1, AQP3 and AQP5 could also be related to their interactions with other proteins. For instance, AQP1 interacts with the amyloid precursor protein, which can upregulate AQP1 expression in Alzheimer’s disease patients’ brains [104]. AQP3 forms complexes with ClC-3 chloride channels, regulating ClC-3 channel gating, with both proteins involved in cell volume regulation in nasopharyngeal carcinoma cells [105,106]. Moreover, AQP3 binds to phospholipase D2 (PLD2) in keratinocytes, facilitating glycerol transport to support PLD2-mediated phosphatidylglycerol synthesis [107]. In extravillous trophoblast cells, the disruption of the complex AQP3-caveolin-1 can contribute to pregnancy disorders [108]. In addition, AQP3 can interact with the lipid droplet protein perilipin 1 (PLIN1) [109]. Regarding AQP5-interacting partners, AQP5 co-localizes with the Na+-K+-Cl co-transporter 1 (NKCC1) and anion exchanger 2 (AE2) in mouse salivary gland acinar cells and human embryonic kidney (HEK293) cells [110]. Similar to ezrin, prolactin-inducible protein (PIP) can interact with AQP5 [111], controlling its localization in Sjögren’s syndrome patients’ salivary glands [112]. In addition, AQP5 interacts with MUC5AC in rabbit and mouse conjunctival epithelial cells, both being upregulated during acute dry eye stress [113]. In HEK293 cells, AQP5 interaction with ZO-1, plakoglobin, β-catenin and desmoglein-2 resulted in their decreased expression [114]. Importantly, phosphorylated AQP5 binds to c-Src in non-small cell lung cancer cells, promoting EMT [90].
These findings indicate a potential role of AQPs as transceptors, acting both as transporters and receptors in the cell membranes [93]. This novel concept for aquaporins can be explored by investigating their interplay with signaling pathways and cytoskeleton elements and the identification of novel potent and selective AQP modulators that can impact tumor growth and development (Figure 3).

4. AQP1, AQP3 and AQP5 as Druggable Targets for Pharmacological Modulators

Considering their involvement in a wide array of diseases, AQPs have emerged as promising targets for drug discovery, prompting the identification of new potent and selective AQP modulators with therapeutic potential. However, the discovery of selective AQP inhibitors has proven to be very challenging due to the high similarity between AQP isoforms, their widespread expression across different types of tissues and cells and the difficulty of targeting their narrow pores [25,115]. Several pharmacological molecules have been reported to modulate AQP activity and/or expression, demonstrating their therapeutic effects on diseases with abnormal AQP expression (Table 1).
Mercury compounds, such as HgCl2, have been found to inhibit AQP1 by binding to the Cys189 residue and preventing the passage of water molecules [116] and were also reported to inhibit other AQPs, including AQP3 and AQP5 [78,117,118]. Interestingly, AQP3 inhibition by HgCl2 can improve the sensitivity of prostate cancer cells to cryotherapy [119]. However, the high toxicity and non-specificity of these compounds renders them unsuitable for therapeutic applications [120,121]. In addition, silver compounds, including silver nitrate and silver sulfadiazine, have demonstrated a rapid and irreversible inhibition of AQP1 [122]. Organogold compounds have emerged as potent AQP inhibitors, specifically targeting aquaglyceroporins such as AQP3 and AQP10. In particular, the gold(III) complex [Au(phen)Cl2]Cl (phen = 1,10-phenantroline, Auphen) [123] was reported to decrease cell proliferation in AQP3-expressing cells [124] and exhibited an inhibitory effect in red blood cells that endogenously express AQP3, with an IC50 of 0.8 ± 0.08 µM [123]. The binding mechanism is likely to occur at the Cys40 residue of AQP3, leading to a conformational change of the protein and consequent inhibition of glycerol permeability [125]. This gold compound was also found to affect AQP7 activity in adipocytes [126,127]. Auphen was reported to exert a therapeutic effect in several pathologies, including hepatocellular carcinoma [128] and inflammation [89], where AQP3 plays an important role. Recently, a new gold compound derivative with the general formula [Au(C^N)Cl2] (C^N = cyclometalated ligand) was found to irreversibly inhibit human AQP10 (hAQP10) in a yeast cell model overexpressing this AQP through the formation of a gold adduct followed by cysteine arylation, leading to the inhibition of AQP10-mediated glycerol transport [129]. Moreover, this new class of organogold compounds has demonstrated strong inhibition of AQP3 peroxiporin activity in melanoma cell lines, leading to an impairment in melanoma cell adhesion, cell proliferation and cell migration [57]. Remarkably, these compounds have also exhibited anticancer activity in vivo [130]. Recently, the organogold complex ST004 was identified as an AQP3 inhibitor, with its liposomal formulation demonstrating anticancer activity against melanomas in both in vitro and in vivo models [131]. Metallodrugs such as [Cu(phen)Cl2] (Cuphen) and polyoxotungstate P2W18 have also been described as potent AQP3 inhibitors, showing anticancer properties [132,133,134,135].
Alongside metallodrugs, small molecules have been validated as potential AQP modulators. Quaternary ammonium ion tetraethylammonium (TEA) and acetazolamide were identified as AQP1 inhibitors in AQP1-expressing Xenopus laevis oocytes [136,137] while bumetanide derivatives (AqB013 and AqB011) act as AQP1 blockers [138,139] and furosemide derivative AqF026 functions as an AQP1 activator [140]. However, the modulation of AQP1 activity by TEA, acetazolamide, AqB013 and AqF026 was not validated in other cell models, disclosing the importance of confirming AQP modulation in various AQP expression systems [141,142]. Ciglitazone, a peroxisome proliferator-activated receptor gamma (PPARγ) activator; suberanilohydroxamic acid (SAHA), a histone deacetylase (HDAC) inhibitor; and monomethylfumarate (MMF), an antipsoriatic agent, were reported to increase AQP3 expression levels in human and mouse keratinocytes, promoting glycerol uptake [143,144,145], with SAHA also upregulating AQP5 expression in mouse lung epithelial cells [146]. Bisacodyl decreased AQP3 expression in rat colons, leading to an increase in fecal water content [147]. Moreover, compounds DFP00173 and Z433927330 were shown as potent AQP3 and AQP7 inhibitors, exerting their inhibitory effect through the AQP cytoplasmic entrance [148]. Recently, DFP00173 was found to reduce multiple myeloma cell viability, tumor growth, mitochondrial respiration and electron transport chain complex I activity through AQP3 blockage [149]. Niclosamide was found to upregulate AQP5 expression, which could be beneficial in pathologies with reduced AQP5 levels such as acute lung injury [150], while methazolamide was reported to reduce AQP5 expression, with an impact on the migration of immune cells [151].
Several natural compounds have been identified as modulators of AQP expression. Bacopaside II blocks AQP1-mediated H2O2 permeability, decreasing the stress-induced hypertrophic remodeling of the heart [152]. All-trans retinoic acid (atRA), chrysin and glycolic acid can reverse the UV-induced reduction in AQP3 expression in human keratinocytes, highlighting their protective role against photoaging [153,154,155], with both atRA and chrysin upregulating AQP3 expression through the ERK and redox signaling pathways [153,154]. In human keratinocytes, resveratrol can reduce AQP3 expression via SIRT1/ARNT/ERK signaling, decreasing cell proliferation [156] while 18β-glycyrrhetinic acid derivative upregulates AQP3 and stimulating dermal fibroblast proliferation and migration [157]. In addition, curcumin inhibits EGF-induced AQP3 upregulation, impairing ovarian cancer cell migration [158]. Daiokanzoto and a resin glycoside fraction from Pharbitis Semen (RFP) reduce AQP3 expression in rat colons, inducing diarrhea [159,160], while naringenin upregulates AQP3 expression levels in a mouse model of constipation [161]. Additionally, β-patchoulene was found to decrease AQP3 expression levels in rats with induced intestinal mucositis through inactivation of the cAMP/PKA/CREB signaling pathway [162]. Recently, we identified rottlerin as an AQP3 inhibitor, acting as a stereochemical lid on the AQP3 pore entrance, blocking AQP3-mediated glycerol transport [163].
AQP1, AQP3 and AQP5 expression and function can also be affected by naturally occurring molecules with potential therapeutic effects, including hormones, microRNAs (miRs) and antibodies. Steroid hormones, including estrogen and progesterone, and arachidonic acid were found to significantly increase AQP1 and AQP5 expression in porcine uteri through the modulation of the PKA and MAPK signaling pathways [164,165,166]. AQP3 is also upregulated by estrogen, being implicated in the stimulation of cell migration and invasion in ER-positive breast cancer cells through the modulation of EMT-related factors and the reorganization of the actin cytoskeleton [97] and in the development of the chicken oviduct and ovarian cancer [167]. In fact, both the AQP3 and AQP5 promoter regions contain a functional estrogen response element that can be activated directly by estrogen [97,168]. In addition, Skowronski and collaborators reported that gonadotropins, prolactin and growth hormone upregulate AQP1 levels in porcine ovarian follicular cells [169]. Testosterone was also reported to upregulate AQP1, AQP3 and AQP5 levels in ovariectomized rats [170,171]. Uroguanylin can trigger lipolysis in human visceral adipocytes via the upregulation of lypolisis-related genes such as AQP3, being usually decreased in obese patients with type 2 diabetes [172]. In addition, leptin administration can ameliorate non-alcoholic fatty liver (NAFLD) disease in leptin-deficient mice decreasing AQP3 expression levels [173]. In acute renal failure, erythropoietin was able to hinder the ischemia-induced downregulation of AQP3, impacting the urinary concentrating ability [174]. Furthermore, both AQP3 and AQP5 were upregulated by dexamethasone and ambroxol in human airway epithelial cells, being able to regulate airway hypersecretion, a typical symptom in several pulmonary diseases [175].
Recently, miRs have been highlighted as a promising tool to modulate AQP expression and function, with therapeutic potential in pathologies characterized by AQP dysregulation. miR-320 can reduce AQP1 expression in breast cancer, decreasing anthracycline chemosensitivity and impairing tumor proliferation, migration and invasion [176,177]. Similarly, miR-1226-3p, miR-19a-3p and miR-19b-3p suppress AQP5 expression in breast cancer, contributing to reduced cell migration [178]. miR-874 targets both AQP1 and AQP3 in several pathologies, mitigating inflammation and myocardial disfunction in sepsis by inhibiting AQP1 [179], and downregulating AQP3 expression in various cancers including gastric cancer [180], pancreatic ductal adenocarcinoma [28] and non-small cell lung cancer (NSCLC) [181], impairing tumor development. This miRNA also contributes to intestinal barrier dysfunction via AQP3 modulation [182,183]. miR-185 regulates AQP3 and AQP5 expression, exerting its therapeutic effect by compromising squamous cell carcinoma and colorectal cancer progression [184,185], whereas miR-877 can suppress AQP3 in gastric cancer, promoting apoptosis and reducing cell proliferation, invasion and EMT [186]. In hepatocellular carcinoma, miR-124 can decrease AQP3 expression, inhibiting cell proliferation and migration [60], while miR-1271-5p can prevent hepatitis B-virus-mediated liver cancer growth in vivo by decreasing the expression of AQP5 [187]. In addition, miR-21 can downregulate AQP5 in gallbladder carcinoma [188] and miR-29a can increase the intestinal membrane permeability of colonic epithelial cells by reducing AQP1 and AQP3 expression in diarrhea-predominant irritable bowel syndrome (IBS-D) [189]. In disseminated intravascular coagulation, miR-96 and miR-330 can lower AQP5 expression, leading to pulmonary edema [190]. In sepsis, miR-133a-3p downregulates AQP1, resulting in increased expression of inflammatory cytokines [191]. In acute lung injury, miR-126-5p and miR-144-3p were found to reduce AQP1 expression, improving fluid clearance and promoting apoptosis [161,192]. miR-495 can inhibit AQP1 expression through the p38 MAPK signaling pathway, enhancing osteoblast proliferation and differentiation in mice with tibial fractures [193].
The urgent need for specific AQP modulators has prompted efforts to develop antibodies capable of selectively targeting AQP isoforms. An anti-AQP3 monoclonal antibody was designed to target a specific extracellular epitope of AQP3 in macrophages. In a mouse model of liver injury, the anti-AQP3 antibody suppressed inflammation by inhibiting AQP3-mediated H2O2 permeability, thereby downregulating the NF-ĸB signaling pathway and reducing macrophage activation [88]. Recently, this antibody exhibited therapeutic effects in colorectal cancer by suppressing tumor growth, reducing immunosuppressive M2-like tumor-associated macrophages, and maintaining T cell anti-tumor activity within the tumor microenvironment through AQP3 targeting [194]. In addition, the anti-AQP3 monoclonal antibody was found to reduce multiple myeloma cell viability, tumor growth, mitochondrial respiration and electron transport chain complex I activity via AQP3 inhibition [149]. These findings disclose the pivotal role of AQP3 in oxidative stress, inflammation and cancer, while also highlighting the potential of antibody-based therapies for the treatment of AQP-overexpressing pathologies.
Further research is needed to address the current challenges in AQP druggability by improving the selectivity and efficiency of aquaporin modulators and developing targeted therapies for their precise delivery to cancer tissues. Significant progress has been made via the development of anti-AQP antibodies [88] and via the encapsulation of AQP3 inhibitors such as Cuphen [132,134] and ST004 [131] in liposomal formulations designed to target cancer tissues. In addition, the development of AQP modulators that selectively affect AQP receptor activity without influencing their transporter function would significantly improve our understanding of their roles in cancer development and metastasis.
Table 1. Modulators of AQP1 AQP3 and AQP5 expression and function.
Table 1. Modulators of AQP1 AQP3 and AQP5 expression and function.
AQPModulatorEffect on AQPTargeted DiseaseReferences
AQP1HgCl2Inhibition-[116]
Silver nitrateInhibition-[122]
Silver sulfadiazineInhibition-[122]
AqB011InhibitionColon cancer[139]
Bacopaside IIInhibitionCardiac hypertrophy[152]
EstrogenUpregulation-[164,165,166]
ProgesteroneUpregulation-[164,165,166]
Arachidonic acidUpregulation-[164,165,166]
GonadotropinsUpregulation-[169]
ProlactinUpregulation-[169]
Growth hormoneUpregulation-[169]
TestosteroneUpregulation-[171]
miR-29aDownregulationIBS-D[189]
miR-126-5pDownregulationAcute lung injury[192]
miR-133a-3pDownregulationSepsis[191]
miR-144-3pDownregulationAcute lung injury[195]
miR-320DownregulationBreast cancer[176,177]
miR-495Downregulation-[193]
miR-874DownregulationSepsis[179]
AQP3HgCl2InhibitionProstate cancer[117,118,119]
AuphenInhibitionEpidermoid carcinoma; hepatocellular carcinoma; inflammation; melanoma; triple-negative breast cancer[57,89,123,124,125,128,130]
Au(III) CNHNInhibitionMelanoma[57]
Au(III) CCONInhibitionMelanoma[57]
ST004InhibitionMelanoma[131]
CuphenInhibitionMelanoma; colon cancer[132,133,134]
P2W18InhibitionMelanoma[135]
CiglitazoneUpregulation-[143]
SAHAUpregulation-[144]
MMFUpregulationPsoriasis[145]
BisacodylDownregulationConstipation[147]
DFP00173InhibitionMultiple myeloma[148,149]
atRAUpregulationSkin photoaging[153]
ChrysinUpregulationSkin photoaging[154]
Glycolic acidUpregulationSkin photoaging[155]
ResveratrolDownregulation-[156]
18β-glycyrrhetinic acidUpregulation-[157]
CurcuminDownregulationOvarian cancer[158]
DaiokanzotoDownregulationConstipation[159]
RFPDownregulationConstipation[160]
NaringeninUpregulationConstipation[161]
β-patchouleneDownregulationIntestinal mucositis[162]
RottlerinInhibition-[163]
EstrogenUpregulationER-positive breast cancer; ovarian cancer[97,167]
TestosteroneUpregulation-[170]
UroguanylinUpregulation-[172]
LeptinDownregulationObesity; NAFLD[173]
ErythropoietinUpregulationAcute renal failure[174]
DexamethasoneUpregulation-[175]
AmbroxolUpregulation-[175]
miR-29aDownregulationIBS-D[189]
miR-124DownregulationHepatocellular carcinoma[60]
miR-185DownregulationSquamous cell carcinoma[184]
miR-874DownregulationGastric cancer; pancreatic ductal adenocarcinoma; NSCLC; intestinal barrier dysfunction[28,180,181,182,183]
miR-877DownregulationGastric cancer[186]
Anti-AQP3 monoclonal antibodyInhibitionLiver injury; colorectal cancer; multiple myeloma[88,149,194]
AQP5HgCl2InhibitionPancreatic ductal adenocarcinoma[78]
SAHAUpregulation-[146]
NiclosamideUpregulation-[150]
MethazolamideDownregulationSepsis[151]
EstrogenUpregulation-[164,165,166]
ProgesteroneUpregulation-[164,165,166]
Arachidonic acidUpregulation-[164,165,166]
TestosteroneUpregulation-[171]
DexamethasoneUpregulation-[175]
AmbroxolUpregulation-[175]
miR-19a-3pDownregulationBreast cancer[178]
miR-19b-3pDownregulationBreast cancer[178]
miR-21DownregulationGallbladder carcinoma[188]
miR-96DownregulationDisseminated intravascular coagulation[190]
miR-185DownregulationColorectal cancer[185]
miR-330DownregulationDisseminated intravascular coagulation[190]
miR-1226-3pDownregulationBreast cancer[178]
miR-1271-5pDownregulationHepatitis B-virus-mediated liver cancer[187]

5. Final Remarks

This review focuses on the pivotal roles of aquaporins AQP1, AQP3 and AQP5 in tumor biology, emphasizing their significant contributions to cancer progression. The overexpression of these aquaporins has been strongly associated with enhanced tumor growth, increased metastatic potential and poor clinical outcomes in patients. While traditionally recognized for their primary role in facilitating the transport of water, glycerol and other small molecules, such as H2O2, across cellular membranes, recent studies have unveiled their broader functionality. These proteins also act as transceptors, integrating transport and signaling roles to mediate signal transduction and the activation of critical pathways involved in tumor initiation and progression.
Given their multifaceted contributions to cancer biology, tumor aquaporins emerge as compelling therapeutic targets. Their involvement extends beyond basic transport functions to include regulatory roles in key oncogenic signaling cascades. This dual functionality underscores the need for innovative therapeutic strategies aimed at modulating their activity. Consequently, there is a growing interest in designing and identifying novel, potent and selective modulators capable of targeting the unique properties of AQP-overexpressing tumors. By addressing the challenges associated with their dysregulation, these advancements could pave the way for more effective treatments and improved prognoses for patients with AQP-driven cancers.

Author Contributions

Conceptualization: C.P. and G.S.; initial draft preparation, C.P., I.V.d.S., G.S.; illustration: C.P. and I.V.d.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by FCT—Fundação para a Ciência e Tecnologia, Portugal, grant number 2022.06601.PTDC, 2020.04974.BD to C.P., researcher contract 10.54499/2022.03691.CEECIND/CP1743/CT0001 to I.V.d.S., and strategic project UID 04138—Instituto de Investigação do Medicamento (iMed.ULisboa).

Data Availability Statement

No new data were created or analyzed in this study.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Verkman, A.S.; Anderson, M.O.; Papadopoulos, M.C. Aquaporins: Important but elusive drug targets. Nat. Rev. Drug Discov. 2014, 13, 259–277. [Google Scholar] [CrossRef] [PubMed]
  2. Zardoya, R. Phylogeny and evolution of the major intrinsic protein family. Biol. Cell 2005, 97, 397–414. [Google Scholar] [CrossRef]
  3. Soveral, G.; Nielsen, S.; Casini, A. Aquaporins in Health and Disease; CRC Press: Boca Raton, FL, USA, 2018. [Google Scholar]
  4. Ishibashi, K.; Tanaka, Y.; Morishita, Y. The role of mammalian superaquaporins inside the cell. Biochim. Biophys. Acta 2014, 1840, 1507–1512. [Google Scholar] [CrossRef]
  5. Ishibashi, K.; Tanaka, Y.; Morishita, Y. The role of mammalian superaquaporins inside the cell: An update. Biochim. Biophys. Acta Biomembr. 2021, 1863, 183617. [Google Scholar] [CrossRef] [PubMed]
  6. Madeira, A.; Fernandez-Veledo, S.; Camps, M.; Zorzano, A.; Moura, T.F.; Ceperuelo-Mallafre, V.; Vendrell, J.; Soveral, G. Human aquaporin-11 is a water and glycerol channel and localizes in the vicinity of lipid droplets in human adipocytes. Obesity 2014, 22, 2010–2017. [Google Scholar] [CrossRef] [PubMed]
  7. Jahn, T.P.; Moller, A.L.; Zeuthen, T.; Holm, L.M.; Klaerke, D.A.; Mohsin, B.; Kuhlbrandt, W.; Schjoerring, J.K. Aquaporin homologues in plants and mammals transport ammonia. FEBS Lett. 2004, 574, 31–36. [Google Scholar] [CrossRef] [PubMed]
  8. Geyer, R.R.; Musa-Aziz, R.; Qin, X.; Boron, W.F. Relative CO2/NH3 selectivities of mammalian aquaporins 0–9. Am. J. Physiol. Cell Physiol. 2013, 304, C985–C994. [Google Scholar] [CrossRef]
  9. Zwiazek, J.J.; Xu, H.; Tan, X.; Navarro-Rodenas, A.; Morte, A. Significance of oxygen transport through aquaporins. Sci. Rep. 2017, 7, 40411. [Google Scholar] [CrossRef] [PubMed]
  10. Herrera, M.; Hong, N.J.; Garvin, J.L. Aquaporin-1 transports NO across cell membranes. Hypertension 2006, 48, 157–164. [Google Scholar] [CrossRef]
  11. Prata, C.; Hrelia, S.; Fiorentini, D. Peroxiporins in Cancer. Int. J. Mol. Sci. 2019, 20, 1371. [Google Scholar] [CrossRef] [PubMed]
  12. da Silva, I.V.; Mlinaric, M.; Lourenco, A.R.; Perez-Garcia, O.; Cipak Gasparovic, A.; Soveral, G. Peroxiporins and Oxidative Stress: Promising Targets to Tackle Inflammation and Cancer. Int. J. Mol. Sci. 2024, 25, 8381. [Google Scholar] [CrossRef]
  13. King, L.S.; Kozono, D.; Agre, P. From structure to disease: The evolving tale of aquaporin biology. Nat. Rev. Mol. Cell Biol. 2004, 5, 687–698. [Google Scholar] [CrossRef] [PubMed]
  14. Verkman, A.S.; Mitra, A.K. Structure and function of aquaporin water channels. Am. J. Physiol. Ren. Physiol. 2000, 278, F13–F28. [Google Scholar] [CrossRef] [PubMed]
  15. Murata, K.; Mitsuoka, K.; Hirai, T.; Walz, T.; Agre, P.; Heymann, J.B.; Engel, A.; Fujiyoshi, Y. Structural determinants of water permeation through aquaporin-1. Nature 2000, 407, 599–605. [Google Scholar] [CrossRef] [PubMed]
  16. de Groot, B.L.; Frigato, T.; Helms, V.; Grubmuller, H. The mechanism of proton exclusion in the aquaporin-1 water channel. J. Mol. Biol. 2003, 333, 279–293. [Google Scholar] [CrossRef] [PubMed]
  17. Wu, B.; Steinbronn, C.; Alsterfjord, M.; Zeuthen, T.; Beitz, E. Concerted action of two cation filters in the aquaporin water channel. EMBO J. 2009, 28, 2188–2194. [Google Scholar] [CrossRef] [PubMed]
  18. Wree, D.; Wu, B.; Zeuthen, T.; Beitz, E. Requirement for asparagine in the aquaporin NPA sequence signature motifs for cation exclusion. FEBS J. 2011, 278, 740–748. [Google Scholar] [CrossRef] [PubMed]
  19. da Silva, I.V.; Rodrigues, J.S.; Rebelo, I.; Miranda, J.P.G.; Soveral, G. Revisiting the metabolic syndrome: The emerging role of aquaglyceroporins. Cell Mol. Life Sci. 2018, 75, 1973–1988. [Google Scholar] [CrossRef] [PubMed]
  20. da Silva, I.V.; Soveral, G. Aquaporins in Immune Cells and Inflammation: New Targets for Drug Development. Int. J. Mol. Sci. 2021, 22, 1845. [Google Scholar] [CrossRef] [PubMed]
  21. Abe, Y.; Yasui, M. Aquaporin-4 in Neuromyelitis Optica Spectrum Disorders: A Target of Autoimmunity in the Central Nervous System. Biomolecules 2022, 12, 591. [Google Scholar] [CrossRef]
  22. Bhattacharjee, A.; Jana, A.; Bhattacharjee, S.; Mitra, S.; De, S.; Alghamdi, B.S.; Alam, M.Z.; Mahmoud, A.B.; Al Shareef, Z.; Abdel-Rahman, W.M.; et al. The role of Aquaporins in tumorigenesis: Implications for therapeutic development. Cell Commun. Signal. 2024, 22, 106. [Google Scholar] [CrossRef] [PubMed]
  23. Papadopoulos, M.C.; Saadoun, S. Key roles of aquaporins in tumor biology. Biochim. Biophys. Acta 2015, 1848, 2576–2583. [Google Scholar] [CrossRef] [PubMed]
  24. da Silva, I.V.; Silva, A.G.; Pimpao, C.; Soveral, G. Skin aquaporins as druggable targets: Promoting health by addressing the disease. Biochimie 2021, 188, 35–44. [Google Scholar] [CrossRef]
  25. Pimpao, C.; Wragg, D.; da Silva, I.V.; Casini, A.; Soveral, G. Aquaglyceroporin Modulators as Emergent Pharmacological Molecules for Human Diseases. Front. Mol. Biosci. 2022, 9, 845237. [Google Scholar] [CrossRef]
  26. Wei, X.; Dong, J. Aquaporin 1 promotes the proliferation and migration of lung cancer cell in vitro. Oncol. Rep. 2015, 34, 1440–1448. [Google Scholar] [CrossRef]
  27. Oishi, M.; Munesue, S.; Harashima, A.; Nakada, M.; Yamamoto, Y.; Hayashi, Y. Aquaporin 1 elicits cell motility and coordinates vascular bed formation by downregulating thrombospondin type-1 domain-containing 7A in glioblastoma. Cancer Med. 2020, 9, 3904–3917. [Google Scholar] [CrossRef] [PubMed]
  28. Huang, X.; Huang, L.; Shao, M. Aquaporin 3 facilitates tumor growth in pancreatic cancer by modulating mTOR signaling. Biochem. Biophys. Res. Commun. 2017, 486, 1097–1102. [Google Scholar] [CrossRef]
  29. Zhang, Z.; Chen, Z.; Song, Y.; Zhang, P.; Hu, J.; Bai, C. Expression of aquaporin 5 increases proliferation and metastasis potential of lung cancer. J. Pathol. 2010, 221, 210–220. [Google Scholar] [CrossRef]
  30. Yoshida, T.; Hojo, S.; Sekine, S.; Sawada, S.; Okumura, T.; Nagata, T.; Shimada, Y.; Tsukada, K. Expression of aquaporin-1 is a poor prognostic factor for stage II and III colon cancer. Mol. Clin. Oncol. 2013, 1, 953–958. [Google Scholar] [CrossRef] [PubMed]
  31. Guo, X.; Sun, T.; Yang, M.; Li, Z.; Li, Z.; Gao, Y. Prognostic value of combined aquaporin 3 and aquaporin 5 overexpression in hepatocellular carcinoma. Biomed. Res. Int. 2013, 2013, 206525. [Google Scholar] [CrossRef] [PubMed]
  32. Direito, I.; Paulino, J.; Vigia, E.; Brito, M.A.; Soveral, G. Differential expression of aquaporin-3 and aquaporin-5 in pancreatic ductal adenocarcinoma. J. Surg. Oncol. 2017, 115, 980–996. [Google Scholar] [CrossRef] [PubMed]
  33. Ding, T.; Ma, Y.; Li, W.; Liu, X.; Ying, G.; Fu, L.; Gu, F. Role of aquaporin-4 in the regulation of migration and invasion of human glioma cells. Int. J. Oncol. 2011, 38, 1521–1531. [Google Scholar] [CrossRef] [PubMed]
  34. Zhu, J.; Shi, L.; Su, Y. Aquaporin-4 as a New Potential Molecular Biomarker for Prognosis of Low-Grade Glioma: Comprehensive Analysis Based on Online Platforms. World Neurosurg. 2023, 175, e713–e722. [Google Scholar] [CrossRef] [PubMed]
  35. Wang, R.; Peng, L.; Xiao, Y.; Zhou, Q.; Wang, Z.; Tang, L.; Xiao, H.; Yang, K.; Liu, H.; Li, L. Single-cell RNA sequencing reveals changes in glioma-associated macrophage polarization and cellular states of malignant gliomas with high AQP4 expression. Cancer Gene Ther. 2023, 30, 716–726. [Google Scholar] [CrossRef]
  36. Jing, J.; Sun, J.; Wu, Y.; Zhang, N.; Liu, C.; Chen, S.; Li, W.; Hong, C.; Xu, B.; Chen, M. AQP9 Is a Prognostic Factor for Kidney Cancer and a Promising Indicator for M2 TAM Polarization and CD8+ T-Cell Recruitment. Front. Oncol. 2021, 11, 770565. [Google Scholar] [CrossRef]
  37. Gao, C.; Shen, J.; Yao, L.; Xia, Z.; Liang, X.; Zhu, R.; Chen, Z. Low expression of AQP9 and its value in hepatocellular carcinoma. Transl. Cancer Res. 2021, 10, 1826–1841. [Google Scholar] [CrossRef] [PubMed]
  38. Shi, Y.; Yasui, M.; Hara-Chikuma, M. AQP9 transports lactate in tumor-associated macrophages to stimulate an M2-like polarization that promotes colon cancer progression. Biochem. Biophys. Rep. 2022, 31, 101317. [Google Scholar] [CrossRef] [PubMed]
  39. Zou, L.B.; Zhang, R.J.; Tan, Y.J.; Ding, G.L.; Shi, S.; Zhang, D.; He, R.H.; Liu, A.X.; Wang, T.T.; Leung, P.C.; et al. Identification of estrogen response element in the aquaporin-2 gene that mediates estrogen-induced cell migration and invasion in human endometrial carcinoma. J. Clin. Endocrinol. Metab. 2011, 96, E1399–E1408. [Google Scholar] [CrossRef] [PubMed]
  40. Pellavio, G.; Martinotti, S.; Patrone, M.; Ranzato, E.; Laforenza, U. Aquaporin-6 May Increase the Resistance to Oxidative Stress of Malignant Pleural Mesothelioma Cells. Cells 2022, 11, 1892. [Google Scholar] [CrossRef]
  41. Dai, C.; Charlestin, V.; Wang, M.; Walker, Z.T.; Miranda-Vergara, M.C.; Facchine, B.A.; Wu, J.; Kaliney, W.J.; Dovichi, N.J.; Li, J.; et al. Aquaporin-7 Regulates the Response to Cellular Stress in Breast Cancer. Cancer Res. 2020, 80, 4071–4086. [Google Scholar] [CrossRef]
  42. Wu, Q.; Yang, Z.F.; Wang, K.J.; Feng, X.Y.; Lv, Z.J.; Li, Y.; Jian, Z.X. AQP8 inhibits colorectal cancer growth and metastasis by down-regulating PI3K/AKT signaling and PCDH7 expression. Am. J. Cancer Res. 2018, 8, 266–279. [Google Scholar]
  43. Hao, Z.; Huajun, S.; Zhen, G.; Yu, X.; Qian, L.; Ziling, C.; Zihao, S.; Qingqian, X.; Shujuan, Z. AQP8 promotes glioma proliferation and growth, possibly through the ROS/PTEN/AKT signaling pathway. BMC Cancer 2023, 23, 516. [Google Scholar] [CrossRef] [PubMed]
  44. Li, W.; Song, Y.; Pan, C.; Yu, J.; Zhang, J.; Zhu, X. Aquaporin-8 is a novel marker for progression of human cervical cancer cells. Cancer Biomark. 2021, 32, 391–400. [Google Scholar] [CrossRef]
  45. Saadoun, S.; Papadopoulos, M.C.; Hara-Chikuma, M.; Verkman, A.S. Impairment of angiogenesis and cell migration by targeted aquaporin-1 gene disruption. Nature 2005, 434, 786–792. [Google Scholar] [CrossRef] [PubMed]
  46. Papadopoulos, M.C.; Saadoun, S.; Verkman, A.S. Aquaporins and cell migration. Pflug. Arch. 2008, 456, 693–700. [Google Scholar] [CrossRef]
  47. Nicchia, G.P.; Stigliano, C.; Sparaneo, A.; Rossi, A.; Frigeri, A.; Svelto, M. Inhibition of aquaporin-1 dependent angiogenesis impairs tumour growth in a mouse model of melanoma. J. Mol. Med. 2012, 91, 613–623. [Google Scholar] [CrossRef]
  48. Esteva-Font, C.; Jin, B.J.; Verkman, A.S. Aquaporin-1 gene deletion reduces breast tumor growth and lung metastasis in tumor-producing MMTV-PyVT mice. FASEB J. 2013, 28, 1446–1453. [Google Scholar] [CrossRef] [PubMed]
  49. Ji, Y.; Liao, X.; Jiang, Y.; Wei, W.; Yang, H. Aquaporin 1 knockdown inhibits triple-negative breast cancer cell proliferation and invasion in vitro and in vivo. Oncol. Lett. 2021, 21, 437. [Google Scholar] [CrossRef]
  50. Jiang, Y. Aquaporin-1 activity of plasma membrane affects HT20 colon cancer cell migration. IUBMB Life 2009, 61, 1001–1009. [Google Scholar] [CrossRef] [PubMed]
  51. Pan, H.; Sun, C.C.; Zhou, C.Y.; Huang, H.F. Expression of aquaporin-1 in normal, hyperplasic, and carcinomatous endometria. Int. J. Gynaecol. Obs. 2008, 101, 239–244. [Google Scholar] [CrossRef] [PubMed]
  52. Moon, C.S.; Moon, D.; Kang, S.K. Aquaporins in Cancer Biology. Front. Oncol. 2022, 12, 782829. [Google Scholar] [CrossRef]
  53. Kaneko, K.; Yagui, K.; Tanaka, A.; Yoshihara, K.; Ishikawa, K.; Takahashi, K.; Bujo, H.; Sakurai, K.; Saito, Y. Aquaporin 1 is required for hypoxia-inducible angiogenesis in human retinal vascular endothelial cells. Microvasc. Res. 2008, 75, 297–301. [Google Scholar] [CrossRef] [PubMed]
  54. Tie, L.; Lu, N.; Pan, X.Y.; Pan, Y.; An, Y.; Gao, J.W.; Lin, Y.H.; Yu, H.M.; Li, X.J. Hypoxia-induced up-regulation of aquaporin-1 protein in prostate cancer cells in a p38-dependent manner. Cell Physiol. Biochem. 2012, 29, 269–280. [Google Scholar] [CrossRef]
  55. Yun, S.; Sun, P.L.; Jin, Y.; Kim, H.; Park, E.; Park, S.Y.; Lee, K.; Lee, K.; Chung, J.H. Aquaporin 1 Is an Independent Marker of Poor Prognosis in Lung Adenocarcinoma. J. Pathol. Transl. Med. 2016, 50, 251–257. [Google Scholar] [CrossRef]
  56. Hara-Chikuma, M.; Verkman, A.S. Prevention of skin tumorigenesis and impairment of epidermal cell proliferation by targeted aquaporin-3 gene disruption. Mol. Cell Biol. 2008, 28, 326–332. [Google Scholar] [CrossRef]
  57. da Silva, I.V.; Pimpao, C.; Paccetti-Alves, I.; Thomas, S.R.; Barateiro, A.; Casini, A.; Soveral, G. Blockage of aquaporin-3 peroxiporin activity by organogold compounds affects melanoma cell adhesion, proliferation and migration. J. Physiol. 2024, 602, 3111–3129. [Google Scholar] [CrossRef] [PubMed]
  58. Liu, Y.L.; Matsuzaki, T.; Nakazawa, T.; Murata, S.; Nakamura, N.; Kondo, T.; Iwashina, M.; Mochizuki, K.; Yamane, T.; Takata, K.; et al. Expression of aquaporin 3 (AQP3) in normal and neoplastic lung tissues. Hum. Pathol. 2007, 38, 171–178. [Google Scholar] [CrossRef] [PubMed]
  59. Zhang, M.; Li, T.; Zhu, J.; Tuo, B.; Liu, X. Physiological and pathophysiological role of ion channels and transporters in the colorectum and colorectal cancer. J. Cell Mol. Med. 2020, 24, 9486–9494. [Google Scholar] [CrossRef] [PubMed]
  60. Chen, G.; Shi, Y.; Liu, M.; Sun, J. circHIPK3 regulates cell proliferation and migration by sponging miR-124 and regulating AQP3 expression in hepatocellular carcinoma. Cell Death Dis. 2018, 9, 175. [Google Scholar] [CrossRef] [PubMed]
  61. De Ieso, M.L.; Yool, A.J. Mechanisms of Aquaporin-Facilitated Cancer Invasion and Metastasis. Front. Chem. 2018, 6, 135. [Google Scholar] [CrossRef]
  62. Li, A.; Lu, D.; Zhang, Y.; Li, J.; Fang, Y.; Li, F.; Sun, J. Critical role of aquaporin-3 in epidermal growth factor-induced migration of colorectal carcinoma cells and its clinical significance. Oncol. Rep. 2013, 29, 535–540. [Google Scholar] [CrossRef] [PubMed]
  63. Chen, J.; Wang, T.; Zhou, Y.C.; Gao, F.; Zhang, Z.H.; Xu, H.; Wang, S.L.; Shen, L.Z. Aquaporin 3 promotes epithelial-mesenchymal transition in gastric cancer. J. Exp. Clin. Cancer Res. 2014, 33, 38. [Google Scholar] [CrossRef]
  64. Zhou, Y.; Wang, Y.; Wen, J.; Zhao, H.; Dong, X.; Zhang, Z.; Wang, S.; Shen, L. Aquaporin 3 promotes the stem-like properties of gastric cancer cells via Wnt/GSK-3beta/beta-catenin pathway. Oncotarget 2016, 7, 16529–16541. [Google Scholar] [CrossRef] [PubMed]
  65. Chen, J.; Wang, Z.; Xu, D.; Liu, Y.; Gao, Y. Aquaporin 3 promotes prostate cancer cell motility and invasion via extracellular signal-regulated kinase 1/2-mediated matrix metalloproteinase-3 secretion. Mol. Med. Rep. 2015, 11, 2882–2888. [Google Scholar] [CrossRef] [PubMed]
  66. Kusayama, M.; Wada, K.; Nagata, M.; Ishimoto, S.; Takahashi, H.; Yoneda, M.; Nakajima, A.; Okura, M.; Kogo, M.; Kamisaki, Y. Critical role of aquaporin 3 on growth of human esophageal and oral squamous cell carcinoma. Cancer Sci. 2011, 102, 1128–1136. [Google Scholar] [CrossRef]
  67. Direito, I.; Madeira, A.; Brito, M.A.; Soveral, G. Aquaporin-5: From structure to function and dysfunction in cancer. Cell Mol. Life Sci. 2016, 73, 1623–1640. [Google Scholar] [CrossRef] [PubMed]
  68. Kang, S.K.; Chae, Y.K.; Woo, J.; Kim, M.S.; Park, J.C.; Lee, J.; Soria, J.C.; Jang, S.J.; Sidransky, D.; Moon, C. Role of human aquaporin 5 in colorectal carcinogenesis. Am. J. Pathol. 2008, 173, 518–525. [Google Scholar] [CrossRef] [PubMed]
  69. Woo, J.; Lee, J.; Kim, M.S.; Jang, S.J.; Sidransky, D.; Moon, C. The effect of aquaporin 5 overexpression on the Ras signaling pathway. Biochem. Biophys. Res. Commun. 2008, 367, 291–298. [Google Scholar] [CrossRef]
  70. Chen, C.; Ma, T.; Zhang, C.; Zhang, H.; Bai, L.; Kong, L.; Luo, J. Down-regulation of aquaporin 5-mediated epithelial-mesenchymal transition and anti-metastatic effect by natural product Cairicoside E in colorectal cancer. Mol. Carcinog. 2017, 56, 2692–2705. [Google Scholar] [CrossRef]
  71. Wang, W.; Li, Q.; Yang, T.; Li, D.; Ding, F.; Sun, H.; Bai, G. Anti-cancer effect of Aquaporin 5 silencing in colorectal cancer cells in association with inhibition of Wnt/beta-catenin pathway. Cytotechnology 2018, 70, 615–624. [Google Scholar] [CrossRef] [PubMed]
  72. He, Z.; Dong, W.; Hu, J.; Ren, X. AQP5 promotes hepatocellular carcinoma metastasis via NF-kappaB-regulated epithelial-mesenchymal transition. Biochem. Biophys. Res. Commun. 2017, 490, 343–348. [Google Scholar] [CrossRef] [PubMed]
  73. Yi, Y.; Qiu, G.; Liu, H.; Gao, F.; Liu, X.; Chen, Y.; Yang, M. Hypotonic induction of aquaporin5 expression in rat astrocytes through p38 MAPK pathway. Anat. Histol. Embryol. 2022, 51, 769–780. [Google Scholar] [CrossRef]
  74. Moon, C.; Soria, J.C.; Jang, S.J.; Lee, J.; Obaidul Hoque, M.; Sibony, M.; Trink, B.; Chang, Y.S.; Sidransky, D.; Mao, L. Involvement of aquaporins in colorectal carcinogenesis. Oncogene 2003, 22, 6699–6703. [Google Scholar] [CrossRef]
  75. Zhu, Z.; Jiao, L.; Li, T.; Wang, H.; Wei, W.; Qian, H. Expression of AQP3 and AQP5 as a prognostic marker in triple-negative breast cancer. Oncol. Lett. 2018, 16, 2661–2667. [Google Scholar] [CrossRef]
  76. Edamana, S.; Login, F.H.; Riishede, A.; Dam, V.S.; Tramm, T.; Nejsum, L.N. The cell polarity protein Scribble is downregulated by the water channel aquaporin-5 in breast cancer cells. Am. J. Physiol. Cell Physiol. 2023, 324, C307–C319. [Google Scholar] [CrossRef] [PubMed]
  77. Edamana, S.; Login, F.H.; Riishede, A.; Dam, V.S.; Kirkegaard, T.; Nejsum, L.N. The water channels aquaporin-1 and aquaporin-3 interact with and affect the cell polarity protein Scribble in 3D in vitro models of breast cancer. Am. J. Physiol. Cell Physiol. 2024, 327, C1323–C1334. [Google Scholar] [CrossRef] [PubMed]
  78. Rodrigues, C.; Pimpao, C.; Mosca, A.F.; Coxixo, A.S.; Lopes, D.; da Silva, I.V.; Pedersen, P.A.; Antunes, F.; Soveral, G. Human Aquaporin-5 Facilitates Hydrogen Peroxide Permeation Affecting Adaption to Oxidative Stress and Cancer Cell Migration. Cancers 2019, 11, 932. [Google Scholar] [CrossRef]
  79. Silva, P.M.; da Silva, I.V.; Sarmento, M.J.; Silva, I.C.; Carvalho, F.A.; Soveral, G.; Santos, N.C. Aquaporin-3 and Aquaporin-5 Facilitate Migration and Cell-Cell Adhesion in Pancreatic Cancer by Modulating Cell Biomechanical Properties. Cells 2022, 11, 1308. [Google Scholar] [CrossRef]
  80. Marinho, H.S.; Real, C.; Cyrne, L.; Soares, H.; Antunes, F. Hydrogen peroxide sensing, signaling and regulation of transcription factors. Redox Biol. 2014, 2, 535–562. [Google Scholar] [CrossRef] [PubMed]
  81. Lennicke, C.; Rahn, J.; Lichtenfels, R.; Wessjohann, L.A.; Seliger, B. Hydrogen peroxide—Production, fate and role in redox signaling of tumor cells. Cell Commun. Signal 2015, 13, 39. [Google Scholar] [CrossRef]
  82. Acharya, A.; Das, I.; Chandhok, D.; Saha, T. Redox regulation in cancer: A double-edged sword with therapeutic potential. Oxid. Med. Cell Longev. 2010, 3, 23–34. [Google Scholar] [CrossRef] [PubMed]
  83. Di Marzo, N.; Chisci, E.; Giovannoni, R. The Role of Hydrogen Peroxide in Redox-Dependent Signaling: Homeostatic and Pathological Responses in Mammalian Cells. Cells 2018, 7, 156. [Google Scholar] [CrossRef]
  84. Sies, H.; Jones, D.P. Reactive oxygen species (ROS) as pleiotropic physiological signalling agents. Nat. Rev. Mol. Cell Biol. 2020, 21, 363–383. [Google Scholar] [CrossRef] [PubMed]
  85. Milković, L.; Čipak Gašparović, A. AQP3 and AQP5—Potential Regulators of Redox Status in Breast Cancer. Molecules 2021, 26, 2613. [Google Scholar] [CrossRef] [PubMed]
  86. Hara-Chikuma, M.; Watanabe, S.; Satooka, H. Involvement of aquaporin-3 in epidermal growth factor receptor signaling via hydrogen peroxide transport in cancer cells. Biochem. Biophys. Res. Commun. 2016, 471, 603–609. [Google Scholar] [CrossRef] [PubMed]
  87. Wang, Q.; Lin, B.; Wei, H.; Wang, X.; Nie, X.; Shi, Y. AQP3 Promotes the Invasion and Metastasis in Cervical Cancer by Regulating NOX4-derived H2O2 Activation of Syk/PI3K/Akt Signaling Axis. J. Cancer 2024, 15, 1124–1137. [Google Scholar] [CrossRef]
  88. Hara-Chikuma, M.; Tanaka, M.; Verkman, A.S.; Yasui, M. Inhibition of aquaporin-3 in macrophages by a monoclonal antibody as potential therapy for liver injury. Nat. Commun. 2020, 11, 5666. [Google Scholar] [CrossRef]
  89. da Silva, I.V.; Cardoso, C.; Martínez-Banaclocha, H.; Casini, A.; Pelegrín, P.; Soveral, G. Aquaporin-3 is involved in NLRP3-inflammasome activation contributing to the setting of inflammatory response. Cell. Mol. Life Sci. 2020, 78, 3073–3085. [Google Scholar] [CrossRef]
  90. Chae, Y.K.; Woo, J.; Kim, M.J.; Kang, S.K.; Kim, M.S.; Lee, J.; Lee, S.K.; Gong, G.; Kim, Y.H.; Soria, J.C.; et al. Expression of aquaporin 5 (AQP5) promotes tumor invasion in human non small cell lung cancer. PLoS ONE 2008, 3, e2162. [Google Scholar] [CrossRef]
  91. Cipak Gasparovic, A.; Milkovic, L.; Rodrigues, C.; Mlinaric, M.; Soveral, G. Peroxiporins Are Induced upon Oxidative Stress Insult and Are Associated with Oxidative Stress Resistance in Colon Cancer Cell Lines. Antioxidants 2021, 10, 1856. [Google Scholar] [CrossRef]
  92. Rodrigues, C.; Milkovic, L.; Bujak, I.T.; Tomljanovic, M.; Soveral, G.; Cipak Gasparovic, A. Lipid Profile and Aquaporin Expression under Oxidative Stress in Breast Cancer Cells of Different Malignancies. Oxid. Med. Cell Longev. 2019, 2019, 2061830. [Google Scholar] [CrossRef]
  93. Brown, D. Aquaporin Function: Seek and You Shall Find! Function 2021, 2, zqaa041. [Google Scholar] [CrossRef] [PubMed]
  94. McLennan, R.; McKinney, M.C.; Teddy, J.M.; Morrison, J.A.; Kasemeier-Kulesa, J.C.; Ridenour, D.A.; Manthe, C.A.; Giniunaite, R.; Robinson, M.; Baker, R.E.; et al. Neural crest cells bulldoze through the microenvironment using Aquaporin 1 to stabilize filopodia. Development 2020, 147, dev185231. [Google Scholar] [CrossRef]
  95. Meng, F.; Rui, Y.; Xu, L.; Wan, C.; Jiang, X.; Li, G. Aqp1 enhances migration of bone marrow mesenchymal stem cells through regulation of FAK and beta-catenin. Stem Cells Dev. 2014, 23, 66–75. [Google Scholar] [CrossRef] [PubMed]
  96. Monzani, E.; Bazzotti, R.; Perego, C.; La Porta, C.A. AQP1 is not only a water channel: It contributes to cell migration through Lin7/beta-catenin. PLoS ONE 2009, 4, e6167. [Google Scholar] [CrossRef] [PubMed]
  97. Huang, Y.T.; Zhou, J.; Shi, S.; Xu, H.Y.; Qu, F.; Zhang, D.; Chen, Y.D.; Yang, J.; Huang, H.F.; Sheng, J.Z. Identification of Estrogen Response Element in Aquaporin-3 Gene that Mediates Estrogen-induced Cell Migration and Invasion in Estrogen Receptor-positive Breast Cancer. Sci. Rep. 2015, 5, 12484. [Google Scholar] [CrossRef]
  98. Cui, D.; Sui, L.; Han, X.; Zhang, M.; Guo, Z.; Chen, W.; Yu, X.; Sun, Q.; Dong, M.; Ma, T.; et al. Aquaporin-3 mediates ovarian steroid hormone-induced motility of endometrial epithelial cells. Hum. Reprod. 2018, 33, 2060–2073. [Google Scholar] [CrossRef] [PubMed]
  99. Chivasso, C.; Hagstromer, C.J.; Rose, K.L.; Lhotellerie, F.; Leblanc, L.; Wang, Z.; Moscato, S.; Chevalier, C.; Zindy, E.; Martin, M.; et al. Ezrin Is a Novel Protein Partner of Aquaporin-5 in Human Salivary Glands and Shows Altered Expression and Cellular Localization in Sjogren’s Syndrome. Int. J. Mol. Sci. 2021, 22, 9213. [Google Scholar] [CrossRef] [PubMed]
  100. Tada, J.; Sawa, T.; Yamanaka, N.; Shono, M.; Akamatsu, T.; Tsumura, K.; Parvin, M.N.; Kanamori, N.; Hosoi, K. Involvement of vesicle-cytoskeleton interaction in AQP5 trafficking in AQP5-gene-transfected HSG cells. Biochem. Biophys. Res. Commun. 1999, 266, 443–447. [Google Scholar] [CrossRef] [PubMed]
  101. Muroi, S.I.; Isohama, Y. Ezrin Regulates Ca2+ Ionophore-Induced Plasma Membrane Translocation of Aquaporin-5. Int. J. Mol. Sci. 2021, 22, 13505. [Google Scholar] [CrossRef]
  102. Tietz, P.S.; McNiven, M.A.; Splinter, P.L.; Huang, B.Q.; Larusso, N.F. Cytoskeletal and motor proteins facilitate trafficking of AQP1-containing vesicles in cholangiocytes. Biol. Cell 2006, 98, 43–52. [Google Scholar] [CrossRef] [PubMed]
  103. Sidhaye, V.K.; Chau, E.; Srivastava, V.; Sirimalle, S.; Balabhadrapatruni, C.; Aggarwal, N.R.; D’Alessio, F.R.; Robinson, D.N.; King, L.S. A novel role for aquaporin-5 in enhancing microtubule organization and stability. PLoS ONE 2012, 7, e38717. [Google Scholar] [CrossRef]
  104. Huysseune, S.; Kienlen-Campard, P.; Hebert, S.; Tasiaux, B.; Leroy, K.; Devuyst, O.; Brion, J.P.; De Strooper, B.; Octave, J.N. Epigenetic control of aquaporin 1 expression by the amyloid precursor protein. FASEB J. 2009, 23, 4158–4167. [Google Scholar] [CrossRef] [PubMed]
  105. Zhang, H.; Li, H.; Liu, E.; Guang, Y.; Yang, L.; Mao, J.; Zhu, L.; Chen, L.; Wang, L. The AQP-3 water channel and the ClC-3 chloride channel coordinate the hypotonicity-induced swelling volume in nasopharyngeal carcinoma cells. Int. J. Biochem. Cell Biol. 2014, 57, 96–107. [Google Scholar] [CrossRef] [PubMed]
  106. Zhang, H.; Deng, Z.; Yang, L.; Luo, H.; Liu, S.; Li, Y.; Wei, Y.; Peng, S.; Zhu, L.; Wang, L.; et al. The AQP-3 water channel is a pivotal modulator of glycerol-induced chloride channel activation in nasopharyngeal carcinoma cells. Int. J. Biochem. Cell Biol. 2016, 72, 89–99. [Google Scholar] [CrossRef] [PubMed]
  107. Zheng, X.; Bollinger Bollag, W. Aquaporin 3 colocates with phospholipase d2 in caveolin-rich membrane microdomains and is downregulated upon keratinocyte differentiation. J. Investig. Dermatol. 2003, 121, 1487–1495. [Google Scholar] [CrossRef] [PubMed]
  108. Reppetti, J.; Reca, A.; Seyahian, E.A.; Medina, Y.; Martinez, N.; Szpilbarg, N.; Damiano, A.E. Intact caveolae are required for proper extravillous trophoblast migration and differentiation. J. Cell Physiol. 2020, 235, 3382–3392. [Google Scholar] [CrossRef] [PubMed]
  109. Huang, P.; Hansen, J.S.; Saba, K.H.; Bergman, A.; Negoita, F.; Gourdon, P.; Hagstrom-Andersson, A.; Lindkvist-Petersson, K. Aquaglyceroporins and orthodox aquaporins in human adipocytes. Biochim. Biophys. Acta Biomembr. 2022, 1864, 183795. [Google Scholar] [CrossRef] [PubMed]
  110. Hwang, S.; Kang, J.Y.; Kim, M.J.; Shin, D.M.; Hong, J.H. Carbonic anhydrase 12 mutation modulates membrane stability and volume regulation of aquaporin 5. J. Enzym. Inhib. Med. Chem. 2019, 34, 179–188. [Google Scholar] [CrossRef] [PubMed]
  111. Ohashi, Y.; Tsuzaka, K.; Takeuchi, T.; Sasaki, Y.; Tsubota, K. Altered distribution of aquaporin 5 and its C-terminal binding protein in the lacrimal glands of a mouse model for Sjogren’s syndrome. Curr. Eye Res. 2008, 33, 621–629. [Google Scholar] [CrossRef]
  112. Chivasso, C.; Nesverova, V.; Jarva, M.; Blanchard, A.; Rose, K.L.; Oberg, F.K.; Wang, Z.; Martin, M.; Lhotellerie, F.; Zindy, E.; et al. Unraveling Human AQP5-PIP Molecular Interaction and Effect on AQP5 Salivary Glands Localization in SS Patients. Cells 2021, 10, 2108. [Google Scholar] [CrossRef] [PubMed]
  113. Chin, W.-C.; Bhattacharya, D.; Yu, L.; Wang, M. Expression patterns of conjunctival mucin 5AC and aquaporin 5 in response to acute dry eye stress. PLoS ONE 2017, 12, e0187188. [Google Scholar] [CrossRef]
  114. Login, F.H.; Palmfeldt, J.; Cheah, J.S.; Yamada, S.; Nejsum, L.N. Aquaporin-5 regulation of cell-cell adhesion proteins: An elusive “tail” story. Am. J. Physiol. Cell Physiol. 2021, 320, C282–C292. [Google Scholar] [CrossRef] [PubMed]
  115. Tradtrantip, L.; Jin, B.J.; Yao, X.; Anderson, M.O.; Verkman, A.S. Aquaporin-Targeted Therapeutics: State-of-the-Field. Adv. Exp. Med. Biol. 2017, 969, 239–250. [Google Scholar] [CrossRef] [PubMed]
  116. Zhang, R.; van Hoek, A.N.; Biwersi, J.; Verkman, A.S. A point mutation at cysteine 189 blocks the water permeability of rat kidney water channel CHIP28k. Biochemistry 1993, 32, 2938–2941. [Google Scholar] [CrossRef] [PubMed]
  117. Kuwahara, M.; Gu, Y.; Ishibashi, K.; Marumo, F.; Sasaki, S. Mercury-sensitive residues and pore site in AQP3 water channel. Biochemistry 1997, 36, 13973–13978. [Google Scholar] [CrossRef]
  118. Muller-Lucks, A.; Gena, P.; Frascaria, D.; Altamura, N.; Svelto, M.; Beitz, E.; Calamita, G. Preparative scale production and functional reconstitution of a human aquaglyceroporin (AQP3) using a cell free expression system. N. Biotechnol. 2013, 30, 545–551. [Google Scholar] [CrossRef] [PubMed]
  119. Ismail, M.; Bokaee, S.; Morgan, R.; Davies, J.; Harrington, K.J.; Pandha, H. Inhibition of the aquaporin 3 water channel increases the sensitivity of prostate cancer cells to cryotherapy. Br. J. Cancer 2009, 100, 1889–1895. [Google Scholar] [CrossRef] [PubMed]
  120. Castle, N.A. Aquaporins as targets for drug discovery. Drug Discov. Today 2005, 10, 485–493. [Google Scholar] [CrossRef]
  121. Rubenwolf, P.C.; Georgopoulos, N.T.; Kirkwood, L.A.; Baker, S.C.; Southgate, J. Aquaporin expression contributes to human transurothelial permeability in vitro and is modulated by NaCl. PLoS ONE 2012, 7, e45339. [Google Scholar] [CrossRef]
  122. Niemietz, C.M.; Tyerman, S.D. New potent inhibitors of aquaporins: Silver and gold compounds inhibit aquaporins of plant and human origin. FEBS Lett. 2002, 531, 443–447. [Google Scholar] [CrossRef]
  123. Martins, A.P.; Marrone, A.; Ciancetta, A.; Galan Cobo, A.; Echevarria, M.; Moura, T.F.; Re, N.; Casini, A.; Soveral, G. Targeting aquaporin function: Potent inhibition of aquaglyceroporin-3 by a gold-based compound. PLoS ONE 2012, 7, e37435. [Google Scholar] [CrossRef]
  124. Serna, A.; Galan-Cobo, A.; Rodrigues, C.; Sanchez-Gomar, I.; Toledo-Aral, J.J.; Moura, T.F.; Casini, A.; Soveral, G.; Echevarria, M. Functional inhibition of aquaporin-3 with a gold-based compound induces blockage of cell proliferation. J. Cell Physiol. 2014, 229, 1787–1801. [Google Scholar] [CrossRef]
  125. Martins, A.P.; Ciancetta, A.; de Almeida, A.; Marrone, A.; Re, N.; Soveral, G.; Casini, A. Aquaporin inhibition by gold(III) compounds: New insights. ChemMedChem 2013, 8, 1086–1092. [Google Scholar] [CrossRef] [PubMed]
  126. Madeira, A.; Camps, M.; Zorzano, A.; Moura, T.F.; Soveral, G. Biophysical assessment of human aquaporin-7 as a water and glycerol channel in 3T3-L1 adipocytes. PLoS ONE 2013, 8, e83442. [Google Scholar] [CrossRef] [PubMed]
  127. Madeira, A.; de Almeida, A.; de Graaf, C.; Camps, M.; Zorzano, A.; Moura, T.F.; Casini, A.; Soveral, G. A gold coordination compound as a chemical probe to unravel aquaporin-7 function. Chembiochem 2014, 15, 1487–1494. [Google Scholar] [CrossRef] [PubMed]
  128. Peng, R.; Zhao, G.X.; Li, J.; Zhang, Y.; Shen, X.Z.; Wang, J.Y.; Sun, J.Y. Auphen and dibutyryl cAMP suppress growth of hepatocellular carcinoma by regulating expression of aquaporins 3 and 9 in vivo. World J. Gastroenterol. 2016, 22, 3341–3354. [Google Scholar] [CrossRef]
  129. Pimpao, C.; Wragg, D.; Bonsignore, R.; Aikman, B.; Pedersen, P.A.; Leoni, S.; Soveral, G.; Casini, A. Mechanisms of irreversible aquaporin-10 inhibition by organogold compounds studied by combined biophysical methods and atomistic simulations. Metallomics 2021, 13, mfab053. [Google Scholar] [CrossRef] [PubMed]
  130. Babak, M.V.; Chong, K.R.; Rapta, P.; Zannikou, M.; Tang, H.M.; Reichert, L.; Chang, M.R.; Kushnarev, V.; Heffeter, P.; Meier-Menches, S.M.; et al. Interfering with Metabolic Profile of Triple-Negative Breast Cancers Using Rationally Designed Metformin Prodrugs. Angew. Chem. Int. Ed. 2021, 60, 13405–13413. [Google Scholar] [CrossRef] [PubMed]
  131. Pinho, J.O.; Coelho, M.; Pimpão, C.; Konwar, J.; Godinho-Santos, A.; Noiva, R.M.; Thomas, S.R.; Casini, A.; Soveral, G.; Gaspar, M.M. Liposomal Formulation of an Organogold Complex Enhancing Its Activity as Antimelanoma Agent—In Vitro and In Vivo Studies. Pharmaceutics 2024, 16, 1566. [Google Scholar] [CrossRef] [PubMed]
  132. Nave, M.; Castro, R.E.; Rodrigues, C.M.; Casini, A.; Soveral, G.; Gaspar, M.M. Nanoformulations of a potent copper-based aquaporin inhibitor with cytotoxic effect against cancer cells. Nanomedicine 2016, 11, 1817–1830. [Google Scholar] [CrossRef] [PubMed]
  133. Pinho, J.O.; Amaral, J.D.; Castro, R.E.; Rodrigues, C.M.; Casini, A.; Soveral, G.; Gaspar, M.M. Copper complex nanoformulations featuring highly promising therapeutic potential in murine melanoma models. Nanomedicine 2019, 14, 835–850. [Google Scholar] [CrossRef]
  134. Pinho, J.O.; da Silva, I.V.; Amaral, J.D.; Rodrigues, C.M.P.; Casini, A.; Soveral, G.; Gaspar, M.M. Therapeutic potential of a copper complex loaded in pH-sensitive long circulating liposomes for colon cancer management. Int. J. Pharm. 2021, 599, 120463. [Google Scholar] [CrossRef]
  135. Pimpao, C.; da Silva, I.V.; Mosca, A.F.; Pinho, J.O.; Gaspar, M.M.; Gumerova, N.I.; Rompel, A.; Aureliano, M.; Soveral, G. The Aquaporin-3-Inhibiting Potential of Polyoxotungstates. Int. J. Mol. Sci. 2020, 21, 2467. [Google Scholar] [CrossRef]
  136. Detmers, F.J.; de Groot, B.L.; Muller, E.M.; Hinton, A.; Konings, I.B.; Sze, M.; Flitsch, S.L.; Grubmuller, H.; Deen, P.M. Quaternary ammonium compounds as water channel blockers. Specificity, potency, and site of action. J. Biol. Chem. 2006, 281, 14207–14214. [Google Scholar] [CrossRef]
  137. Seeliger, D.; Zapater, C.; Krenc, D.; Haddoub, R.; Flitsch, S.; Beitz, E.; Cerda, J.; de Groot, B.L. Discovery of novel human aquaporin-1 blockers. ACS Chem. Biol. 2013, 8, 249–256. [Google Scholar] [CrossRef]
  138. Migliati, E.; Meurice, N.; DuBois, P.; Fang, J.S.; Somasekharan, S.; Beckett, E.; Flynn, G.; Yool, A.J. Inhibition of aquaporin-1 and aquaporin-4 water permeability by a derivative of the loop diuretic bumetanide acting at an internal pore-occluding binding site. Mol. Pharmacol. 2009, 76, 105–112. [Google Scholar] [CrossRef] [PubMed]
  139. Kourghi, M.; Pei, J.V.; De Ieso, M.L.; Flynn, G.; Yool, A.J. Bumetanide Derivatives AqB007 and AqB011 Selectively Block the Aquaporin-1 Ion Channel Conductance and Slow Cancer Cell Migration. Mol. Pharmacol. 2016, 89, 133–140. [Google Scholar] [CrossRef] [PubMed]
  140. Yool, A.J.; Morelle, J.; Cnops, Y.; Verbavatz, J.M.; Campbell, E.M.; Beckett, E.A.; Booker, G.W.; Flynn, G.; Devuyst, O. AqF026 is a pharmacologic agonist of the water channel aquaporin-1. J. Am. Soc. Nephrol. 2013, 24, 1045–1052. [Google Scholar] [CrossRef] [PubMed]
  141. Yang, B.; Kim, J.K.; Verkman, A.S. Comparative efficacy of HgCl2 with candidate aquaporin-1 inhibitors DMSO, gold, TEA+ and acetazolamide. FEBS Lett. 2006, 580, 6679–6684. [Google Scholar] [CrossRef] [PubMed]
  142. Esteva-Font, C.; Jin, B.J.; Lee, S.; Phuan, P.W.; Anderson, M.O.; Verkman, A.S. Experimental Evaluation of Proposed Small-Molecule Inhibitors of Water Channel Aquaporin-1. Mol. Pharmacol. 2016, 89, 686–693. [Google Scholar] [CrossRef] [PubMed]
  143. Jiang, Y.J.; Kim, P.; Lu, Y.F.; Feingold, K.R. PPARgamma activators stimulate aquaporin 3 expression in keratinocytes/epidermis. Exp. Dermatol. 2011, 20, 595–599. [Google Scholar] [CrossRef] [PubMed]
  144. Choudhary, V.; Olala, L.O.; Kagha, K.; Pan, Z.Q.; Chen, X.; Yang, R.; Cline, A.; Helwa, I.; Marshall, L.; Kaddour-Djebbar, I.; et al. Regulation of the Glycerol Transporter, Aquaporin-3, by Histone Deacetylase-3 and p53 in Keratinocytes. J. Investig. Dermatol. 2017, 137, 1935–1944. [Google Scholar] [CrossRef]
  145. Helwa, I.; Choudhary, V.; Chen, X.; Kaddour-Djebbar, I.; Bollag, W.B. Anti-Psoriatic Drug Monomethylfumarate Increases Nuclear Factor Erythroid 2-Related Factor 2 Levels and Induces Aquaporin-3 mRNA and Protein Expression. J. Pharmacol. Exp. Ther. 2017, 362, 243–253. [Google Scholar] [CrossRef] [PubMed]
  146. Flodby, P.; Li, C.; Liu, Y.; Wang, H.; Rieger, M.E.; Minoo, P.; Crandall, E.D.; Ann, D.K.; Borok, Z.; Zhou, B. Cell-specific expression of aquaporin-5 (Aqp5) in alveolar epithelium is directed by GATA6/Sp1 via histone acetylation. Sci. Rep. 2017, 7, 3473. [Google Scholar] [CrossRef]
  147. Ikarashi, N.; Baba, K.; Ushiki, T.; Kon, R.; Mimura, A.; Toda, T.; Ishii, M.; Ochiai, W.; Sugiyama, K. The laxative effect of bisacodyl is attributable to decreased aquaporin-3 expression in the colon induced by increased PGE2 secretion from macrophages. Am. J. Physiol. Gastrointest. Liver Physiol. 2011, 301, G887–G895. [Google Scholar] [CrossRef]
  148. Sonntag, Y.; Gena, P.; Maggio, A.; Singh, T.; Artner, I.; Oklinski, M.K.; Johanson, U.; Kjellbom, P.; Nieland, J.D.; Nielsen, S.; et al. Identification and characterization of potent and selective aquaporin-3 and aquaporin-7 inhibitors. J. Biol. Chem. 2019, 294, 7377–7387. [Google Scholar] [CrossRef] [PubMed]
  149. Tanaka, M.; Yasui, M.; Hara-Chikuma, M. Aquaporin 3 inhibition suppresses the mitochondrial respiration rate and viability of multiple myeloma cells. Biochem. Biophys. Res. Commun. 2023, 676, 158–164. [Google Scholar] [CrossRef]
  150. Villandre, J.; White, V.; Lear, T.B.; Chen, Y.; Tuncer, F.; Vaiz, E.; Tuncer, B.; Lockwood, K.; Camarco, D.; Liu, Y.; et al. A Repurposed Drug Screen for Compounds Regulating Aquaporin 5 Stability in Lung Epithelial Cells. Front. Pharmacol. 2022, 13, 828643. [Google Scholar] [CrossRef] [PubMed]
  151. Rump, K.; Koos, B.; Ziehe, D.; Thon, P.; Rahmel, T.; Palmowski, L.; Marko, B.; Wolf, A.; Witowski, A.; Bazzi, Z.; et al. Methazolamide Reduces the AQP5 mRNA Expression and Immune Cell Migration-A New Potential Drug in Sepsis Therapy? Int. J. Mol. Sci. 2024, 25, 610. [Google Scholar] [CrossRef]
  152. Montiel, V.; Bella, R.; Michel, L.Y.M.; Esfahani, H.; De Mulder, D.; Robinson, E.L.; Deglasse, J.-P.; Tiburcy, M.; Chow, P.H.; Jonas, J.-C.; et al. Inhibition of aquaporin-1 prevents myocardial remodeling by blocking the transmembrane transport of hydrogen peroxide. Sci. Transl. Med. 2020, 12, eaay2176. [Google Scholar] [CrossRef]
  153. Cao, C.; Wan, S.; Jiang, Q.; Amaral, A.; Lu, S.; Hu, G.; Bi, Z.; Kouttab, N.; Chu, W.; Wan, Y. All-trans retinoic acid attenuates ultraviolet radiation-induced down-regulation of aquaporin-3 and water permeability in human keratinocytes. J. Cell Physiol. 2008, 215, 506–516. [Google Scholar] [CrossRef] [PubMed]
  154. Wu, N.L.; Fang, J.Y.; Chen, M.; Wu, C.J.; Huang, C.C.; Hung, C.F. Chrysin protects epidermal keratinocytes from UVA- and UVB-induced damage. J. Agric. Food Chem. 2011, 59, 8391–8400. [Google Scholar] [CrossRef]
  155. Tang, S.C.; Tang, L.C.; Liu, C.H.; Liao, P.Y.; Lai, J.C.; Yang, J.H. Glycolic acid attenuates UVB-induced aquaporin-3, matrix metalloproteinase-9 expression, and collagen degradation in keratinocytes and mouse skin. Biochem. J. 2019, 476, 1387–1400. [Google Scholar] [CrossRef]
  156. Wu, Z.; Uchi, H.; Morino-Koga, S.; Shi, W.; Furue, M. Resveratrol inhibition of human keratinocyte proliferation via SIRT1/ARNT/ERK dependent downregulation of aquaporin 3. J. Dermatol. Sci. 2014, 75, 16–23. [Google Scholar] [CrossRef] [PubMed]
  157. Hung, C.F.; Hsiao, C.Y.; Hsieh, W.H.; Li, H.J.; Tsai, Y.J.; Lin, C.N.; Chang, H.H.; Wu, N.L. 18ss-glycyrrhetinic acid derivative promotes proliferation, migration and aquaporin-3 expression in human dermal fibroblasts. PLoS ONE 2017, 12, e0182981. [Google Scholar] [CrossRef]
  158. Ji, C.; Cao, C.; Lu, S.; Kivlin, R.; Amaral, A.; Kouttab, N.; Yang, H.; Chu, W.; Bi, Z.; Di, W.; et al. Curcumin attenuates EGF-induced AQP3 up-regulation and cell migration in human ovarian cancer cells. Cancer Chemother. Pharmacol. 2008, 62, 857–865. [Google Scholar] [CrossRef]
  159. Kon, R.; Yamamura, M.; Matsunaga, Y.; Kimura, H.; Minami, M.; Kato, S.; Ikarashi, N.; Sugiyama, K. Laxative effect of repeated Daiokanzoto is attributable to decrease in aquaporin-3 expression in the colon. J. Nat. Med. 2018, 72, 493–502. [Google Scholar] [CrossRef] [PubMed]
  160. Zhu, D.; Chen, C.; Bai, L.; Kong, L.; Luo, J. Downregulation of Aquaporin 3 Mediated the Laxative Effect in the Rat Colon by a Purified Resin Glycoside Fraction from Pharbitis Semen. Evid. Based Complement. Altern. Med. 2019, 2019, 9406342. [Google Scholar] [CrossRef] [PubMed]
  161. Yin, J.; Liang, Y.; Wang, D.; Yan, Z.; Yin, H.; Wu, D.; Su, Q. Naringenin induces laxative effects by upregulating the expression levels of c-Kit and SCF, as well as those of aquaporin 3 in mice with loperamide-induced constipation. Int. J. Mol. Med. 2018, 41, 649–658. [Google Scholar] [CrossRef] [PubMed]
  162. Wu, J.; Gan, Y.; Luo, H.; Xu, N.; Chen, L.; Li, M.; Guan, F.; Su, Z.; Lin, Z.; Xie, J.; et al. beta-Patchoulene Ameliorates Water Transport and the Mucus Barrier in 5-Fluorouracil-Induced Intestinal Mucositis Rats via the cAMP/PKA/CREB Signaling Pathway. Front. Pharmacol. 2021, 12, 689491. [Google Scholar] [CrossRef]
  163. Paccetti-Alves, I.; Batista, M.S.P.; Pimpao, C.; Victor, B.L.; Soveral, G. Unraveling the Aquaporin-3 Inhibitory Effect of Rottlerin by Experimental and Computational Approaches. Int. J. Mol. Sci. 2023, 24, 6004. [Google Scholar] [CrossRef] [PubMed]
  164. Skowronska, A.; Mlotkowska, P.; Nielsen, S.; Skowronski, M.T. Difference in expression between AQP1 and AQP5 in porcine endometrium and myometrium in response to steroid hormones, oxytocin, arachidonic acid, forskolin and cAMP during the mid-luteal phase of the estrous cycle and luteolysis. Reprod. Biol. Endocrinol. 2015, 13, 131. [Google Scholar] [CrossRef]
  165. Skowronska, A.; Mlotkowska, P.; Wojciechowicz, B.; Okrasa, S.; Nielsen, S.; Skowronski, M.T. Progesterone, estradiol, arachidonic acid, oxytocin, forskolin and cAMP influence on aquaporin 1 and 5 expression in porcine uterine explants during the mid-luteal phase of the estrous cycle and luteolysis: An in vitro study. Reprod. Biol. Endocrinol. 2015, 13, 7. [Google Scholar] [CrossRef]
  166. Tanski, D.; Skowronska, A.; Tanska, M.; Lepiarczyk, E.; Skowronski, M.T. The In Vitro Effect of Steroid Hormones, Arachidonic Acid, and Kinases Inhibitors on Aquaporin 1, 2, 5, and 7 Gene Expression in the Porcine Uterine Luminal Epithelial Cells during the Estrous Cycle. Cells 2021, 10, 832. [Google Scholar] [CrossRef] [PubMed]
  167. Yang, C.; Lim, W.; Bae, H.; Song, G. Aquaporin 3 is regulated by estrogen in the chicken oviduct and is involved in progression of epithelial cell-derived ovarian carcinomas. Domest. Anim. Endocrinol. 2016, 55, 97–106. [Google Scholar] [CrossRef] [PubMed]
  168. Kobayashi, M.; Takahashi, E.; Miyagawa, S.; Watanabe, H.; Iguchi, T. Chromatin immunoprecipitation-mediated target identification proved aquaporin 5 is regulated directly by estrogen in the uterus. Genes. Cells 2006, 11, 1133–1143. [Google Scholar] [CrossRef]
  169. Skowronski, M.; Mlotkowska, P.; Tanski, D.; Lepiarczyk, E.; Oklinski, M.; Nielsen, S.; Skowronska, A. Pituitary Gonadotropins, Prolactin and Growth Hormone Differentially Regulate AQP1 Expression in the Porcine Ovarian Follicular Cells. Int. J. Mol. Sci. 2017, 19, 5. [Google Scholar] [CrossRef] [PubMed]
  170. Hermo, L.; Krzeczunowicz, D.; Ruz, R. Cell Specificity of Aquaporins 0, 3, and 10 Expressed in the Testis, Efferent Ducts, and Epididymis of Adult Rats. J. Androl. 2013, 25, 494–505. [Google Scholar] [CrossRef] [PubMed]
  171. Salleh, N.; Mokhtar, H.M.; Kassim, N.M.; Giribabu, N. Testosterone Induces Increase in Aquaporin (AQP)-1, 5, and 7 Expressions in the Uteri of Ovariectomized Rats. J. Membr. Biol. 2015, 248, 1097–1105. [Google Scholar] [CrossRef] [PubMed]
  172. Rodríguez, A.; Gómez-Ambrosi, J.; Catalán, V.; Ezquerro, S.; Méndez-Giménez, L.; Becerril, S.; Ibáñez, P.; Vila, N.; Margall, M.A.; Moncada, R.; et al. Guanylin and uroguanylin stimulate lipolysis in human visceral adipocytes. Int. J. Obes. 2016, 40, 1405–1415. [Google Scholar] [CrossRef] [PubMed]
  173. Rodriguez, A.; Moreno, N.R.; Balaguer, I.; Mendez-Gimenez, L.; Becerril, S.; Catalan, V.; Gomez-Ambrosi, J.; Portincasa, P.; Calamita, G.; Soveral, G.; et al. Leptin administration restores the altered adipose and hepatic expression of aquaglyceroporins improving the non-alcoholic fatty liver of ob/ob mice. Sci. Rep. 2015, 5, 12067. [Google Scholar] [CrossRef] [PubMed]
  174. Gong, H.; Wang, W.; Kwon, T.H.; Jonassen, T.; Li, C.; Ring, T.; Froki, A.J.; Nielsen, S. EPO and alpha-MSH prevent ischemia/reperfusion-induced down-regulation of AQPs and sodium transporters in rat kidney. Kidney Int. 2004, 66, 683–695. [Google Scholar] [CrossRef]
  175. Ben, Y.; Chen, J.; Zhu, R.; Gao, L.; Bai, C. Upregulation of AQP3 and AQP5 induced by dexamethasone and ambroxol in A549 cells. Respir. Physiol. Neurobiol. 2008, 161, 111–118. [Google Scholar] [CrossRef] [PubMed]
  176. Chong, W.; Zhang, H.; Guo, Z.; Yang, L.; Shao, Y.; Liu, X.; Zhao, Y.; Wang, Z.; Zhang, M.; Guo, C.; et al. Aquaporin 1 promotes sensitivity of anthracycline chemotherapy in breast cancer by inhibiting beta-catenin degradation to enhance TopoIIalpha activity. Cell Death Differ. 2021, 28, 382–400. [Google Scholar] [CrossRef]
  177. Luo, L.; Yang, R.; Zhao, S.; Chen, Y.; Hong, S.; Wang, K.; Wang, T.; Cheng, J.; Zhang, T.; Chen, D. Decreased miR-320 expression is associated with breast cancer progression, cell migration, and invasiveness via targeting Aquaporin 1. Acta Biochim. Et. Biophys. Sin. 2018, 50, 473–480. [Google Scholar] [CrossRef] [PubMed]
  178. Park, E.J.; Jung, H.J.; Choi, H.J.; Jang, H.J.; Park, H.J.; Nejsum, L.N.; Kwon, T.H. Exosomes co-expressing AQP5-targeting miRNAs and IL-4 receptor-binding peptide inhibit the migration of human breast cancer cells. FASEB J. 2020, 34, 3379–3398. [Google Scholar] [CrossRef] [PubMed]
  179. Fang, Y.; Hu, J.; Wang, Z.; Zong, H.; Zhang, L.; Zhang, R.; Sun, L. LncRNA H19 functions as an Aquaporin 1 competitive endogenous RNA to regulate microRNA-874 expression in LPS sepsis. Biomed. Pharmacother. 2018, 105, 1183–1191. [Google Scholar] [CrossRef]
  180. Jiang, B.; Li, Z.; Zhang, W.; Wang, H.; Zhi, X.; Feng, J.; Chen, Z.; Zhu, Y.; Yang, L.; Xu, H.; et al. miR-874 inhibits cell proliferation, migration and invasion through targeting aquaporin-3 in gastric cancer. J. Gastroenterol. 2014, 49, 1011–1025. [Google Scholar] [CrossRef] [PubMed]
  181. Wang, S.; Wu, Y.; Yang, S.; Liu, X.; Lu, Y.; Liu, F.; Li, G.; Tian, G. miR-874 directly targets AQP3 to inhibit cell proliferation, mobility and EMT in non-small cell lung cancer. Thorac. Cancer 2020, 11, 1550–1558. [Google Scholar] [CrossRef] [PubMed]
  182. Zhi, X.; Tao, J.; Li, Z.; Jiang, B.; Feng, J.; Yang, L.; Xu, H.; Xu, Z. MiR-874 promotes intestinal barrier dysfunction through targeting AQP3 following intestinal ischemic injury. FEBS Lett. 2014, 588, 757–763. [Google Scholar] [CrossRef] [PubMed]
  183. Su, Z.; Zhi, X.; Zhang, Q.; Yang, L.; Xu, H.; Xu, Z. LncRNA H19 functions as a competing endogenous RNA to regulate AQP3 expression by sponging miR-874 in the intestinal barrier. FEBS Lett. 2016, 590, 1354–1364. [Google Scholar] [CrossRef] [PubMed]
  184. Ratovitski, E.A. Phospho-DeltaNp63alpha regulates AQP3, ALOX12B, CASP14 and CLDN1 expression through transcription and microRNA modulation. FEBS Lett. 2013, 587, 3581–3586. [Google Scholar] [CrossRef] [PubMed]
  185. Zhou, C.; Kong, W.; Ju, T.; Xie, Q.; Zhai, L. MiR-185-3p mimic promotes the chemosensitivity of CRC cells via AQP5. Cancer Biol. Ther. 2020, 21, 790–798. [Google Scholar] [CrossRef] [PubMed]
  186. Zhu, H.; Wu, Y.; Kang, M.; Zhang, B. MiR-877 suppresses gastric cancer progression by downregulating AQP3. J. Int. Med. Res. 2020, 48, 0300060520903661. [Google Scholar] [CrossRef]
  187. Li, Z.; Ma, L.; Di, L.; Lin, X. MicroRNA-1271-5p alleviates the malignant development of hepatitis B virus-mediated liver cancer via binding to AQP5. Mol. Med. Rep. 2021, 23, 386. [Google Scholar] [CrossRef]
  188. Sekine, S.; Shimada, Y.; Nagata, T.; Sawada, S.; Yoshioka, I.; Matsui, K.; Moriyama, M.; Omura, T.; Osawa, S.; Shibuya, K.; et al. Role of aquaporin-5 in gallbladder carcinoma. Eur. Surg. Res. 2013, 51, 108–117. [Google Scholar] [CrossRef] [PubMed]
  189. Chao, G.; Wang, Y.; Zhang, S.; Yang, W.; Ni, Z.; Zheng, X. Correction: MicroRNA-29a increased the intestinal membrane permeability of colonic epithelial cells in irritable bowel syndrome rats. Oncotarget 2018, 9, 15816. [Google Scholar] [CrossRef] [PubMed]
  190. Zhang, Y.; Chen, M.; Zhang, Y.; Peng, P.; Li, J.; Xin, X. miR-96 and miR-330 overexpressed and targeted AQP5 in lipopolysaccharide-induced rat lung damage of disseminated intravascular coagulation. Blood Coagul. Fibrinolysis 2014, 25, 731–737. [Google Scholar] [CrossRef] [PubMed]
  191. Chu, Y.; Wang, X.; Yu, N.; Li, Y.; Kan, J. Long non-coding RNA FGD5-AS1/microRNA-133a-3p upregulates aquaporin 1 to decrease the inflammatory response in LPS-induced sepsis. Mol. Med. Rep. 2021, 24, 784. [Google Scholar] [CrossRef] [PubMed]
  192. Tang, R.; Pei, L.; Bai, T.; Wang, J. Down-regulation of microRNA-126-5p contributes to overexpression of VEGFA in lipopolysaccharide-induced acute lung injury. Biotechnol. Lett. 2016, 38, 1277–1284. [Google Scholar] [CrossRef]
  193. Zhu, L.; Lin, Z.W.; Wang, G.; Zhang, H.; Liu, B.; Xu, Q.J. MicroRNA-495 downregulates AQP1 and facilitates proliferation and differentiation of osteoblasts in mice with tibial fracture through activation of p38 MAPK signaling pathway. Sci. Rep. 2019, 9, 16171. [Google Scholar] [CrossRef]
  194. Tanaka, M.; Ito, A.; Shiozawa, S.; Hara-Chikuma, M. Anti-tumor effect of aquaporin 3 monoclonal antibody on syngeneic mouse tumor model. Transl. Oncol. 2022, 24, 101498. [Google Scholar] [CrossRef] [PubMed]
  195. Li, H.; Shi, H.; Gao, M.; Ma, N.; Sun, R. Long non-coding RNA CASC2 improved acute lung injury by regulating miR-144-3p/AQP1 axis to reduce lung epithelial cell apoptosis. Cell Biosci. 2018, 8, 15. [Google Scholar] [CrossRef] [PubMed]
Figure 1. General three-dimensional structure of aquaporins. (A) Side and (B) top view of human AQP5 tetrameric structure, depicted in ribbon representation. (C) Side view of AQP5 monomer hourglass structure. Picture edited in UCSF Chimera software 1.15 (PDB code: 3D9S). (D) Schematic representation of aquaporin topology within membrane. Each monomer comprises six transmembrane helices (H1–H6) connected by five loops (A–E). Loops B and E fold into membrane, constituting two helices containing conserved NPA (Asn-Pro-Ala) motifs, with both N-terminal and C-terminal domains located in cytoplasm.
Figure 1. General three-dimensional structure of aquaporins. (A) Side and (B) top view of human AQP5 tetrameric structure, depicted in ribbon representation. (C) Side view of AQP5 monomer hourglass structure. Picture edited in UCSF Chimera software 1.15 (PDB code: 3D9S). (D) Schematic representation of aquaporin topology within membrane. Each monomer comprises six transmembrane helices (H1–H6) connected by five loops (A–E). Loops B and E fold into membrane, constituting two helices containing conserved NPA (Asn-Pro-Ala) motifs, with both N-terminal and C-terminal domains located in cytoplasm.
Ijms 26 01330 g001
Figure 2. Role of aquaporins (AQPs) in cancer cell migration and proliferation. (A) AQPs are involved in cancer cell migration by facilitating fast water fluxes, generating local hydrostatic pressure that allows lamellipodium expansion for actin polymerization. (B) AQPs are overexpressed in cancer cells, influencing their proliferation through regulation of glycerol metabolism and modulation of signaling pathways such as PI3K, ERK and Wnt.
Figure 2. Role of aquaporins (AQPs) in cancer cell migration and proliferation. (A) AQPs are involved in cancer cell migration by facilitating fast water fluxes, generating local hydrostatic pressure that allows lamellipodium expansion for actin polymerization. (B) AQPs are overexpressed in cancer cells, influencing their proliferation through regulation of glycerol metabolism and modulation of signaling pathways such as PI3K, ERK and Wnt.
Ijms 26 01330 g002
Figure 3. A schematic representation of the role of AQPs as transceptors in cancer. AQPs are overexpressed in cancer cells, contributing to tumor progression through the modulation of key signaling pathways, including Wnt, PI3K and MAPK, and by interacting with the cytoskeleton, probably through the activation of their receptor activity by effector molecules (E). Targeting AQPs with specific inhibitors (I) can disrupt actin cytoskeleton organization and impair the activation of the downstream effectors of cancer signaling, thereby affecting tumor growth and spread. Arrows on the left represent the activation of signaling pathways and tumorigenic processes, while arrows on the right indicate their inhibition. The red “X” represents the blockage of AQP activity.
Figure 3. A schematic representation of the role of AQPs as transceptors in cancer. AQPs are overexpressed in cancer cells, contributing to tumor progression through the modulation of key signaling pathways, including Wnt, PI3K and MAPK, and by interacting with the cytoskeleton, probably through the activation of their receptor activity by effector molecules (E). Targeting AQPs with specific inhibitors (I) can disrupt actin cytoskeleton organization and impair the activation of the downstream effectors of cancer signaling, thereby affecting tumor growth and spread. Arrows on the left represent the activation of signaling pathways and tumorigenic processes, while arrows on the right indicate their inhibition. The red “X” represents the blockage of AQP activity.
Ijms 26 01330 g003
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

Pimpão, C.; da Silva, I.V.; Soveral, G. The Expanding Role of Aquaporin-1, Aquaporin-3 and Aquaporin-5 as Transceptors: Involvement in Cancer Development and Potential Druggability. Int. J. Mol. Sci. 2025, 26, 1330. https://doi.org/10.3390/ijms26031330

AMA Style

Pimpão C, da Silva IV, Soveral G. The Expanding Role of Aquaporin-1, Aquaporin-3 and Aquaporin-5 as Transceptors: Involvement in Cancer Development and Potential Druggability. International Journal of Molecular Sciences. 2025; 26(3):1330. https://doi.org/10.3390/ijms26031330

Chicago/Turabian Style

Pimpão, Catarina, Inês V. da Silva, and Graça Soveral. 2025. "The Expanding Role of Aquaporin-1, Aquaporin-3 and Aquaporin-5 as Transceptors: Involvement in Cancer Development and Potential Druggability" International Journal of Molecular Sciences 26, no. 3: 1330. https://doi.org/10.3390/ijms26031330

APA Style

Pimpão, C., da Silva, I. V., & Soveral, G. (2025). The Expanding Role of Aquaporin-1, Aquaporin-3 and Aquaporin-5 as Transceptors: Involvement in Cancer Development and Potential Druggability. International Journal of Molecular Sciences, 26(3), 1330. https://doi.org/10.3390/ijms26031330

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