Aquaporin: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Contributor:

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 enabling the transmembrane diffusion of small neutral solutes (such as urea, glycerol, and hydrogen peroxide) and ions. AQPs are expressed throughout the human body.

  • aquaporin (AQP)
  • membranes
  • water
  • fluid
  • secretion

1. Introduction

Aquaporins (AQPs) have diverse roles in mammals, ranging from fluid homeostasis, glandular secretions, barrier function, immunity and inflammation, cell migration, and angiogenesis to signal transduction and sensation. It is now clear that AQP functions are more complex than simply mediating the passive flow of water across biological membranes [1]. Understanding their underlying regulatory mechanisms along with the discovery and development of small-molecule AQP inhibitors for use in research and therapeutic development is expected to lead to new insights into the basic biology of and novel treatments for the wide range of AQP-associated disorders.

2. Distribution and Classification of AQPs in the Human Body

The essential role of membrane intrinsic protein channels in the regulation of water transport and homeostasis was discovered in 1986 [2][3]. The molecular characterization of the archetypal water channel protein, aquaporin-1 (AQP1), launched the research field in the early 1990s with the cloning and characterization of CHIP28 (later renamed AQP1 [4][5]). Recognizing earlier work, the lens major intrinsic protein (MIP), cloned in 1984 [6], was renamed AQP0 on the basis that it formed ion channels in lipid bilayers [7] and transported water, albeit at a considerably slower rate than AQP1 [8]. Additional members of the mammalian AQP family were cloned and characterized over the next decade; 13 paralogs have currently been identified in higher mammals, showing differential patterns of tissue and cell expression throughout the body (Figure 1). Their diverse functions include the transport of water, glycerol, gases, hydrogen peroxide (H2O2), ammonia, and ions [9][10][11][12].
Figure 1. AQP distribution in the human body. Expression of AQP paralogs in the (a) brain, (b) blood–brain barrier, (c) eye, (d) exocrine glands, (e) inner ear, (f) cardiovascular system, (g) spine, (h) heart, (i) respiratory tract (trachea and lung; inset showing alveoli), (j) skeletal muscle, (k) pancreas, (l) liver, (m) gastrointestinal tract, (n) kidney, (o) skin (inset showing adipose tissue), and (p) female as well as (q) male reproductive tracts. This summary is not comprehensive; minor AQP subtypes are omitted for clarity. Bold text is used to highlight the major AQPs studied in the selected tissues [12].
The traditional categorization of aquaporins (AQPs) into orthodox (water-selective) and glycerol-permeable subtypes, with a third poorly understood ‘super’ or ‘subcellular’ group, no longer encompasses the expanding appreciation of AQPs as multi-functional channels. A broad repertoire of permeable substrates is evident not only in mammalian classes but in AQP classes across the kingdoms of life [13]. In the traditional scheme, orthodox mammalian AQPs comprise AQP0, 1, 2, 4, 5, and 6. This group includes the classic water channels, but interestingly also includes all the mammalian AQPs known thus far to have dual roles as water and ion channels (which are AQP0, 1, and 6). No ion channel function has yet been detected for AQP4 and AQP5 [14], though it is possible that the key stimuli remain to be identified, a concept to be considered for any AQPs that appear to be nonfunctional in experimental assays. When a multiple-sequence alignment analysis is run for human AQPs combined with known non-mammalian AQP ion channels, AQP8 falls on a distant branch of the orthodox group alongside the soybean AQP channel nodulin 26, which conducts water, glycerol, ammonia, and ions [1][15][16]. Although AQP8 in other alignments has been assigned to the ‘super-aquaporin’ group with AQP11 and AQP12 [17], its capacity for conducting ammonia [18] presents an interesting similarity with nodulin 26.
AQP0 was the first mammalian water channel suggested to mediate an ion conductance, in addition to its function as a water channel [19][20]. AQP1′s function as a gated ion channel was proposed in 1996 [21], and has since been refined to clarify that AQP1 activation occurs via the direct binding of cyclic guanosine monophosphate (cGMP) [22][23][24]. The subsequent identification of other members of the AQP family as dual water and ion channels has added the mammalian water channel AQP6, the insect water channel Drosophila big brain (DmBIB), and plant membrane intrinsic proteins such as the Arabidopsis plasma membrane intrinsic protein (PIP2;1) to the list [14][25][26].
The aquaglyceroporin group (AQP3, 7, 9, and 10) captures well-characterized glycerol-permeable channels. The most-recently cloned AQPs from higher mammalian orders include AQP11 and 12 [27]. Initial tests found no water permeability for AQP11 expressed in Xenopus oocytes [28]; however, reconstituted in membrane vesicles, AQP11 showed a low but detectable water permeability that was sensitive to mercury [29]. At positions which correspond to the conserved arginine aromatic barrier in most AQPs, residues in AQP11 and 12 are uniquely hydrophobic, lacking both the aromatic and basic elements of this functionally important site. Although this should not be compatible with the transport of water, alternative barrier sites have been proposed [30].

3. Structural Biology of the AQP Family

Owing to the relative ease of crystallizing AQP proteins, the structural biology of the transmembrane domains of the AQP family is well-established. A conserved signature fold consists of six transmembrane helices. Two re-entrant helix-forming loops stack one on top of the other, with the family’s signature asparagine–proline–alanine (NPA) motif present in both helices at their interface (Figure 2A). In contrast to the transmembrane domains, little is known about the structures of the intracellular amino- and carboxy-termini of AQP proteins. These less-ordered regions are usually removed to facilitate crystallization. Complex formation by calmodulin (CaM) binding to the unstructured C-terminus of AQP4 causes it to adopt an α-helical conformation [31]. The short C-terminal domain of AQP2 exemplifies this flexibility; the C-termini in each monomer adopted four different conformations within the tetrameric unit cell in an X-ray crystal structure [32].
Figure 2. Structural biology of the AQP family. (A) The signature fold of the AQP family consists of six transmembrane helices and two helix-forming re-entrant loops containing the signature NPA motif. (B,C) Water transport and selectivity is facilitated by the NPA motifs (green) found at the interface of the two helical re-entrant loops (red) and the aromatic/arginine selectivity filter (blue). Water molecules (a single water oxygen at the selectivity filter is indicated by a purple sphere) traverse the pore in single-file. (DF) The central pore formed at the fourfold axis of AQP1 contains two rings of bulky hydrophobic residues (orange) that prevent pore hydration in the absence of a cGMP signal. cGMP binding at loop D (green) activates the ion channel. Created with Biorender.com.
Following a pattern observed for cyclic-nucleotide-gated channels and potassium channels [33], AQP tetramers have a central pore at the four-fold axis of symmetry [34][35][36], which remains incompletely characterized for most AQP classes. Pharmacological and functional analyses have shown in AQP1 that ion and water transport occurs through independent parallel pathways [11].
Despite the ubiquity of tetramerization in the AQP family, the water pores reside and function within monomers, as evidenced by AQP structures in which permeable substrates have been co-crystallized in the intrasubunit pore in single-file [34][37][38][39]. AQP1 fusion proteins, linking one functional and one non-water-conducting monomer into dimers, were used to demonstrate that each subunit contains an independent water pore pathway [40]. In human AQP4, mutations in the loop D domain were shown to reduce oligomerization, impairing membrane trafficking and its responsiveness to osmotic stimuli [41].
The molecular mechanisms of ion permeation and gating differ between AQP classes. In AQP1, cGMP-activated cation currents are thought to flow through the central pore [36]. Molecular dynamic simulations and mutagenesis revealed that cGMP interacts with an arginine-rich region in loop D, causing loop displacement and conformational changes, and widening the central pore to enable hydration and then ion permeation [24][42][43]. In the closed central pore, hydrophobic barriers restrict ion permeation but could allow the permeation of gases, such as CO2 [44]. The loop D domain of AQP1, modeled as the gate for cGMP-dependent ion-channel-opening, might interact with the C-terminus, which modulates activation rates [22][23][42][43][45].
Amino acid sequence similarities between the AQP1 C-terminus and other known cGMP-interacting proteins fit a proposed modulatory role for the C-terminal domain [45]. The activation of ion conductance is impaired in AQP1 channels with truncated or mutated C-termini [46]. In contrast, in AQP6 the Hg2+-inducible permeation pathways for anions appear to reside in the intrasubunit (monomeric) pores, based on the Hg2+-sensitive cysteine locations and the lack of allostery in Hg2+-induced activation [47]. AQP6 channel properties are affected by the conformational flexibility of transmembrane helices involving conserved glycines at the crossing point of transmembrane domains TM2 and TM5; the mutation of a glutamine uniquely present in rat AQP6 into a glycine typical of most AQPs (N60G) abolished anion conduction while increasing water permeability [48]. The molecular basis of ion transport through AQP0 remains unexplored.

4. AQP Permeabilities: An Expanding Repertoire

In contrast to initial expectations, AQPs have turned out to be more than simple water channels, and display properties of a diverse multifunctional protein family that is still not fully characterized. The spectrum of roles now recognized for AQPs include water, glycerol, urea, ammonia, nitric oxide, and H2O2 transport; ion conductance; direct mediation of cell–cell adhesion; and regulation of the plasma membrane abundance of other membrane proteins (Table 1). Emerging permeability properties do not segregate neatly into the traditional orthodox and aquaglyceroporin classification scheme. Classes of AQPs that facilitate H2O2 transport, described as peroxiporins, include AQP1 [49], AQP3 [50], AQP5 [51], AQP8 [50][52][53], and AQP9 [54], although earlier work on AQP1 did not find peroxiporin activity [55]. AQP8 is expressed in the inner membrane of mitochondria and involved in H2O2 transport linked with the accumulation of reactive oxygen species (ROS) [52], revealing an unexpected breadth of physiologically important roles for AQPs across phyla [13][56] and highlighting many gaps that are yet to be addressed. Recent perspectives have provided comprehensive synopses of AQP-related diseases [57][58][59][60][61][62][63][64][65][66][67][68].
Table 1. AQP classification with permeant substrates and main sites of expression. Chromosome location and water permeability data adapted from [69]. Asterisks (*) highlight permeability results that are controversial or not yet accepted as readily reproducible.

Aquaporin

Chromosome

Water Permeability (Pf)

[×10−14 cm3 s−1]

Permeability to Molecules Other Than Water

Main Expression Sites

Orthodox (classical) AQPs

AQP0

12q13

0.25

Ions [19][20]

Eye lens

AQP1

7p14

6.0

Monovalent cations [24][36][42], nitric oxide [70], H2O2 [49][55], and glycerol * [71]

Central nervous system (CNS), inner ear, eye, kidney, endothelium, lung, skeletal muscle, cartilage, and erythrocytes

AQP2

12q13

3.3

None known

Kidney, inner ear, and reproductive tract

AQP4

18q22

24

Nitric oxide [72]

CNS, inner ear, retina, kidney, gastrointestinal tract (GIT), lung, and skeletal muscle

AQP5

12q13

5.0

H2O2 [51]

Secretory glands, inner ear, eye, kidney, GIT, and lung

AQP6

12q13

Low; no quantitative data

Ammonia [73], glycerol, urea [74], nitrate [75], and anions (NO3, Cl) [76]

Inner ear, kidney

AQP8

16p12

No quantitative data

Urea, ammonia, and H2O2 [77]

Liver, kidney, adipose tissue, pancreas, GIT, and reproductive tract

Aquaglyceroporins

AQP3

9p13

2.1

Glycerol [78], H2O2 [9], urea * [78], and ammonia [79]

Skin, inner ear, eye, adipose tissue, kidney, GIT, heart, lung, reproductive tract, and cartilage

AQP7

9p13

No quantitative data

Arsenite [80], glyerol and urea [81], and ammonia [82]

Adipose tissue, pancreas, liver, kidney, inner ear, GIT, heart, reproductive tract

AQP9

15q22

No quantitative data

Arsenite [80], carbamides, polyols, purines, pyrimidines [83], ketone bodies [84], lactate [85], ammonia [86], glycerol, urea [83][87][88], and H2O2 [54]

Liver, adipose tissue, CNS (unclear for humans), inner ear, and reproductive tract

AQP10

1q21

No quantitative data

Glycerol [89]

Adipose tissue and reproductive tract

Unorthodox AQPs/S-aquaporins

AQP11

11q13

~2

Glycerol [29][90][91]

Retina, kidney, GIT, and reproductive tract

AQP12

2q37

No quantitative data

Unknown

Pancreas

The diverse array of AQP functions in human physiology and pathophysiology has been divided into four overarching themes. These are: fluid homeostasis and secretion; signal transduction and sensory functions; defense and metabolism; and motility and cancer (Figure 3).
Figure 3. Functional roles of AQPs. (A) Fluid homeostasis and secretion: In the kidney, AQP1 regulates water reabsorption in the proximal tubules, while AQP2–4 are involved in urine concentration. In the central nervous system (CNS), AQP1 is involved in cerebrospinal fluid (CSF) production in the choroid plexus. In the lungs, AQPs facilitate transendothelial and transepithelial water flow. (B) Signal transduction and sensor function: AQP4 is involved in skeletal muscle contraction and viability. In the spinal cord, AQP1 is thought to contribute to pain processing and promote axonal growth as well as the regeneration of dorsal root ganglia (DRG). In the inner ear, AQPs are involved in balance and hearing. In the eye, AQP0 facilitates the structural integrity and transparency of the lens. (C) Defense, protection, and support: AQP4 is involved in blood–brain barrier (BBB) integrity, astrocyte plasticity, glial scar formation, and cerebral waste clearance. AQP3 supports skin hydration and wound healing. AQP1, 3, 5, 7, and 9 are involved in immune cell activation and pathogen elimination (phagocytosis). AQP7, 9, and 10 are involved in the glycerol transport that supports energy metabolism. (D) Cell motility: AQP1, 4, 5, and 9 are polarized at the leading edge of migrating cells and are thought to promote the cellular migration stages of polarization, protrusion, adhesion, and retraction. Additionally, AQP1, 3, 4, and 9 are assumed to enhance the degradation of the extracellular matrix (ECM). Created with BioRender.com.

This entry is adapted from the peer-reviewed paper 10.3390/ijms23031388

References

  1. Salman, M.M.; Kitchen, P.; Yool, A.J.; Bill, R.M. Recent breakthroughs and future directions in drugging aquaporins. Trends Pharm. Sci. 2022, 43, 30–42.
  2. Benga, G.; Popescu, O.; Borza, V.; Pop, V.I.; Muresan, A.; Mocsy, I.; Brain, A.; Wrigglesworth, J.M. Water permeability in human erythrocytes: Identification of membrane proteins involved in water transport. Eur. J. Cell Biol. 1986, 41, 252–262.
  3. Benga, G.; Popescu, O.; Pop, V.I.; Holmes, R.P. p-(Chloromercuri)benzenesulfonate binding by membrane proteins and the inhibition of water transport in human erythrocytes. Biochemistry 1986, 25, 1535–1538.
  4. Preston, G.M.; Agre, P. Isolation of the cDNA for erythrocyte integral membrane protein of 28 kilodaltons: Member of an ancient channel family. Proc. Natl. Acad. Sci. USA 1991, 88, 11110–11114.
  5. Preston, G.M.; Carroll, T.P.; Guggino, W.B.; Agre, P. Appearance of water channels in Xenopus oocytes expressing red cell CHIP28 protein. Science 1992, 256, 385–387.
  6. Gorin, M.B.; Yancey, S.B.; Cline, J.; Revel, J.P.; Horwitz, J. The major intrinsic protein (MIP) of the bovine lens fiber membrane: Characterization and structure based on cDNA cloning. Cell 1984, 39, 49–59.
  7. Zampighi, G.A.; Hall, J.E.; Kreman, M. Purified lens junctional protein forms channels in planar lipid films. Proc. Natl. Acad. Sci. USA 1985, 82, 8468–8472.
  8. Chandy, G.; Zampighi, G.A.; Kreman, M.; Hall, J.E. Comparison of the water transporting properties of MIP and AQP1. J. Membr. Biol. 1997, 159, 29–39.
  9. Bienert, G.P.; Chaumont, F. Aquaporin-facilitated transmembrane diffusion of hydrogen peroxide. Biochim. Biophys. Acta 2014, 1840, 1596–1604.
  10. Madeira, A.; Moura, T.F.; Soveral, G. Aquaglyceroporins: Implications in adipose biology and obesity. Cell. Mol. Life Sci. 2015, 72, 759–771.
  11. Yool, A.J.; Campbell, E.M. Structure, function and translational relevance of aquaporin dual water and ion channels. Mol. Aspects Med. 2012, 33, 553–561.
  12. Day, R.E.; Kitchen, P.; Owen, D.S.; Bland, C.; Marshall, L.; Conner, A.C.; Bill, R.M.; Conner, M.T. Human aquaporins: Regulators of transcellular water flow. Biochim. Biophys. Acta 2014, 1840, 1492–1506.
  13. Tyerman, S.D.; McGaughey, S.A.; Qiu, J.; Yool, A.J.; Byrt, C.S. Adaptable and Multifunctional Ion-Conducting Aquaporins. Annu. Rev. Plant Biol. 2021, 72, 703–736.
  14. Kourghi, M.; Nourmohammadi, S.; Pei, J.V.; Qiu, J.; McGaughey, S.; Tyerman, S.D.; Byrt, C.S.; Yool, A.J. Divalent Cations Regulate the Ion Conductance Properties of Diverse Classes of Aquaporins. Int. J. Mol. Sci. 2017, 18, 2323.
  15. Weaver, C.D.; Shomer, N.H.; Louis, C.F.; Roberts, D.M. Nodulin 26, a nodule-specific symbiosome membrane protein from soybean, is an ion channel. J. Biol. Chem. 1994, 269, 17858–17862.
  16. Hwang, J.H.; Ellingson, S.R.; Roberts, D.M. Ammonia permeability of the soybean nodulin 26 channel. FEBS Lett. 2010, 584, 4339–4343.
  17. Gonen, T.; Walz, T. The structure of aquaporins. Q. Rev. Biophys. 2006, 39, 361–396.
  18. Krenc, D.; Song, J.; Almasalmeh, A.; Wu, B.; Beitz, E. The arginine-facing amino acid residue of the rat aquaporin 1 constriction determines solute selectivity according to its size and lipophilicity. Mol. Membr. Biol. 2014, 31, 228–238.
  19. Kushmerick, C.; Rice, S.J.; Baldo, G.J.; Haspel, H.C.; Mathias, R.T. Ion, water and neutral solute transport in Xenopus oocytes expressing frog lens MIP. Exp. Eye Res. 1995, 61, 351–362.
  20. Ehring, G.R.; Zampighi, G.; Horwitz, J.; Bok, D.; Hall, J.E. Properties of channels reconstituted from the major intrinsic protein of lens fiber membranes. J. Gen. Physiol. 1990, 96, 631–664.
  21. Yool, A.J.; Stamer, W.D.; Regan, J.W. Forskolin stimulation of water and cation permeability in aquaporin 1 water channels. Science 1996, 273, 1216–1218.
  22. Saparov, S.M.; Kozono, D.; Rothe, U.; Agre, P.; Pohl, P. Water and ion permeation of aquaporin-1 in planar lipid bilayers. Major differences in structural determinants and stoichiometry. J. Biol. Chem. 2001, 276, 31515–31520.
  23. Anthony, T.L.; Brooks, H.L.; Boassa, D.; Leonov, S.; Yanochko, G.M.; Regan, J.W.; Yool, A.J. Cloned human aquaporin-1 is a cyclic GMP-gated ion channel. Mol. Pharmacol. 2000, 57, 576–588.
  24. Campbell, E.M.; Birdsell, D.N.; Yool, A.J. The activity of human aquaporin 1 as a cGMP-gated cation channel is regulated by tyrosine phosphorylation in the carboxyl-terminal domain. Mol. Pharmacol. 2012, 81, 97–105.
  25. Yanochko, G.M.; Yool, A.J. Regulated cationic channel function in Xenopus oocytes expressing Drosophila big brain. J. Neurosci. 2002, 22, 2530–2540.
  26. Yasui, M.; Hazama, A.; Kwon, T.H.; Nielsen, S.; Guggino, W.B.; Agre, P. Rapid gating and anion permeability of an intracellular aquaporin. Nature 1999, 402, 184–187.
  27. Ishibashi, K.; Tanaka, Y.; Morishita, Y. The role of mammalian superaquaporins inside the cell. Biochim. Biophys. Acta 2014, 1840, 1507–1512.
  28. Gorelick, D.A.; Praetorius, J.; Tsunenari, T.; Nielsen, S.; Agre, P. Aquaporin-11: A channel protein lacking apparent transport function expressed in brain. BMC Biochem. 2006, 7, 14.
  29. Yakata, K.; Hiroaki, Y.; Ishibashi, K.; Sohara, E.; Sasaki, S.; Mitsuoka, K.; Fujiyoshi, Y. Aquaporin-11 containing a divergent NPA motif has normal water channel activity. Biochim. Biophys. Acta 2007, 1768, 688–693.
  30. Calvanese, L.; Pellegrini-Calace, M.; Oliva, R. In silico study of human aquaporin AQP11 and AQP12 channels. Protein Sci. 2013, 22, 455–466.
  31. Kitchen, P.; Salman, M.M.; Halsey, A.M.; Clarke-Bland, C.; MacDonald, J.A.; Ishida, H.; Vogel, H.J.; Almutiri, S.; Logan, A.; Kreida, S.; et al. Targeting Aquaporin-4 Subcellular Localization to Treat Central Nervous System Edema. Cell 2020, 181, 784–799.
  32. Frick, A.; Eriksson, U.K.; de Mattia, F.; Oberg, F.; Hedfalk, K.; Neutze, R.; de Grip, W.J.; Deen, P.M.; Tornroth-Horsefield, S. X-ray structure of human aquaporin 2 and its implications for nephrogenic diabetes insipidus and trafficking. Proc. Natl. Acad. Sci. USA 2014, 111, 6305–6310.
  33. Jan, L.Y.; Jan, Y.N. Structural Elements Involved in Specific K+ Channel Functions. Annu. Rev. Physiol. 1992, 54, 537–555.
  34. 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.
  35. Walz, T.; Hirai, T.; Murata, K.; Heymann, J.B.; Mitsuoka, K.; Fujiyoshi, Y.; Smith, B.L.; Agre, P.; Engel, A. The three-dimensional structure of aquaporin-1. Nature 1997, 387, 624–627.
  36. Yool, A.J.; Weinstein, A.M. New roles for old holes: Ion channel function in aquaporin-1. News Physiol. Sci. 2002, 17, 68–72.
  37. Sui, H.; Han, B.G.; Lee, J.K.; Walian, P.; Jap, B.K. Structural basis of water-specific transport through the AQP1 water channel. Nature 2001, 414, 872–878.
  38. Fu, D.; Libson, A.; Miercke, L.J.; Weitzman, C.; Nollert, P.; Krucinski, J.; Stroud, R.M. Structure of a glycerol-conducting channel and the basis for its selectivity. Science 2000, 290, 481–486.
  39. Ho, J.D.; Yeh, R.; Sandstrom, A.; Chorny, I.; Harries, W.E.; Robbins, R.A.; Miercke, L.J.; Stroud, R.M. Crystal structure of human aquaporin 4 at 1.8 A and its mechanism of conductance. Proc. Natl. Acad. Sci. USA 2009, 106, 7437–7442.
  40. Mathai, J.C.; Agre, P. Hourglass pore-forming domains restrict aquaporin-1 tetramer assembly. Biochemistry 1999, 38, 923–928.
  41. Kitchen, P.; Conner, M.T.; Bill, R.M.; Conner, A.C. Structural Determinants of Oligomerization of the Aquaporin-4 Channel. J. Biol. Chem. 2016, 291, 6858–6871.
  42. Yu, J.; Yool, A.J.; Schulten, K.; Tajkhorshid, E. Mechanism of gating and ion conductivity of a possible tetrameric pore in aquaporin-1. Structure 2006, 14, 1411–1423.
  43. Kourghi, M.; De Ieso, M.L.; Nourmohammadi, S.; Pei, J.V.; Yool, A.J. Identification of Loop D Domain Amino Acids in the Human Aquaporin-1 Channel Involved in Activation of the Ionic Conductance and Inhibition by AqB011. Front. Chem. 2018, 6, 142.
  44. Endeward, V.; Musa-Aziz, R.; Cooper, G.J.; Chen, L.M.; Pelletier, M.F.; Virkki, L.V.; Supuran, C.T.; King, L.S.; Boron, W.F.; Gros, G. Evidence that aquaporin 1 is a major pathway for CO2 transport across the human erythrocyte membrane. FASEB J. 2006, 20, 1974–1981.
  45. Boassa, D.; Yool, A.J. A fascinating tail: cGMP activation of aquaporin-1 ion channels. Trends Pharmacol. Sci. 2002, 23, 558–562.
  46. Boassa, D.; Yool, A.J. Single amino acids in the carboxyl terminal domain of aquaporin-1 contribute to cGMP-dependent ion channel activation. BMC Physiol. 2003, 3, 12.
  47. Hazama, A.; Kozono, D.; Guggino, W.B.; Agre, P.; Yasui, M. Ion permeation of AQP6 water channel protein. Single channel recordings after Hg2+ activation. J. Biol. Chem. 2002, 277, 29224–29230.
  48. Liu, K.; Kozono, D.; Kato, Y.; Agre, P.; Hazama, A.; Yasui, M. Conversion of aquaporin 6 from an anion channel to a water-selective channel by a single amino acid substitution. Proc. Natl. Acad. Sci. USA 2005, 102, 2192–2197.
  49. 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.
  50. Miller, E.W.; Dickinson, B.C.; Chang, C.J. Aquaporin-3 mediates hydrogen peroxide uptake to regulate downstream intracellular signaling. Proc. Natl. Acad. Sci. USA 2010, 107, 15681–15686.
  51. Rodrigues, C.; Pimpão, C.; Mósca, 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.
  52. Marchissio, M.J.; Francés, D.E.; Carnovale, C.E.; Marinelli, R.A. Mitochondrial aquaporin-8 knockdown in human hepatoma HepG2 cells causes ROS-induced mitochondrial depolarization and loss of viability. Toxicol Appl. Pharm. 2012, 264, 246–254.
  53. Bertolotti, M.; Bestetti, S.; García-Manteiga, J.M.; Medraño-Fernandez, I.; Dal Mas, A.; Malosio, M.L.; Sitia, R. Tyrosine kinase signal modulation: A matter of H2O2 membrane permeability? Antioxid Redox Signal 2013, 19, 1447–1451.
  54. Watanabe, S.; Moniaga, C.S.; Nielsen, S.; Hara-Chikuma, M. Aquaporin-9 facilitates membrane transport of hydrogen peroxide in mammalian cells. Biochem. Biophys. Res. Commun. 2016, 471, 191–197.
  55. Almasalmeh, A.; Krenc, D.; Wu, B.; Beitz, E. Structural determinants of the hydrogen peroxide permeability of aquaporins. FEBS J. 2014, 281, 647–656.
  56. Kitchen, P.; Day, R.E.; Salman, M.M.; Conner, M.T.; Bill, R.M.; Conner, A.C. Beyond water homeostasis: Diverse functional roles of mammalian aquaporins. Biochim. Biophys. Acta 2015, 1850, 2410–2421.
  57. Lv, H.; Li, Y.; Xue, C.; Dong, N.; Bi, C.; Shan, A. Aquaporin: Targets for dietary nutrients to regulate intestinal health. J. Anim. Physiol. Anim. Nutr. 2021, 106, 167–180.
  58. Olesen, E.T.B.; Fenton, R.A. Aquaporin 2 regulation: Implications for water balance and polycystic kidney diseases. Nat. Rev. Nephrol. 2021, 17, 765–781.
  59. Tardelli, M.; Stulnig, T.M. Aquaporin regulation in metabolic organs. Vitam. Horm. 2020, 112, 71–93.
  60. Bollag, W.B.; Aitkens, L.; White, J.; Hyndman, K.A. Aquaporin-3 in the epidermis: More than skin deep. Am. J. Physiol. Cell Physiol. 2020, 318, C1144–C1153.
  61. Mogensen, F.L.; Delle, C.; Nedergaard, M. The Glymphatic System (En)during Inflammation. Int. J. Mol. Sci. 2021, 22, 7491.
  62. Noda, Y.; Sasaki, S. Updates and Perspectives on Aquaporin-2 and Water Balance Disorders. Int. J. Mol. Sci. 2021, 22, 12950.
  63. Valenti, G.; Tamma, G. The vasopressin-aquaporin-2 pathway syndromes. Handb. Clin. Neurol. 2021, 181, 249–259.
  64. Salman, M.M.; Kitchen, P.; Iliff, J.J.; Bill, R.M. Aquaporin 4 and glymphatic flow have central roles in brain fluid homeostasis. Nat. Rev. Neurosci. 2021, 22, 650–651.
  65. Salman, M.M.; Kitchen, P.; Halsey, A.; Wang, M.X.; Tornroth-Horsefield, S.; Conner, A.C.; Badaut, J.; Iliff, J.J.; Bill, R.M. Emerging roles for dynamic aquaporin-4 subcellular relocalization in CNS water homeostasis. Brain 2021.
  66. Markou, A.; Unger, L.; Abir-Awan, M.; Saadallah, A.; Halsey, A.; Balklava, Z.; Conner, M.; Törnroth-Horsefield, S.; Greenhill, S.D.; Conner, A.; et al. Molecular mechanisms governing aquaporin relocalisation. Biochim. Biophys. Acta Biomembr. 2021, 1864, 183853.
  67. De Ieso, M.L.; Yool, A.J. Mechanisms of Aquaporin-Facilitated Cancer Invasion and Metastasis. Front. Chem. 2018, 6, 135.
  68. Yool, A.J.; Ramesh, S. Molecular Targets for Combined Therapeutic Strategies to Limit Glioblastoma Cell Migration and Invasion. Front. Pharm. 2020, 11, 358.
  69. Castle, N. Aquaporins as targets for drug discovery. Drug Discov. Today 2005, 10, 485–493.
  70. Herrera, M.; Hong, N.J.; Garvin, J.L. Aquaporin-1 transports NO across cell membranes. Hypertension 2006, 48, 157–164.
  71. Abrami, L.; Tacnet, F.; Ripoche, P. Evidence for a glycerol pathway through aquaporin 1 (CHIP28) channels. Pflügers Archiv 1995, 430, 447–458.
  72. Wang, Y.; Tajkhorshid, E. Nitric oxide conduction by the brain aquaporin AQP4. Proteins 2010, 78, 661–670.
  73. Soria, L.R.; Fanelli, E.; Altamura, N.; Svelto, M.; Marinelli, R.A.; Calamita, G. Aquaporin-8-facilitated mitochondrial ammonia transport. Biochem. Biophys. Res. Commun. 2010, 393, 217–221.
  74. Holm, L.M.; Klaerke, D.A.; Zeuthen, T. Aquaporin 6 is permeable to glycerol and urea. Pflügers Archiv 2004, 448, 181–186.
  75. Ikeda, M.; Beitz, E.; Kozono, D.; Guggino, W.B.; Agre, P.; Yasui, M. Characterization of aquaporin-6 as a nitrate channel in mammalian cells. Requirement of pore-lining residue threonine 63. J. Biol. Chem. 2002, 277, 39873–39879.
  76. Rambow, J.; Wu, B.; Rönfeldt, D.; Beitz, E. Aquaporins with anion/monocarboxylate permeability: Mechanisms, relevance for pathogenic “host interactions. Front. Pharmacol. 2014, 5, 199.
  77. Chauvigné, F.; Yilmaz, O.; Ferré, A.; Fjelldal, P.G.; Finn, R.N.; Cerdà, J. The vertebrate Aqp14 water channel is a neuropeptide-regulated polytransporter. Commun. Biol. 2019, 2, 462.
  78. Ishibashi, K.; Sasaki, S.; Fushimi, K.; Uchida, S.; Kuwahara, M.; Saito, H.; Furukawa, T.; Nakajima, K.; Yamaguchi, Y.; Gojobori, T.; et al. Molecular cloning and expression of a member of the aquaporin family with permeability to glycerol and urea in addition to water expressed at the basolateral membrane of kidney collecting duct cells. Proc. Natl. Acad. Sci. USA 1994, 91, 6269–6273.
  79. Soveral, G.; Nielsen, S.; Casini, A. Aquaporins in Health and Disease: New Molecular Targets for Drug Discovery; Taylor Francis Group (CRC Press): Boca Raton, FL, USA, 2016.
  80. Liu, Z.; Shen, J.; Carbrey, J.M.; Mukhopadhyay, R.; Agre, P.; Rosen, B.P. Arsenite transport by mammalian aquaglyceroporins AQP7 and AQP9. Proc. Natl. Acad. Sci. USA 2002, 99, 6053–6058.
  81. Ishibashi, K.; Kuwahara, M.; Gu, Y.; Kageyama, Y.; Tohsaka, A.; Suzuki, F.; Marumo, F.; Sasaki, S. Cloning and functional expression of a new water channel abundantly expressed in the testis permeable to water, glycerol, and urea. J. Biol. Chem. 1997, 272, 20782–20786.
  82. Geyer, R.R.; Musa-Aziz, R.; Qin, X.; Boron, W.F. Relative CO(2)/NH(3) selectivities of mammalian aquaporins 0-9. Am. J. Physiol. Cell Physiol. 2013, 304, C985–C994.
  83. Tsukaguchi, H.; Shayakul, C.; Berger, U.V.; Mackenzie, B.; Devidas, S.; Guggino, W.B.; van Hoek, A.N.; Hediger, M.A. Molecular characterization of a broad selectivity neutral solute channel. J. Biol. Chem. 1998, 273, 24737–24743.
  84. Elkjaer, M.; Vajda, Z.; Nejsum, L.N.; Kwon, T.; Jensen, U.B.; Amiry-Moghaddam, M.; Frøkiaer, J.; Nielsen, S. Immunolocalization of AQP9 in liver, epididymis, testis, spleen, and brain. Biochem. Biophys. Res. Commun. 2000, 276, 1118–1128.
  85. Akashi, A.; Miki, A.; Kanamori, A.; Nakamura, M. Aquaporin 9 expression is required for l-lactate to maintain retinal neuronal survival. Neurosci. Lett. 2015, 589, 185–190.
  86. Stahl, K.; Rahmani, S.; Prydz, A.; Skauli, N.; MacAulay, N.; Mylonakou, M.N.; Torp, R.; Skare, Ø.; Berg, T.; Leergaard, T.B.; et al. Targeted deletion of the aquaglyceroporin AQP9 is protective in a mouse model of Parkinson’s disease. PLoS ONE 2018, 13, e0194896.
  87. Ishibashi, K.; Kuwahara, M.; Gu, Y.; Tanaka, Y.; Marumo, F.; Sasaki, S. Cloning and functional expression of a new aquaporin (AQP9) abundantly expressed in the peripheral leukocytes permeable to water and urea, but not to glycerol. Biochem. Biophys. Res. Commun. 1998, 244, 268–274.
  88. Tsukaguchi, H.; Weremowicz, S.; Morton, C.C.; Hediger, M.A. Functional and molecular characterization of the human neutral solute channel aquaporin-9. Am. J. Physiol. 1999, 277, F685–F696.
  89. Laforenza, U.; Scaffino, M.F.; Gastaldi, G. Aquaporin-10 Represents an Alternative Pathway for Glycerol Efflux from Human Adipocytes. PLoS ONE 2013, 8, e54474.
  90. Madeira, A.; Fernández-Veledo, S.; Camps, M.; Zorzano, A.; Moura, T.F.; Ceperuelo-Mallafré, 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.
  91. Finn, R.N.; Cerdà, J. Evolution and functional diversity of aquaporins. Biol. Bull. 2015, 229, 6–23.
More
This entry is offline, you can click here to edit this entry!
Video Production Service