1
|
Nourmohammadi S, Henderson SW, Ramesh SA, Yool AJ. Characterization of human aquaporin ion channels in a yeast expression system as a tool for novel ion channel discovery. Biosci Rep 2024; 44:BSR20240542. [PMID: 39069912 PMCID: PMC11358751 DOI: 10.1042/bsr20240542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 07/15/2024] [Accepted: 07/26/2024] [Indexed: 07/30/2024] Open
Abstract
Aquaporin (AQP) channels found in all domains of life are transmembrane proteins which mediate passive transport of water, glycerol, signaling molecules, metabolites, and charged solutes. Discovery of new classes of ion-conducting AQP channels has been slow, likely reflecting time- and labor-intensive methods required for traditional electrophysiology. Work here defines a sensitive mass-throughput system for detecting AQP ion channels, identified by rescue of cell growth in the K+-transport-defective yeast strain CY162 following genetic complementation with heterologously expressed cation-permeable channels, using the well characterized human AQP1 channel for proof of concept. Results showed AQP1 conferred transmembrane permeability to cations which rescued survival in CY162 yeast. Comprehensive testing showed that growth response properties fully recapitulated AQP1 pharmacological agonist and antagonist profiles for activation, inhibition, dose-dependence, and structure-function relationships, demonstrating validity of the yeast screening tool for AQP channel identification and drug discovery efforts. This method also provided new information on divalent cation blockers of AQP1, pH sensitivity of antagonists, and ion permeability of human AQP6. Site-directed mutagenesis of AQP1 channel regulatory domains confirmed that yeast growth rescue was mediated by the introduced channels. Optical monitoring with a lithium-sensitive photoswitchable probe in living cells independently demonstrated monovalent cation permeability of AQP1 channels in yeast plasma membrane. Ion channel properties of AQP1 expressed in yeast were consistent with those of AQP1 expressed in Xenopus laevis oocyte and K+-transport defective Escherichia coli. Outcomes here establish a powerful new approach for efficient screening of phylogenetically diverse AQPs for yet untested functions as cation channels.
Collapse
Affiliation(s)
- Saeed Nourmohammadi
- School of Biomedicine, Faculty of Health and Medical Sciences, and the Institute for Photonics and Advanced Sensing, University of Adelaide, Adelaide, SA 5005, Australia
| | - Sam W Henderson
- School of Biomedicine, Faculty of Health and Medical Sciences, and the Institute for Photonics and Advanced Sensing, University of Adelaide, Adelaide, SA 5005, Australia
| | - Sunita A Ramesh
- Biological Sciences, College of Science and Engineering, Flinders University, Bedford Park, SA 5042, Australia
| | - Andrea J Yool
- School of Biomedicine, Faculty of Health and Medical Sciences, and the Institute for Photonics and Advanced Sensing, University of Adelaide, Adelaide, SA 5005, Australia
| |
Collapse
|
2
|
Sharma Y, Thakral V, Raturi G, Dutta Dubey K, Sonah H, Pareek A, Sharma TR, Deshmukh R. Structural assessment of OsNIP2;1 highlighted critical residues defining solute specificity and functionality of NIP class aquaporins. J Adv Res 2024; 58:1-11. [PMID: 37164213 PMCID: PMC10982858 DOI: 10.1016/j.jare.2023.04.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 04/30/2023] [Accepted: 04/30/2023] [Indexed: 05/12/2023] Open
Abstract
INTRODUCTION Nodulin-26-like intrinsic proteins (NIPs) are integral membrane proteins belonging to the aquaporin family, that facilitate the transport of neutral solutes across the bilayer. The OsNIP2;1 a member of NIP-III class of aquaporins is permeable to beneficial elements like silicon and hazardous arsenic. However, the atomistic cross-talk of these molecules traversing the OsNIP2;1 channel is not well understood. OBJECTIVE Due to the lack of genomic variation but the availability of high confidence crystal structure, this study aims to highlight structural determinants of metalloid permeation through OsNIP2;1. METHODS The molecular simulations, combined with site-directed mutagenesis were used to probe the role of specific residues in the metalloid transport activity of OsNIP2;1. RESULTS We drew energetic landscape of OsNIP2;1, for silicic and arsenous acid transport. Potential Mean Force (PMF) construction illuminate three prominent energetic barriers for metalloid passage through the pore. One corresponds to the extracellular molecular entry in the channel, the second located on ar/R filter, and the third size constriction in the cytoplasmic half. Comparative PMF for silicic acid and arsenous acid elucidate a higher barrier for silicic acid at the cytoplasmic constrict resulting in longer residence time for silicon. Furthermore, our simulation studies explained the importance of conserved residues in loop-C and loop-D with a direct effect on pore dynamics and metalloid transport. Next we assessed contribution of predicted key residues for arsenic uptake, by functional complementation in yeast. With the aim of reducing arsenic uptake while maintaining beneficial elements uptake, we identified novel OsNIP2;1 mutants with substantial reduction in arsenic uptake in yeast. CONCLUSION We provide a comprehensive assessment of pore lining residues of OsNIP2;1 with respect to metalloid uptake. The findings will expand mechanistic understanding of aquaporin's metalloid selectivity and facilitate variant interpretation to develop novel alleles with preference for beneficial metalloid species and reducing hazardous ones.
Collapse
Affiliation(s)
- Yogesh Sharma
- National Agri-Food Biotechnology Institute, Mohali, Punjab, India; Regional Centre for Biotechnology, Faridabad, Haryana (NCR Delhi), India
| | - Vandana Thakral
- National Agri-Food Biotechnology Institute, Mohali, Punjab, India; Department of Biotechnology, Panjab University, Chandigarh, India
| | - Gaurav Raturi
- National Agri-Food Biotechnology Institute, Mohali, Punjab, India; Department of Biotechnology, Panjab University, Chandigarh, India
| | - Kshatresh Dutta Dubey
- Department of Chemistry, School of Natural Sciences, Shiv Nadar Institute of Eminence, Gautam Buddha Nagar, Uttar Pradesh, India
| | - Humira Sonah
- National Agri-Food Biotechnology Institute, Mohali, Punjab, India; Department of Biotechnology, Central University of Haryana, Mahendragarh, Haryana, India
| | - Ashwani Pareek
- National Agri-Food Biotechnology Institute, Mohali, Punjab, India
| | - Tilak Raj Sharma
- National Agri-Food Biotechnology Institute, Mohali, Punjab, India; Indian Council of Agricultural Research, Division of Crop Science, Krishi Bhavan, New Delhi, India
| | - Rupesh Deshmukh
- National Agri-Food Biotechnology Institute, Mohali, Punjab, India; Plaksha University, Mohali, Punjab, India; Department of Biotechnology, Central University of Haryana, Mahendragarh, Haryana, India.
| |
Collapse
|
3
|
Khan S, Lokman NA, Oehler MK, Ricciardelli C, Yool AJ. Reducing the Invasiveness of Low- and High-Grade Endometrial Cancers in Both Primary Human Cancer Biopsies and Cell Lines by the Inhibition of Aquaporin-1 Channels. Cancers (Basel) 2023; 15:4507. [PMID: 37760476 PMCID: PMC10526386 DOI: 10.3390/cancers15184507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/29/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
Aquaporin (AQP) channels in endometrial cancer (EC) cells are of interest as pharmacological targets to reduce tumor progression. A panel of compounds, including AQP1 ion channel inhibitors (AqB011 and 5-(phenoxymethyl) furan-2-carbaldehyde, PMFC), were used to test the hypothesis that inhibition of key AQPs can limit the invasiveness of low- and high-grade EC cells. We evaluated the effects on transwell migration in EC cell lines (Ishikawa, MFE-280) and primary EC cells established from surgical tissues (n = 8). Quantitative PCR uncovered classes of AQPs not previously reported in EC that are differentially regulated by hormonal signaling. With estradiol, Ishikawa showed increased AQPs 5, 11, 12, and decreased AQPs 0 and 4; MFE-280 showed increased AQPs 0, 1, 3, 4, 8, and decreased AQP11. Protein expression was confirmed by Western blot and immunocytochemistry. AQPs 1, 4, and 11 were colocalized with plasma membrane marker; AQP8 was intracellular in Ishikawa and not detectable in MFE-280. AQP1 ion channel inhibitors (AqB011; PMFC) reduced invasiveness of EC cell lines in transwell chamber and spheroid dispersal assays. In Ishikawa cells, transwell invasiveness was reduced ~41% by 80 µM AqB011 and ~55% by 0.5 mM 5-PMFC. In MFE-280, 5-PMFC inhibited invasion by ~77%. In contrast, proposed inhibitors of AQP water pores (acetazolamide, ginsenoside, KeenMind, TGN-020, IMD-0354) were not effective. Treatments of cultured primary EC cells with AqB011 or PMFC significantly reduced the invasiveness of both low- and high-grade primary EC cells in transwell chambers. We confirmed the tumors expressed moderate to high levels of AQP1 detected by immunohistochemistry, whereas expression levels of AQP4, AQP8, and AQP11 were substantially lower. The anti-invasive potency of AqB011 treatment for EC tumor tissues showed a positive linear correlation with AQP1 expression levels. In summary, AQP1 ion channels are important for motility in both low- and high-grade EC subtypes. Inhibition of AQP1 is a promising strategy to inhibit EC invasiveness and improve patient outcomes.
Collapse
Affiliation(s)
- Sidra Khan
- School of Biomedicine, University of Adelaide, Adelaide, SA 5000, Australia;
| | - Noor A. Lokman
- Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide, SA 5000, Australia; (N.A.L.); (M.K.O.)
| | - Martin K. Oehler
- Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide, SA 5000, Australia; (N.A.L.); (M.K.O.)
- Department of Gynaecological Oncology, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
| | - Carmela Ricciardelli
- Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide, SA 5000, Australia; (N.A.L.); (M.K.O.)
| | - Andrea J. Yool
- School of Biomedicine, University of Adelaide, Adelaide, SA 5000, Australia;
| |
Collapse
|
4
|
Henderson SW, Nakayama Y, Whitelaw ML, Bruning JB, Anderson PA, Tyerman SD, Ramesh SA, Martinac B, Yool AJ. Proteoliposomes reconstituted with human aquaporin-1 reveal novel single-ion-channel properties. BIOPHYSICAL REPORTS 2023; 3:100100. [PMID: 36949749 PMCID: PMC10025285 DOI: 10.1016/j.bpr.2023.100100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 01/11/2023] [Indexed: 01/15/2023]
Abstract
Human aquaporin 1 (hAQP1) forms homotetrameric channels that facilitate fluxes of water and small solutes across cell membranes. In addition to water channel activity, hAQP1 displays non-selective monovalent cation-channel activity gated by intracellular cyclic GMP. Dual water and ion-channel activity of hAQP1, thought to regulate cell shape and volume, could offer a target for novel therapeutics relevant to controlling cancer cell invasiveness. This study probed properties of hAQP1 ion channels using proteoliposomes, which, unlike conventional cell-based systems such as Xenopus laevis oocytes, are relatively free of background ion channels. Histidine-tagged recombinant hAQP1 protein was synthesized and purified from the methylotrophic yeast, Pichia pastoris, and reconstituted into proteoliposomes for biophysical analyses. Osmotic water channel activity confirmed correct folding and channel assembly. Ion-channel activity of hAQP1-Myc-His6 was recorded by patch-clamp electrophysiology with excised patches. In symmetrical potassium, the hAQP1-Myc-His6 channels displayed coordinated gating, a single-channel conductance of approximately 75 pS, and multiple subconductance states. Applicability of this method for structure-function analyses was tested using hAQP1-Myc-His6 D48A/D185A channels modified by site-directed mutations of charged Asp residues estimated to be adjacent to the central ion-conducting pore of the tetramer. No differences in conductance were detected between mutant and wild-type constructs, suggesting the open-state conformation could differ substantially from expectations based on crystal structures. Nonetheless, the method pioneered here for AQP1 demonstrates feasibility for future work defining structure-function relationships, screening pharmacological inhibitors, and testing other classes in the broad family of aquaporins for previously undiscovered ion-conducting capabilities.
Collapse
Affiliation(s)
- Sam W. Henderson
- School of Biomedicine, University of Adelaide, Adelaide, SA 5005, Australia
| | - Yoshitaka Nakayama
- Victor Chang Cardiac Research Institute, Lowy Packer Building, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, UNSW Medicine & Health, St Vincent’s Healthcare Clinical Campus, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW Australia
| | - Murray L. Whitelaw
- Institute of Photonics and Advanced Sensing, The School of Biological Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| | - John B. Bruning
- Institute of Photonics and Advanced Sensing, The School of Biological Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| | - Peter A. Anderson
- School of Biological Sciences, Faculty of Science and Engineering, Flinders University, Adelaide, SA 5001, Australia
| | - Stephen D. Tyerman
- ARC Centre of Excellence in Plant Energy Biology, School of Agriculture, Food and Wine & Waite Research Institute, University of Adelaide, Glen Osmond, SA 5064, Australia
| | - Sunita A. Ramesh
- School of Biological Sciences, Faculty of Science and Engineering, Flinders University, Adelaide, SA 5001, Australia
| | - Boris Martinac
- Victor Chang Cardiac Research Institute, Lowy Packer Building, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, UNSW Medicine & Health, St Vincent’s Healthcare Clinical Campus, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW Australia
| | - Andrea J. Yool
- School of Biomedicine, University of Adelaide, Adelaide, SA 5005, Australia
| |
Collapse
|
5
|
Aquaporins Display a Diversity in their Substrates. J Membr Biol 2023; 256:1-23. [PMID: 35986775 DOI: 10.1007/s00232-022-00257-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 07/13/2022] [Indexed: 02/07/2023]
Abstract
Aquaporins constitute a family of transmembrane proteins that function to transport water and other small solutes across the cell membrane. Aquaporins family members are found in diverse life forms. Aquaporins share the common structural fold consisting of six transmembrane alpha helices with a central water-transporting channel. Four such monomers assemble together to form tetramers as their biological unit. Initially, aquaporins were discovered as water-transporting channels, but several studies supported their involvement in mediating the facilitated diffusion of different solutes. The so-called water channel is able to transport a variety of substrates ranging from a neutral molecule to a charged molecule or a small molecule to a bulky molecule or even a gas molecule. This article gives an overview of a diverse range of substrates conducted by aquaporin family members. Prime focus is on human aquaporins where aquaporins show a wide tissue distribution and substrate specificity leading to various physiological functions. This review also highlights the structural mechanisms leading to the transport of water and glycerol. More research is needed to understand how one common fold enables the aquaporins to transport an array of solutes.
Collapse
|
6
|
Ozu M, Alvear-Arias JJ, Fernandez M, Caviglia A, Peña-Pichicoi A, Carrillo C, Carmona E, Otero-Gonzalez A, Garate JA, Amodeo G, Gonzalez C. Aquaporin Gating: A New Twist to Unravel Permeation through Water Channels. Int J Mol Sci 2022; 23:12317. [PMID: 36293170 PMCID: PMC9604103 DOI: 10.3390/ijms232012317] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 09/30/2022] [Accepted: 10/05/2022] [Indexed: 11/17/2022] Open
Abstract
Aquaporins (AQPs) are small transmembrane tetrameric proteins that facilitate water, solute and gas exchange. Their presence has been extensively reported in the biological membranes of almost all living organisms. Although their discovery is much more recent than ion transport systems, different biophysical approaches have contributed to confirm that permeation through each monomer is consistent with closed and open states, introducing the term gating mechanism into the field. The study of AQPs in their native membrane or overexpressed in heterologous systems have experimentally demonstrated that water membrane permeability can be reversibly modified in response to specific modulators. For some regulation mechanisms, such as pH changes, evidence for gating is also supported by high-resolution structures of the water channel in different configurations as well as molecular dynamics simulation. Both experimental and simulation approaches sustain that the rearrangement of conserved residues contributes to occlude the cavity of the channel restricting water permeation. Interestingly, specific charged and conserved residues are present in the environment of the pore and, thus, the tetrameric structure can be subjected to alter the positions of these charges to sustain gating. Thus, is it possible to explore whether the displacement of these charges (gating current) leads to conformational changes? To our knowledge, this question has not yet been addressed at all. In this review, we intend to analyze the suitability of this proposal for the first time.
Collapse
Affiliation(s)
- Marcelo Ozu
- Department of Biodiversity and Experimental Biology, Faculty of Exact & Natural Sciences, University of Buenos Aires, Buenos Aires C1053, Argentina
- CONICET—Institute of Biodiversity and Experimental and Applied Biology CONICET (IBBEA), University of Buenos Aires, Buenos Aires C1053, Argentina
| | - Juan José Alvear-Arias
- Interdisciplinary Center of Neurosciences of Valparaiso, University of Valparaiso, CINV, Valparaíso 2360102, Chile
- Millennium Nucleus in NanoBioPhysics, Scientific and Technologic Center of Excellence of Science and Life, Santiago 7750000, Chile
| | - Miguel Fernandez
- Interdisciplinary Center of Neurosciences of Valparaiso, University of Valparaiso, CINV, Valparaíso 2360102, Chile
- Millennium Nucleus in NanoBioPhysics, Scientific and Technologic Center of Excellence of Science and Life, Santiago 7750000, Chile
| | - Agustín Caviglia
- CONICET—Institute of Biodiversity and Experimental and Applied Biology CONICET (IBBEA), University of Buenos Aires, Buenos Aires C1053, Argentina
| | - Antonio Peña-Pichicoi
- Interdisciplinary Center of Neurosciences of Valparaiso, University of Valparaiso, CINV, Valparaíso 2360102, Chile
- Millennium Nucleus in NanoBioPhysics, Scientific and Technologic Center of Excellence of Science and Life, Santiago 7750000, Chile
| | - Christian Carrillo
- Interdisciplinary Center of Neurosciences of Valparaiso, University of Valparaiso, CINV, Valparaíso 2360102, Chile
- Millennium Nucleus in NanoBioPhysics, Scientific and Technologic Center of Excellence of Science and Life, Santiago 7750000, Chile
| | - Emerson Carmona
- Cell Physiology and Molecular Biophysics Department and the Center for Membrane Protein Research, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Anselmo Otero-Gonzalez
- Center of Protein Study, Faculty of Biology, University of Havana, La Habana 10400, Cuba
| | - José Antonio Garate
- Interdisciplinary Center of Neurosciences of Valparaiso, University of Valparaiso, CINV, Valparaíso 2360102, Chile
- Millennium Nucleus in NanoBioPhysics, Scientific and Technologic Center of Excellence of Science and Life, Santiago 7750000, Chile
- Faculty of Engineering and Technology, University of San Sebastian, Santiago 8420524, Chile
| | - Gabriela Amodeo
- Department of Biodiversity and Experimental Biology, Faculty of Exact & Natural Sciences, University of Buenos Aires, Buenos Aires C1053, Argentina
- CONICET—Institute of Biodiversity and Experimental and Applied Biology CONICET (IBBEA), University of Buenos Aires, Buenos Aires C1053, Argentina
| | - Carlos Gonzalez
- Interdisciplinary Center of Neurosciences of Valparaiso, University of Valparaiso, CINV, Valparaíso 2360102, Chile
- Millennium Nucleus in NanoBioPhysics, Scientific and Technologic Center of Excellence of Science and Life, Santiago 7750000, Chile
- Department of Physiology and Biophysics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
7
|
Critical Role of Aquaporins in Cancer: Focus on Hematological Malignancies. Cancers (Basel) 2022; 14:cancers14174182. [PMID: 36077720 PMCID: PMC9455074 DOI: 10.3390/cancers14174182] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/23/2022] [Accepted: 08/26/2022] [Indexed: 12/04/2022] Open
Abstract
Simple Summary Aquaporins are proteins able to regulate the transfer of water and other small substances such as ions, glycerol, urea, and hydrogen peroxide across cellular membranes. AQPs provide for a huge variety of physiological phenomena; their alteration provokes several types of pathologies including cancer and hematological malignancies. Our review presents data revealing the possibility of employing aquaporins as biomarkers in patients with hematological malignancies and evaluates the possibility that interfering with the expression of aquaporins could represent an effective treatment for hematological malignancies. Abstract Aquaporins are transmembrane molecules regulating the transfer of water and other compounds such as ions, glycerol, urea, and hydrogen peroxide. Their alteration has been reported in several conditions such as cancer. Tumor progression might be enhanced by aquaporins in modifying tumor angiogenesis, cell volume adaptation, proteases activity, cell–matrix adhesions, actin cytoskeleton, epithelial–mesenchymal transitions, and acting on several signaling pathways facilitating cancer progression. Close connections have also been identified between the aquaporins and hematological malignancies. However, it is difficult to identify a unique action exerted by aquaporins in different hemopathies, and each aquaporin has specific effects that vary according to the class of aquaporin examined and to the different neoplastic cells. However, the expression of aquaporins is altered in cell cultures and in patients with acute and chronic myeloid leukemia, in lymphoproliferative diseases and in multiple myeloma, and the different expression of aquaporins seems to be able to influence the efficacy of treatment and could have a prognostic significance, as greater expression of aquaporins is correlated to improved overall survival in leukemia patients. Finally, we assessed the possibility that modifying the aquaporin expression using aquaporin-targeting regulators, specific monoclonal antibodies, and even aquaporin gene transfer could represent an effective therapy of hematological malignancies.
Collapse
|
8
|
Markou A, Unger L, Abir-Awan M, Saadallah A, Halsey A, Balklava Z, Conner M, Törnroth-Horsefield S, Greenhill SD, Conner A, Bill RM, Salman MM, Kitchen P. Molecular mechanisms governing aquaporin relocalisation. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2022; 1864:183853. [PMID: 34973181 PMCID: PMC8825993 DOI: 10.1016/j.bbamem.2021.183853] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 02/07/2023]
Abstract
The aquaporins (AQPs) form a family of integral membrane proteins that facilitate the movement of water across biological membrane by osmosis, as well as facilitating the diffusion of small polar solutes. AQPs have been recognised as drug targets for a variety of disorders associated with disrupted water or solute transport, including brain oedema following stroke or trauma, epilepsy, cancer cell migration and tumour angiogenesis, metabolic disorders, and inflammation. Despite this, drug discovery for AQPs has made little progress due to a lack of reproducible high-throughput assays and difficulties with the druggability of AQP proteins. However, recent studies have suggested that targetting the trafficking of AQP proteins to the plasma membrane is a viable alternative drug target to direct inhibition of the water-conducting pore. Here we review the literature on the trafficking of mammalian AQPs with a view to highlighting potential new drug targets for a variety of conditions associated with disrupted water and solute homeostasis.
Collapse
Affiliation(s)
- Andrea Markou
- College of Health and Life Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK
| | - Lucas Unger
- College of Health and Life Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK
| | - Mohammed Abir-Awan
- College of Health and Life Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK
| | - Ahmed Saadallah
- College of Health and Life Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK
| | - Andrea Halsey
- MRC Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Zita Balklava
- College of Health and Life Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK
| | - Matthew Conner
- School of Sciences, Research Institute in Healthcare Science, University of Wolverhampton, Wolverhampton WV1 1LY, UK
| | | | - Stuart D Greenhill
- College of Health and Life Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK
| | - Alex Conner
- Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Roslyn M Bill
- College of Health and Life Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK
| | - Mootaz M Salman
- Department of Physiology Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK; Oxford Parkinson's Disease Centre, University of Oxford, South Parks Road, Oxford OX1 3QX, UK.
| | - Philip Kitchen
- College of Health and Life Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK.
| |
Collapse
|
9
|
Chow PH, Cox CD, Pei JV, Anabaraonye N, Nourmohammadi S, Henderson SW, Martinac B, Abdulmalik O, Yool AJ. Inhibition of the Aquaporin-1 Cation Conductance by Selected Furan Compounds Reduces Red Blood Cell Sickling. Front Pharmacol 2022; 12:794791. [PMID: 35111062 PMCID: PMC8801817 DOI: 10.3389/fphar.2021.794791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/06/2021] [Indexed: 11/13/2022] Open
Abstract
In sickle cell disease (SCD), the pathological shift of red blood cells (RBCs) into distorted morphologies under hypoxic conditions follows activation of a cationic leak current (Psickle) and cell dehydration. Prior work showed sickling was reduced by 5-hydroxylmethyl-2-furfural (5-HMF), which stabilized mutant hemoglobin and also blocked the Psickle current in RBCs, though the molecular basis of this 5-HMF-sensitive cation current remained a mystery. Work here is the first to test the hypothesis that Aquaporin-1 (AQP1) cation channels contribute to the monovalent component of Psickle. Human AQP1 channels expressed in Xenopus oocytes were evaluated for sensitivity to 5-HMF and four derivatives known to have differential efficacies in preventing RBC sickling. Ion conductances were measured by two-electrode voltage clamp, and osmotic water permeability by optical swelling assays. Compounds tested were: 5-HMF; 5-PMFC (5-(phenoxymethyl)furan-2-carbaldehyde); 5-CMFC (5-(4-chlorophenoxymethyl)furan-2-carbaldehyde); 5-NMFC (5-(2-nitrophenoxymethyl)-furan-2-carbaldehyde); and VZHE006 (tert-butyl (5-formylfuran-2-yl)methyl carbonate). The most effective anti-sickling agent, 5-PMFC, was the most potent inhibitor of the AQP1 ion conductance (98% block at 100 µM). The order of sensitivity of the AQP1 conductance to inhibition was 5-PMFC > VZHE006 > 5-CMFC ≥ 5-NMFC, which corresponded with effectiveness in protecting RBCs from sickling. None of the compounds altered AQP1 water channel activity. Combined application of a selective AQP1 ion channel blocker AqB011 (80 µM) with a selective hemoglobin modifying agent 5-NMFC (2.5 mM) increased anti-sickling effectiveness in red blood cells from human SCD patients. Another non-selective cation channel known to be expressed in RBCs, Piezo1, was unaffected by 2 mM 5-HMF. Results suggest that inhibition of AQP1 ion channels and capacity to modify hemoglobin are combined features of the most effective anti-sickling agents. Future therapeutics aimed at both targets could hold promise for improved treatments for SCD.
Collapse
Affiliation(s)
- Pak Hin Chow
- Aquaporin Physiology and Drug Discovery Program, School of Biomedicine, University of Adelaide, Adelaide, SA, Australia
| | - Charles D Cox
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, University of New South Wales, Darlinghurst, NSW, Australia
| | - Jinxin V Pei
- Research School of Biology, College of Science, Australian National University, Canberra, ACT, Australia
| | - Nancy Anabaraonye
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Saeed Nourmohammadi
- Aquaporin Physiology and Drug Discovery Program, School of Biomedicine, University of Adelaide, Adelaide, SA, Australia
| | - Sam W Henderson
- Aquaporin Physiology and Drug Discovery Program, School of Biomedicine, University of Adelaide, Adelaide, SA, Australia
| | - Boris Martinac
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, University of New South Wales, Darlinghurst, NSW, Australia
| | - Osheiza Abdulmalik
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Andrea J Yool
- Aquaporin Physiology and Drug Discovery Program, School of Biomedicine, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
10
|
Wagner K, Unger L, Salman MM, Kitchen P, Bill RM, Yool AJ. Signaling Mechanisms and Pharmacological Modulators Governing Diverse Aquaporin Functions in Human Health and Disease. Int J Mol Sci 2022; 23:1388. [PMID: 35163313 PMCID: PMC8836214 DOI: 10.3390/ijms23031388] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/18/2022] [Accepted: 01/20/2022] [Indexed: 02/07/2023] Open
Abstract
The aquaporins (AQPs) are a family of small integral membrane proteins that facilitate the bidirectional transport of water across biological membranes in response to osmotic pressure gradients as well as enable the transmembrane diffusion of small neutral solutes (such as urea, glycerol, and hydrogen peroxide) and ions. AQPs are expressed throughout the human body. Here, we review their key roles in fluid homeostasis, glandular secretions, signal transduction and sensation, barrier function, immunity and inflammation, cell migration, and angiogenesis. Evidence from a wide variety of studies now supports a view of the functions of AQPs being much more complex than simply mediating the passive flow of water across biological membranes. The discovery and development of small-molecule AQP inhibitors for research use and therapeutic development will lead to new insights into the basic biology of and novel treatments for the wide range of AQP-associated disorders.
Collapse
Affiliation(s)
- Kim Wagner
- School of Biomedicine, University of Adelaide, Adelaide, SA 5005, Australia;
| | - Lucas Unger
- College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK; (L.U.); (P.K.)
| | - Mootaz M. Salman
- Department of Physiology Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK;
- Oxford Parkinson’s Disease Centre, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| | - Philip Kitchen
- College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK; (L.U.); (P.K.)
| | - Roslyn M. Bill
- College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK; (L.U.); (P.K.)
| | - Andrea J. Yool
- School of Biomedicine, University of Adelaide, Adelaide, SA 5005, Australia;
| |
Collapse
|
11
|
Salman MM, Kitchen P, Yool AJ, Bill RM. Recent breakthroughs and future directions in drugging aquaporins. Trends Pharmacol Sci 2022; 43:30-42. [PMID: 34863533 DOI: 10.1016/j.tips.2021.10.009] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/09/2021] [Accepted: 10/18/2021] [Indexed: 02/06/2023]
Abstract
Aquaporins facilitate the passive transport of water, solutes, or ions across biological membranes. They are implicated in diverse pathologies including brain edema following stroke or trauma, epilepsy, cancer cell migration and tumor angiogenesis, metabolic disorders, and inflammation. Despite this, there is no aquaporin-targeted drug in the clinic and aquaporins have been perceived to be intrinsically non-druggable targets. Here we challenge this idea, as viable routes to inhibition of aquaporin function have recently been identified, including targeting their regulation or their roles as channels for unexpected substrates. Identifying new drug development frameworks for conditions associated with disrupted water and solute homeostasis will meet the urgent, unmet clinical need of millions of patients for whom no pharmacological interventions are available.
Collapse
Affiliation(s)
- Mootaz M Salman
- Department of Physiology, Anatomy and Genetics, Kavli Institute for NanoScience Discovery, University of Oxford, Oxford OX1 3PT, UK; Oxford Parkinson's Disease Centre, University of Oxford, Oxford, UK.
| | - Philip Kitchen
- School of Biosciences, College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK.
| | - Andrea J Yool
- University of Adelaide, School of Biomedicine, Adelaide, South Australia 5005, Australia.
| | - Roslyn M Bill
- School of Biosciences, College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK.
| |
Collapse
|
12
|
Tyerman SD, McGaughey SA, Qiu J, Yool AJ, Byrt CS. Adaptable and Multifunctional Ion-Conducting Aquaporins. ANNUAL REVIEW OF PLANT BIOLOGY 2021; 72:703-736. [PMID: 33577345 DOI: 10.1146/annurev-arplant-081720-013608] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Aquaporins function as water and neutral solute channels, signaling hubs, disease virulence factors, and metabolon components. We consider plant aquaporins that transport ions compared to some animal counterparts. These are candidates for important, as yet unidentified, cation and anion channels in plasma, tonoplast, and symbiotic membranes. For those individual isoforms that transport ions, water, and gases, the permeability spans 12 orders of magnitude. This requires tight regulation of selectivity via protein interactions and posttranslational modifications. A phosphorylation-dependent switch between ion and water permeation in AtPIP2;1 might be explained by coupling between the gates of the four monomer water channels and the central pore of the tetramer. We consider the potential for coupling between ion and water fluxes that could form the basis of an electroosmotic transducer. A grand challenge in understanding the roles of ion transporting aquaporins is their multifunctional modes that are dependent on location, stress, time, and development.
Collapse
Affiliation(s)
- Stephen D Tyerman
- Australian Research Council (ARC) Centre of Excellence in Plant Energy Biology, School of Agriculture, Food and Wine, University of Adelaide, Glen Osmond, South Australia 5064, Australia; ,
| | - Samantha A McGaughey
- ARC Centre of Excellence for Translational Photosynthesis, Division of Plant Sciences, Research School of Biology, Australian National University, Acton, Australian Capital Territory 0200, Australia; ,
| | - Jiaen Qiu
- Australian Research Council (ARC) Centre of Excellence in Plant Energy Biology, School of Agriculture, Food and Wine, University of Adelaide, Glen Osmond, South Australia 5064, Australia; ,
| | - Andrea J Yool
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia 5005, Australia;
| | - Caitlin S Byrt
- ARC Centre of Excellence for Translational Photosynthesis, Division of Plant Sciences, Research School of Biology, Australian National University, Acton, Australian Capital Territory 0200, Australia; ,
| |
Collapse
|
13
|
Chow PH, Kourghi M, Pei JV, Nourmohammadi S, Yool AJ. 5-Hydroxymethyl-Furfural and Structurally Related Compounds Block the Ion Conductance in Human Aquaporin-1 Channels and Slow Cancer Cell Migration and Invasion. Mol Pharmacol 2020; 98:38-48. [PMID: 32434851 DOI: 10.1124/mol.119.119172] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 04/20/2020] [Indexed: 12/18/2022] Open
Abstract
Aquaporin-1 (AQP1) dual water and ion channels enhance migration and invasion when upregulated in leading edges of certain classes of cancer cells. Work here identifies structurally related furan compounds as novel inhibitors of AQP1 ion channels. 5-Hydroxymethyl-2-furfural (5HMF), a component of natural medicinal honeys, and three structurally related compounds, 5-nitro-2-furoic acid (5NFA), 5-acetoxymethyl-2-furaldehyde (5AMF), and methyl-5-nitro-2-furoate (M5NF), were analyzed for effects on water and ion channel activities of human AQP1 channels expressed in Xenopus oocytes. Two-electrode voltage clamp showed dose-dependent block of the AQP1 ion current by 5HMF (IC50 0.43 mM), 5NFA (IC50 1.2 mM), and 5AMF (IC50 ∼3 mM) but no inhibition by M5NF. In silico docking predicted the active ligands interacted with glycine 165, located in loop D gating domains surrounding the intracellular vestibule of the tetrameric central pore. Water fluxes through separate intrasubunit pores were unaltered by the furan compounds (at concentrations up to 5 mM). Effects on cell migration, invasion, and cytoskeletal organization in vitro were tested in high-AQP1-expressing cancer lines, colon cancer (HT29) and AQP1-expressing breast cancer (MDA), and low-AQP1-expressing SW480. 5HMF, 5NFA, and 5AMF selectively impaired cell motility in the AQP1-enriched cell lines. In contrast, M5NF immobilized all the cancer lines by disrupting actin cytoskeleton. No reduction in cell viability was observed at doses that were effective in blocking motility. These results define furans as a new class of AQP1 ion channel inhibitors for basic research and potential lead compounds for development of therapeutic agents targeting aquaporin channel activity. SIGNIFICANCE STATEMENT: 5-Hydroxymethyl-2-furfural (5HMF), a component of natural medicinal honeys, blocks the ion conductance but not the water flux through human Aquaporin-1 (AQP1) channels and impairs AQP1-dependent cell migration and invasiveness in cancer cell lines. Analyses of 5HMT and structural analogs demonstrate a structure-activity relationship for furan compounds, supported by in silico docking modeling. This work identifies new low-cost pharmacological antagonists for AQP1 available to researchers internationally. Furans merit consideration as a new class of therapeutic agents for controlling cancer metastasis.
Collapse
Affiliation(s)
- Pak Hin Chow
- Adelaide Medical School, University of Adelaide, Adelaide, Australia (P.H.C., M.K., J.V.P., S.N., A.J.Y.) and College of Science, The Australian National University, Canberra, Australia (J.V.P.)
| | - Mohamad Kourghi
- Adelaide Medical School, University of Adelaide, Adelaide, Australia (P.H.C., M.K., J.V.P., S.N., A.J.Y.) and College of Science, The Australian National University, Canberra, Australia (J.V.P.)
| | - Jinxin V Pei
- Adelaide Medical School, University of Adelaide, Adelaide, Australia (P.H.C., M.K., J.V.P., S.N., A.J.Y.) and College of Science, The Australian National University, Canberra, Australia (J.V.P.)
| | - Saeed Nourmohammadi
- Adelaide Medical School, University of Adelaide, Adelaide, Australia (P.H.C., M.K., J.V.P., S.N., A.J.Y.) and College of Science, The Australian National University, Canberra, Australia (J.V.P.)
| | - Andrea J Yool
- Adelaide Medical School, University of Adelaide, Adelaide, Australia (P.H.C., M.K., J.V.P., S.N., A.J.Y.) and College of Science, The Australian National University, Canberra, Australia (J.V.P.)
| |
Collapse
|
14
|
Yool AJ, Ramesh S. Molecular Targets for Combined Therapeutic Strategies to Limit Glioblastoma Cell Migration and Invasion. Front Pharmacol 2020; 11:358. [PMID: 32292341 PMCID: PMC7118801 DOI: 10.3389/fphar.2020.00358] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 03/10/2020] [Indexed: 12/21/2022] Open
Abstract
The highly invasive nature of glioblastoma imposes poor prospects for patient survival. Molecular evidence indicates glioblastoma cells undergo an intriguing expansion of phenotypic properties to include neuron-like signaling using excitable membrane ion channels and synaptic proteins, augmenting survival and motility. Neurotransmitter receptors, membrane signaling, excitatory receptors, and Ca2+ responses are important candidates for the design of customized treatments for cancers within the heterogeneous central nervous system. Relatively few published studies of glioblastoma multiforme (GBM) have evaluated pharmacological agents targeted to signaling pathways in limiting cancer cell motility. Transcriptomic analyses here identified classes of ion channels, ionotropic receptors, and synaptic proteins that are enriched in human glioblastoma biopsy samples. The pattern of GBM-enriched gene expression points to a major role for glutamate signaling. However, the predominant role of AMPA receptors in fast excitatory signaling throughout the central nervous system raises a challenge on how to target inhibitors selectively to cancer cells while maintaining tolerability. This review critically evaluates a panel of ligand- and voltage-gated ion channels and synaptic proteins upregulated in GBM, and the evidence for their potential roles in the pathological disease progress. Evidence suggests combinations of therapies could be more effective than single agents alone. Natural plant products used in traditional medicines for the treatment of glioblastoma contain flavonoids, terpenoids, polyphenols, epigallocatechin gallate, quinones, and saponins, which might serendipitously include agents that modulate some classes of signaling compounds highlighted in this review. New therapeutic strategies are likely to exploit evidence-based combinations of selected agents, each at a low dose, to create new cancer cell-specific therapeutics.
Collapse
Affiliation(s)
- Andrea J. Yool
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Sunita Ramesh
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
- College of Science and Engineering, Flinders University, Adelaide, SA, Australia
| |
Collapse
|
15
|
Calcino AD, de Oliveira AL, Simakov O, Schwaha T, Zieger E, Wollesen T, Wanninger A. The quagga mussel genome and the evolution of freshwater tolerance. DNA Res 2020; 26:411-422. [PMID: 31504356 PMCID: PMC6796509 DOI: 10.1093/dnares/dsz019] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Accepted: 08/01/2019] [Indexed: 02/06/2023] Open
Abstract
Freshwater dreissenid mussels evolved from marine ancestors during the Miocene ∼30 million years ago and today include some of the most successful and destructive invasive species of freshwater environments. Here, we sequenced the genome of the quagga mussel Dreissena rostriformis to identify adaptations involved in embryonic osmoregulation. We provide evidence that a lophotrochozoan-specific aquaporin water channel, a vacuolar ATPase subunit and a sodium/hydrogen exchanger are involved in osmoregulation throughout early cleavage, during which time large intercellular fluid-filled 'cleavage cavities' repeatedly form, coalesce and collapse, expelling excess water to the exterior. Independent expansions of aquaporins coinciding with at least five freshwater colonization events confirm their role in freshwater adaptation. Repeated aquaporin expansions and the evolution of membrane-bound fluid-filled osmoregulatory structures in diverse freshwater taxa point to a fundamental principle guiding the evolution of freshwater tolerance and provide a framework for future species control efforts.
Collapse
Affiliation(s)
- Andrew D Calcino
- Department of Integrative Zoology, University of Vienna, Vienna, Austria
| | | | - Oleg Simakov
- Department of Molecular Evolution and Development, University of Vienna, Vienna, Austria
| | - Thomas Schwaha
- Department of Integrative Zoology, University of Vienna, Vienna, Austria
| | - Elisabeth Zieger
- Department of Integrative Zoology, University of Vienna, Vienna, Austria
| | - Tim Wollesen
- Developmental Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Andreas Wanninger
- Department of Integrative Zoology, University of Vienna, Vienna, Austria
| |
Collapse
|
16
|
Wei H, Gao WS, Qi L, Jia L, Qu YJ, Yue SW, Zhang Y. Effect of cGMP-activated aquaporin 1 on TRPV4 in rats with allodynia induced by chronic compression of the dorsal root ganglion. Neurosci Lett 2019; 716:134630. [PMID: 31790718 DOI: 10.1016/j.neulet.2019.134630] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 11/07/2019] [Accepted: 11/13/2019] [Indexed: 02/01/2023]
Abstract
BACKGROUND The aim of this study was to investigate the effects of aquaporin 1 (AQP1) knockdown on allodynia in rats with chronic compression of the dorsal root ganglia (DRG) and the role of TRPV4 in these effects. METHODS Adult male Wistar rats were subjected to chronic compression of the dorsal root ganglia (CCD) via surgery. Behavioral tests were performed to calculate the paw withdrawal mechanical threshold (PWMT). Gene silence was induced by injecting rats with lentivirus expressing AQP1 short hairpin RNA (shRNA, Lv-shAQP1). Western blot analyses were performed to examine AQP1 and TRPV4 protein expression. The concentration of cyclic guanosine monophosphate (cGMP) was determined via enzyme-linked immunosorbent assay. RESULTS AQP1 protein levels in DRG neurons were significantly increased in CCD rats and were accompanied by a decrease in the PWMT. Lentivirus-mediated RNA interference of AQP1 decreased AQP1 protein expression in CCD rats and normalized their PWMT, but not in rats infected with lentivirus-expressing negative control short hairpin RNA. Furthermore, AQP1 was identified as a cGMP-gated channel. cGMP concentration was upregulated in CCD rats. This effect was attenuated by treatment with a cGMP inhibitor. Additionally, the cGMP inhibitor decreased the mechanical allodynia and AQP1 protein expression in CCD rats. Finally, levels of TRPV4 expression were upregulated in DRG neurons and the L4/L5 spinal cord following surgery, and these effects were reversed by treatment with Lv-shAQP1 or a cGMP inhibitor. CONCLUSION AQP1 plays a vital role in CCD-induced allodynia as Lv-shAQP1 significantly reduced the allodynia in CCD rats by inhibiting TRPV4 expression.
Collapse
Affiliation(s)
- Hui Wei
- Department of Physical Medicine & Rehabilitation, Qilu Hospital, Shandong University, Jinan, 250012, China.
| | - Wen-Shuang Gao
- Department of Physical Medicine & Rehabilitation, Qilu Hospital, Shandong University, Jinan, 250012, China.
| | - Lei Qi
- Department of Orthopedics, Qilu Hospital, Shandong University, Jinan 250012, China.
| | - Lei Jia
- Department of Physical Medicine & Rehabilitation, Qilu Hospital, Shandong University, Jinan, 250012, China.
| | - Yu-Juan Qu
- Department of Physical Medicine & Rehabilitation, Qilu Hospital, Shandong University, Jinan, 250012, China.
| | - Shou-Wei Yue
- Department of Physical Medicine & Rehabilitation, Qilu Hospital, Shandong University, Jinan, 250012, China.
| | - Yang Zhang
- Department of Physical Medicine & Rehabilitation, Qilu Hospital, Shandong University, Jinan, 250012, China
| |
Collapse
|
17
|
De Ieso ML, Pei JV, Nourmohammadi S, Smith E, Chow PH, Kourghi M, Hardingham JE, Yool AJ. Combined pharmacological administration of AQP1 ion channel blocker AqB011 and water channel blocker Bacopaside II amplifies inhibition of colon cancer cell migration. Sci Rep 2019; 9:12635. [PMID: 31477744 PMCID: PMC6718670 DOI: 10.1038/s41598-019-49045-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 08/13/2019] [Indexed: 12/14/2022] Open
Abstract
Aquaporin-1 (AQP1) has been proposed as a dual water and cation channel that when upregulated in cancers enhances cell migration rates; however, the mechanism remains unknown. Previous work identified AqB011 as an inhibitor of the gated human AQP1 cation conductance, and bacopaside II as a blocker of AQP1 water pores. In two colorectal adenocarcinoma cell lines, high levels of AQP1 transcript were confirmed in HT29, and low levels in SW480 cells, by quantitative PCR (polymerase chain reaction). Comparable differences in membrane AQP1 protein levels were demonstrated by immunofluorescence imaging. Migration rates were quantified using circular wound closure assays and live-cell tracking. AqB011 and bacopaside II, applied in combination, produced greater inhibitory effects on cell migration than did either agent alone. The high efficacy of AqB011 alone and in combination with bacopaside II in slowing HT29 cell motility correlated with abundant membrane localization of AQP1 protein. In SW480, neither agent alone was effective in blocking cell motility; however, combined application did cause inhibition of motility, consistent with low levels of membrane AQP1 expression. Bacopaside alone or combined with AqB011 also significantly impaired lamellipodial formation in both cell lines. Knockdown of AQP1 with siRNA (confirmed by quantitative PCR) reduced the effectiveness of the combined inhibitors, confirming AQP1 as a target of action. Invasiveness measured using transwell filters layered with extracellular matrix in both cell lines was inhibited by AqB011, with a greater potency in HT29 than SW480. A side effect of bacopaside II at high doses was a potentiation of invasiveness, that was reversed by AqB011. Results here are the first to demonstrate that combined block of the AQP1 ion channel and water pores is more potent in impairing motility across diverse classes of colon cancer cells than single agents alone.
Collapse
Affiliation(s)
- Michael L De Ieso
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Jinxin V Pei
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Saeed Nourmohammadi
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Eric Smith
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5005, Australia
- Oncology Department, The Basil Hetzel Institute, The Queen Elizabeth Hospital, Woodville, SA, 5011, Australia
| | - Pak Hin Chow
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Mohamad Kourghi
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Jennifer E Hardingham
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5005, Australia
- Oncology Department, The Basil Hetzel Institute, The Queen Elizabeth Hospital, Woodville, SA, 5011, Australia
| | - Andrea J Yool
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5005, Australia.
| |
Collapse
|
18
|
Ozu M, Galizia L, Acuña C, Amodeo G. Aquaporins: More Than Functional Monomers in a Tetrameric Arrangement. Cells 2018; 7:E209. [PMID: 30423856 PMCID: PMC6262540 DOI: 10.3390/cells7110209] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 10/27/2018] [Accepted: 11/07/2018] [Indexed: 12/11/2022] Open
Abstract
Aquaporins (AQPs) function as tetrameric structures in which each monomer has its own permeable pathway. The combination of structural biology, molecular dynamics simulations, and experimental approaches has contributed to improve our knowledge of how protein conformational changes can challenge its transport capacity, rapidly altering the membrane permeability. This review is focused on evidence that highlights the functional relationship between the monomers and the tetramer. In this sense, we address AQP permeation capacity as well as regulatory mechanisms that affect the monomer, the tetramer, or tetramers combined in complex structures. We therefore explore: (i) water permeation and recent evidence on ion permeation, including the permeation pathway controversy-each monomer versus the central pore of the tetramer-and (ii) regulatory mechanisms that cannot be attributed to independent monomers. In particular, we discuss channel gating and AQPs that sense membrane tension. For the latter we propose a possible mechanism that includes the monomer (slight changes of pore shape, the number of possible H-bonds between water molecules and pore-lining residues) and the tetramer (interactions among monomers and a positive cooperative effect).
Collapse
Affiliation(s)
- Marcelo Ozu
- Departamento de Biodiversidad y Biología Experimental, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina.
- Instituto de Biodiversidad y Biología Experimental y Aplicada (IBBEA), Universidad de Buenos Aires y Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1428EGA CABA, Argentina.
| | - Luciano Galizia
- Instituto de investigaciones Médicas A. Lanari, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C1427ARO, Argentina.
- Consejo Nacional de Investigaciones Científicas y Técnicas, Laboratorio de Canales Iónicos, Instituto de Investigaciones Médicas (IDIM), Universidad de Buenos Aires, Buenos Aires C1427ARO, Argentina.
| | - Cynthia Acuña
- Departamento de Biodiversidad y Biología Experimental, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina.
- Instituto de Biodiversidad y Biología Experimental y Aplicada (IBBEA), Universidad de Buenos Aires y Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1428EGA CABA, Argentina.
| | - Gabriela Amodeo
- Departamento de Biodiversidad y Biología Experimental, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina.
- Instituto de Biodiversidad y Biología Experimental y Aplicada (IBBEA), Universidad de Buenos Aires y Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1428EGA CABA, Argentina.
| |
Collapse
|
19
|
De Ieso ML, Yool AJ. Mechanisms of Aquaporin-Facilitated Cancer Invasion and Metastasis. Front Chem 2018; 6:135. [PMID: 29922644 PMCID: PMC5996923 DOI: 10.3389/fchem.2018.00135] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 04/09/2018] [Indexed: 01/02/2023] Open
Abstract
Cancer is a leading cause of death worldwide, and its incidence is rising with numbers expected to increase 70% in the next two decades. The fact that current mainline treatments for cancer patients are accompanied by debilitating side effects prompts a growing demand for new therapies that not only inhibit growth and proliferation of cancer cells, but also control invasion and metastasis. One class of targets gaining international attention is the aquaporins, a family of membrane-spanning water channels with diverse physiological functions and extensive tissue-specific distributions in humans. Aquaporins−1,−2,−3,−4,−5,−8, and−9 have been linked to roles in cancer invasion, and metastasis, but their mechanisms of action remain to be fully defined. Aquaporins are implicated in the metastatic cascade in processes of angiogenesis, cellular dissociation, migration, and invasion. Cancer invasion and metastasis are proposed to be potentiated by aquaporins in boosting tumor angiogenesis, enhancing cell volume regulation, regulating cell-cell and cell-matrix adhesions, interacting with actin cytoskeleton, regulating proteases and extracellular-matrix degrading molecules, contributing to the regulation of epithelial-mesenchymal transitions, and interacting with signaling pathways enabling motility and invasion. Pharmacological modulators of aquaporin channels are being identified and tested for therapeutic potential, including compounds derived from loop diuretics, metal-containing organic compounds, plant natural products, and other small molecules. Further studies on aquaporin-dependent functions in cancer metastasis are needed to define the differential contributions of different classes of aquaporin channels to regulation of fluid balance, cell volume, small solute transport, signal transduction, their possible relevance as rate limiting steps, and potential values as therapeutic targets for invasion and metastasis.
Collapse
Affiliation(s)
- Michael L De Ieso
- Department of Physiology, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Andrea J Yool
- Department of Physiology, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|