Next Article in Journal
Genome-Wide Identification and Characterization of the SBP Gene Family in Passion Fruit (Passiflora edulis Sims)
Next Article in Special Issue
Biallelic Variants in TULP1 Are Associated with Heterogeneous Phenotypes of Retinal Dystrophy
Previous Article in Journal
Serum Biomarkers of Renal Fibrosis: A Systematic Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Retinal Cyclic Nucleotide-Gated Channel Regulation by Calmodulin

Department of Chemistry, University of California, Davis, CA 95616, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(22), 14143; https://doi.org/10.3390/ijms232214143
Submission received: 25 October 2022 / Revised: 10 November 2022 / Accepted: 12 November 2022 / Published: 16 November 2022
(This article belongs to the Special Issue Molecular Basis of Sensory Transduction in Health and Disease 2.0)

Abstract

:
Retinal cyclic nucleotide-gated (CNG) ion channels bind to intracellular cGMP and mediate visual phototransduction in photoreceptor rod and cone cells. Retinal rod CNG channels form hetero-tetramers comprised of three CNGA1 and one CNGB1 protein subunits. Cone CNG channels are similar tetramers consisting of three CNGA3 and one CNGB3 subunits. Calmodulin (CaM) binds to two distinct sites (CaM1: residues 565–587 and CaM2: residues 1120–1147) within the cytosolic domains of rod CNGB1. The binding of Ca2+-bound CaM to CNGB1 promotes the Ca2+-induced desensitization of CNG channels in retinal rods that may be important for photoreceptor light adaptation. Mutations that affect Ca2+-dependent CNG channel function are responsible for inherited forms of blindness. In this review, we propose structural models of the rod CNG channel bound to CaM that suggest how CaM might cause channel desensitization and how dysregulation of the channel may lead to retinal disease.

1. Overview of Retinal CNG Channels

Cyclic nucleotide-gated channels [1,2] were first identified in the plasma membrane of vertebrate rod photoreceptor cells, where they serve a functional role in visual phototransduction [3]. CNG channels are cation-specific channels that are opened by the direct binding of intracellular cyclic nucleotides [4]. CNG channels consist of multiple protein subunits coded by six different genes grouped into two subtypes: CNGA and CNGB, as shown in Figure 1A [5]. CNGA1 from the bovine retina was the first gene to be cloned [6] and was originally called the α-subunit of the retinal rod CNG channel. CNGA1 is mainly expressed in rod photoreceptors and recombinant CNGA1 can be expressed as a functional homomeric channel that can be gated by cGMP [7,8]. Mutations in CNGA1 are genetically linked to a recessive form of retinitis pigmentosa [9]. The native rod CNG channel contains a second protein subunit, CNGB1, [10] originally called the β-subunit. The native rod CNG channel forms a hetero-tetramer composed of three CNGA1 and one CNGB1 subunits [11], as seen in recent atomic-level structures of the rod CNG channel [12,13,14]. The CNGA1 and CNGB1 structures (Figure 1B) are both comprised of an N-terminal cytosolic region (not visible in the cryo-EM), six transmembrane spanning helices (channel domain, CD) and an intracellular cGMP-binding domain (CNBD) flanked by a C-terminal leucine zipper domain (CLZ) in CNGA1 or a CaM-binding domain (CaM2) in CNGB1. The sixth transmembrane helix (S6) from each subunit forms the ion-conducting pathway (or channel pore). The CNBD is comprised of three helices and a β-roll containing eight β-strands. An 80-residue linker (C-linker) between the CNBD and the channel domain relays ligand-induced conformational changes to the S6 helix in the channel pore [15].
Cone photoreceptors express CNG channels that contain two different protein subunits, CNGA3 (cone α subunit) and CNGB3 (cone β subunit) [16,17] that are distinct from the protein subunits of the rod channel. Recombinant CNGA3 subunits expressed in non-native cells form functional homomeric channels that are gated by cGMP [16,18,19], similar to that seen for CNGA1 [8]. The native cone CNG channel forms a hetero-tetramer composed of three CNGA3 and one CNGB3 subunits, as seen in the recent cryo-EM structure [20]. Mutations in human CNGA3 and CNGB3 have been linked to achromatopsia (also called color blindness), which is an autosomal recessive congenital disorder characterized by the complete inability to discriminate between colors [21,22].
Figure 1. Structural overview of CNG channels. (A) Phylogenetic analysis of CNG channel subunits. (B) Cryo-EM structure of CNG channel [14] and ribbon diagrams of CNGA1 (blue) and CNGB1 (red) subunits. The structure of the CaM1 site (asterisks) was not visible in the cryo-EM structures and instead was modeled by AlphaFold [23].
Figure 1. Structural overview of CNG channels. (A) Phylogenetic analysis of CNG channel subunits. (B) Cryo-EM structure of CNG channel [14] and ribbon diagrams of CNGA1 (blue) and CNGB1 (red) subunits. The structure of the CaM1 site (asterisks) was not visible in the cryo-EM structures and instead was modeled by AlphaFold [23].
Ijms 23 14143 g001

2. Retinal CNG Channels Are Essential for Visual Phototransduction

Phototransduction in retinal rod photoreceptor cells is triggered by light-activation of the visual pigment rhodopsin (R*) that turns on an enzymatic cascade, leading to the hydrolysis of cGMP catalyzed by phosphodiesterase PDE6 [24,25,26,27] (Figure 2A). The cytosolic cGMP concentration is maintained at a high level in dark-adapted photoreceptors, which allows for CNG channels to remain open in the absence of light [26]. Photoreceptor CNG channels conduct a cationic current only when the cytosolic ligand-binding domain binds to intracellular cGMP (Figure 2B) [3,24]. The light-induced lowering of cytosolic cGMP (that occurs during phototransduction) causes the closure of CNG channels in the photoreceptor’s outer-segment plasma membrane that blocks the entry of Na+ and Ca2+ and generates a neural signal [28,29,30]. The light-induced CNG channel closure during phototransduction causes a decrease in the cytosolic Ca2+ concentration (500 nM in the dark [31] versus 50 nM in the light [32]) that, in turn, modulates CNG channel sensitivity [19,33,34] and activates guanylate cyclase [35,36,37]. CNG channels have the highest open probability at low cytosolic Ca2+ levels in light-activated photoreceptors and the channels become desensitized at elevated Ca2+ levels that exist in dark-adapted photoreceptors ([Ca2+]i = 500 nM [31]). The Ca2+-dependent desensitization of photoreceptor CNG channel activity is mediated by CaM, which is important for promoting light adaptation in rod photoreceptor cells [33,34,38,39]. The Ca2+-dependent CNG channel activity might be due to a Ca2+-induced lowering of the cGMP-binding affinity [19,40,41]. Ca2+-dependent CNG channel activity has also been reported in the cone photoreceptor [42,43,44,45].

3. Calmodulin Mediates Ca2+-Dependent CNG Channel Desensitization

Calmodulin (CaM) is a 16.7 kDa Ca2+ sensor protein that belongs to the EF-hand superfamily [46]. CaM contains four EF-hand Ca2+-binding motifs (EF1, EF2, EF3 and EF4) that form two separate domains (EF1 and EF2 form the CaM N-lobe, while EF3 and EF4 form the CaM C-lobe) [47]. The CaM C-lobe and N-lobe each bind to Ca2+ with a dissociation constant of ~1 μM and 10 μM, respectively [48], which allows for Ca2+ to bind to the C-lobe first before binding to the N-lobe. The Ca2+-saturated form of CaM binds to hundreds of different target proteins, including dozens of enzymes, receptors, ion channels and other Ca2+ transporters [49].
The Ca2+-dependent desensitization of rod CNG channels requires CaM binding to two separate sites within the cytosolic region of CNGB1 (residues 565–587 called CaM1 [7,19,41,50] and residues 1120–1147 called CaM2 [41] in Figure 3A). The CaM1 and CaM2 sites are both conserved in the cone CNGB3 (Figure 3A) and the cone CNG channel undergoes Ca2+-induced desensitization [42]. The Ca2+-dependent activity of cone channels may also be mediated by a novel Ca2+ sensor protein called CNG modulin [43,51]; however, more recent studies suggest that CNG modulin does not regulate CNG channels [52]. The N-terminal CaM1 site in the rod channel was originally identified to bind to Ca2+-bound CaM [41] (but not Ca2+-free CaM [53]) and CaM binding was suggested to inactivate rod CNG channel activity [19,54]. The CaM1 site (in the absence of CaM) was proposed to interact with a C-terminal domain in CNGA1, known as a C-terminal leucine zipper (CLZ) [11], that is immediately adjacent to the cyclic-nucleotide binding domain (Figure 1B). The binding of Ca2+-bound CaM to CaM1 was suggested to block CaM1 binding to the CLZ [7,11]. The N-terminal CaM1 site is not visible in the recent cryo-EM structures [12,13,14,15], suggesting that CaM1 is dynamically disordered or otherwise not visible in these structures. A recent NMR structure of CaM bound to a peptide fragment of CaM1 [53] reveals that the Ca2+-bound CaM N-lobe binds stably to CaM1 (Figure 3B), but the Ca2+-bound CaM C-lobe binds to CaM1 with much lower affinity, which prevents determination of its structure [53]. Thus, CaM1 binds functionally to only the CaM N-lobe, while the lower affinity CaM C-lobe binding may not be physiologically relevant. The CaM1 residue F575 makes extensive hydrophobic contacts with the Ca2+-bound CaM N-lobe (Figure 3B) and the mutation F575E can selectively disable the CaM N-lobe binding to CaM1 [53]. Future electrophysiology studies are needed to study the effect of the F575E mutation on Ca2+-dependent desensitization of CNG channels to test whether CaM1 is essential for CNG channel function.
A closer look at the cryo-EM structure of the rod CNG channel in the apo state [13] revealed faint electron density from CaM that is bound to a site in CNGB1 downstream of the CNBD, dubbed the D-helix (Figure 3C). The structure of CaM bound to the D-helix (Figure 3C) is very similar to the crystal structure of the CaM C-lobe bound to the creatine kinase peptide [55]. Moreover, the CaM/D-helix structure (Figure 3C) is essentially identical (within experimental error) to the very recent NMR structure of the CaM C-lobe bound to the CaM2 peptide (Figure 3D) [53]. Indeed, the CaM C-lobe alone can bind to the CaM2 peptide with nanomolar affinity, in contrast to the CaM N-lobe that binds to CaM2 with lower affinity [53]. The nanomolar binding of the CaM C-lobe to the CaM2 peptide explains in part how the CaM/CaM2 complex can bind to Ca2+ at the very low Ca2+ concentration (80 nM) used in the cryo-EM study [13]. The NMR structure of the Ca2+-bound CaM C-lobe bound to the CaM2 peptide (Figure 3D) reveals CaM2 residues L1132 and L1136 make extensive hydrophobic contact with the CaM C-lobe (Figure 3D), and the mutations (L1132E and L1136E) selectively disable CaM C-lobe binding to the CaM2 peptide [53]. Future electrophysiology studies are needed to study the effect of the L1132E/L1136E mutations on the Ca2+-dependent desensitization of CNG channels and to determine whether CaM binding to the CaM2 site is essential for CNG channel function.

4. Structural Models of CNG Channel Regulation by Calmodulin

4.1. Two-Site Model for CaM Regulation of the Rod CNG Channel

We propose two different structural models of Ca2+-dependent desensitization of CNG channels mediated by CaM: one-site versus two-site model (Figure 4A,B). The two-site model (Figure 4A), originally proposed by A. Bej and J.B. Ames [53], suggests that two separate CaM binding sites (CaM1 and CaM2 in Figure 3A) in CNGB1 bind selectively to each lobe of CaM: CaM1 binds to the CaM N-lobe and CaM2 binds to the CaM C-lobe. At high-cytosolic Ca2+ levels in the dark-adapted photoreceptor, we propose that a single CaM bridges the two CaM binding sites together to bring CaM1 and CaM2 in close proximity in the desensitized-channel state (Figure 4A, left panel). The Ca2+-induced bridging of CaM1 and CaM2 and their close proximity with the CLZ may somehow alter the structure of the adjacent CNBD to decrease the binding affinity of cGMP or otherwise reduce the channel open probability. By contrast, the low cytosolic Ca2+ level in the light-activated photoreceptor should cause Ca2+ dissociation from the CaM N-lobe, which is predicted to disable CaM N-lobe binding to CaM1 and, therefore, allow for CaM1 and CaM2 to move far apart when the channel switches to the highest open probability state (Figure 4A, right panel). In essence, the CaM N-lobe serves as a Ca2+ sensor, while the Ca2+-bound CaM C-lobe is proposed to remain constitutively anchored to CaM2 even at low Ca2+ levels in light-activated rods, because the apparent Ca2+ affinity of CaM bound to CaM2 is estimated to be ~60 nM [53]. This pre-anchoring of the Ca2+-bound CaM C-lobe to the sensitized CNG channel at low Ca2+ levels eliminates any diffusional barrier of Ca2+ sensing and is reminiscent of CaM pre-association with L-type voltage-gate Ca2+ channels [56,57]. The two-site model predicts that Ca2+ binding to the CaM C-lobe (EF3 and EF4) should be essential for anchoring the CaM C-lobe to the sensitized CNG channel (Figure 4A, right panel). Future experiments on the CaM mutant, CaM34 (that disables Ca2+ binding to the CaM C-lobe [58]) are needed to test whether Ca2+-binding to the CaM C-lobe is essential for CNG channel sensitization at low Ca2+ levels in light-activated photoreceptors. Another way to test the two-site model would be to assess the effect of CNG channel mutations F575E and L1132E/L1136E on the channel’s open probability and cGMP binding affinity. The two-site model (Figure 4A) predicts that the F575E mutation should disable CaM N-lobe binding, which should prevent Ca2+-induced channel desensitization and/or permit high-affinity cGMP binding regardless of the Ca2+ level. The L1132E/L1136E mutations should disable CaM binding to CaM2, which, in turn, might destabilize the sensitized channel state, causing a decrease in the channel open probability and perhaps a lower cGMP binding affinity. Future electrophysiology experiments on the F575E and L1132E/L1136E mutations are needed to test the predictions of the two-site model.

4.2. CaM Binding to CaM2 May Be Sufficient for CNG Channel Desensitization

A one-site model for Ca2+-induced CNG channel desensitization is proposed, in which CaM C-lobe binding to CaM2 alone may be sufficient to cause desensitization (Figure 4B). The CaM interaction with CaM1 is not considered here because the CaM1 peptide has relatively low affinity binding to the CaM N-lobe (Kd = 5 μM [53]), which may be outside the physiological range of cellular-free CaM concentration (100 nM [60]). This is in stark contrast to the CaM2 site that binds to CaM with a Kd in the nanomolar range [53]. Moreover, the CaM N-lobe and CaM1 site are both structurally disordered or otherwise not visible in the cryo-EM structures [12,13,14,15], consistent with a lack of CaM1 function. We propose that the partially open channel state described previously in [13,14,15] with CaM bound to CaM2 (Figure 4B) might represent the desensitized CNG channel state in dark-adapted photoreceptors that contain high-cytosolic Ca2+ and cGMP levels. A recent cryo-EM structure of the apo CNG channel revealed that the CaM C-lobe is bound to CaM2 (Figure 3C). Our one-site model proposes that CaM binding to CaM2 may stabilize the CNGB1 subunit in a desensitized conformation in which CNGB1 residues in the S6 helix (residues F872, I876 and R880) point inward to block the ion conduction pathway [13] (Figure 4B, inset). This channel pore blockage by CNGB1 residues (F872, I576 and R880) was previously termed the “CNGB1 gate” [15] (Figure 4C). In essence, CaM binding to CaM2 is proposed to keep CNGB1 in a closed conformation by closing the CNGB1 gate (Figure 4C, left and middle panels). Therefore, CaM binding to CaM2 may be necessary to stabilize the partially open channel state (Figure 4C, middle panel) that we propose may represent the desensitized channel in dark-adapted photoreceptors (Figure 4B). We furthermore suggest that the low-cytosolic Ca2+ level in the light-activated photoreceptor ([Ca2+]i ~50 nM) might cause at least some of the bound Ca2+ to dissociate from the CaM C-lobe (Kd for Ca2+ binding to CaM C-lobe bound to CaM2 is 60–80 nM [53]), which, in turn, might cause Ca2+-free CaM to dissociate from CaM2 and possibly allow the CaM-free channel to convert into a more fully open state (Figure 4C, right panel) akin to the fully open state of the homomeric CNGA1 channel [59]. In summary, we propose that Ca2+-induced CNG channel desensitization occurs when the fully open channel (at low Ca2+ levels) converts into the partially open state stabilized by CaM binding to CaM2 at high Ca2+ levels. This one-site model (Figure 4B) predicts that the L1132E/L1136E mutations (that disable CaM binding to CaM2 [53]) should abolish Ca2+-dependent channel desensitization. An important difference between the one-site and two-site models (Figure 4A,B) is that CaM1 should be essential for Ca2+-induced CNG channel desensitization in the two-site model, but not in the one-site model. Another difference is that CaM binding to CaM2 is predicted to stabilize the partially open (desensitized) state in the one-site model (Figure 4B), in contrast to possibly stabilizing the high open-probability state in the two-site model (Figure 4A, right panel). Future electrophysiology experiments and cryo-EM structures of CNG channels bound to CaM (at both high and low Ca2+ levels) are needed to discriminate the two models and test their predictions.

5. Conclusions

Retinal rod CNG channels exhibit Ca2+-induced channel desensitization caused by CaM that may be important for light adaptation. CaM binding to CNG channels is proposed to decrease the channel open probability and/or decrease its cGMP binding affinity at high-cytosolic Ca2+ levels in dark-adapted photoreceptors. A cryo-EM structure of the apo CNG channel [13] suggests that CaM is bound to a CaM binding site (helix D) in CNGB1 that interacts with the CLZ domain. A recent NMR structural analysis [53] suggests that helix D in CNGB1 may be the C-terminal CaM site (CaM2). The NMR structures of CaM bound to separate CNGB1 sites (CaM1 and CaM2) predict that a single CaM could bind to each CNG channel tetramer by having the CaM C-lobe bind to the CNGB1 C-terminal site (CaM2) and the CaM N-lobe bind to the N-terminal site (CaM1). We propose two possible models to explain the Ca2+-induced desensitization of CNG channels caused by CaM. A two-site model (Figure 4A) proposes that a single CaM binds to two separate sites in CNGB1 (CaM1 and CaM2) to cause channel desensitization. The separate binding of the CaM lobes to CaM1 and CaM2 brings the two sites in close proximity to possibly cause channel inactivation at high Ca2+ levels. An alternative one-site model (Figure 4B) proposes that CaM binding to CaM2 alone may be sufficient to cause channel desensitization. An important difference between the two models is that the CaM1 site is essential for Ca2+-induced CNG channel desensitization in the two-site model (Figure 4A), but not in the one-site model (Figure 4B). Moreover, CaM binding to CaM2 is predicted to increase the channel open probability in the two-site model, in contrast to promoting channel desensitization in the one-site model. Future electrophysiology experiments and cryo-EM structures of CNG channels bound to CaM are needed to test the two models. Mutations that affect the Ca2+-dependent desensitization of CNG channels are linked to inherited forms of blindness. Understanding the structural basis of CaM binding to the CNG channel may provide new insights for the future design of therapeutics.

Author Contributions

A.B. prepared the figures and edited the manuscript. J.B.A. directed the project and wrote the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by a grant from the National Institutes of Health, grant number R01 EY012347.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

This work was supported by a grant to J.B.A. from the National Institutes of Health (R01 EY012347).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Matulef, K.; Zagotta, W.N. Cyclic nucleotide-gated ion channels. Annu. Rev. Cell Dev. Biol. 2003, 19, 23–44. [Google Scholar] [CrossRef] [PubMed]
  2. Trudeau, M.C.; Zagotta, W.N. Calcium/Calmodulin Modulation of Olfactory and Rod Cyclic Nucleotide-gated Ion Channels. J. Biol. Chem. 2003, 278, 18705–18708. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Fesenko, E.E.; Kolesnikov, S.S.; Lyubarsky, A.L. Induction by cyclic GMP of cationic conductance in plasma membrane of retinal rod outer segment. Nature 1985, 313, 310–313. [Google Scholar] [CrossRef]
  4. Yau, K.W.; Baylor, D.A. Cyclic GMP-activated conductance of retinal photoreceptor cells. Annu. Rev. Neurosci. 1989, 12, 289–327. [Google Scholar] [CrossRef] [PubMed]
  5. Bradley, J.; Frings, S.; Yau, K.-W.; Reed, R. Nomenclature for Ion Channel Subunits. Science 2001, 294, 2095–2096. [Google Scholar] [CrossRef] [Green Version]
  6. Kaupp, U.B.; Niidome, T.; Tanabe, T.; Terada, S.; Bönigk, W.; Stühmer, W.; Cook, N.J.; Kangawa, K.; Matsuo, H.; Hirose, T.; et al. Primary structure and functional expression from complementary DNA of the rod photoreceptor cyclic GMP-gated channel. Nature 1989, 342, 762–766. [Google Scholar] [CrossRef] [Green Version]
  7. Trudeau, M.C.; Zagotta, W.N. Mechanism of calcium/calmodulin inhibition of rod cyclic nucleotide-gated channels. Proc. Natl. Acad. Sci. USA 2002, 99, 8424–8429. [Google Scholar] [CrossRef] [Green Version]
  8. Cook, N.J.; Hanke, W.; Kaupp, U.B. Identification, purification, and functional reconstitution of the cyclic GMP-dependent channel from rod photoreceptors. Proc. Natl. Acad. Sci. USA 1987, 84, 585–589. [Google Scholar] [CrossRef] [Green Version]
  9. Dryja, T.P.; Finn, J.T.; Peng, Y.W.; McGee, T.L.; Berson, E.L.; Yau, K.W. Mutations in the gene encoding the alpha subunit of the rod cGMP-gated channel in autosomal recessive retinitis pigmentosa. Proc. Natl. Acad. Sci. USA 1995, 92, 10177–10181. [Google Scholar] [CrossRef] [Green Version]
  10. Chen, T.-Y.; Peng, Y.-W.; Dhallan, R.S.; Ahamed, B.; Reed, R.R.; Yau, K.-W. A new subunit of the cyclic nucleotide-gated cation channel in retinal rods. Nature 1993, 362, 764–767. [Google Scholar] [CrossRef]
  11. Shuart, N.G.; Haitin, Y.; Camp, S.S.; Black, K.D.; Zagotta, W.N. Molecular mechanism for 3:1 subunit stoichiometry of rod cyclic nucleotide-gated ion channels. Nat. Commun. 2011, 2, 457. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Barret, D.C.A.; Schertler, G.F.; Benjamin Kaupp, U.; Marino, J. The structure of the native CNGA1/CNGB1 CNG channel from bovine retinal rods. Nat. Struct. Mol. Biol. 2022, 29, 32–39. [Google Scholar] [CrossRef] [PubMed]
  13. Barret, D.C.A.; Schertler, G.F.; Kaupp, U.B.; Marino, J. Structural basis of the partially open central gate in the human CNGA1/CNGB1 channel explained by additional density for calmodulin in cryo-EM map. J. Struct. Biol. 2021, 214, 107828. [Google Scholar] [CrossRef] [PubMed]
  14. Xue, J.; Han, Y.; Zeng, W.; Wang, Y.; Jiang, Y. Structural mechanisms of gating and selectivity of human rod CNGA1 channel. Neuron 2021, 109, 1302–1313.e4. [Google Scholar] [CrossRef] [PubMed]
  15. Barret, D.C.; Kaupp, U.B.; Marino, J. The structure of cyclic nucleotide-gated channels in rod and cone photoreceptors. Trends Neurosci. 2022, 45, 763–776. [Google Scholar] [CrossRef] [PubMed]
  16. Bonigk, W.; Altenhofen, W.; Müller, F.; Dose, A.; Illing, M.; Molday, R.S.; Kaupp, U.B. Rod and cone receptor cells express distinct genes for cGMP-gated channels. Neuron 1993, 10, 865–877. [Google Scholar] [CrossRef]
  17. Gerstner, A.; Zong, X.; Hofmann, F.; Biel, M. Molecular cloning and functional characterization of a new modulatory cyclic nucleotide-gated channel subunit from mouse retina. J. Neurosci. 2000, 20, 1324–1332. [Google Scholar] [CrossRef] [Green Version]
  18. Bright, S.R.; Brown, T.E.; Varnum, M.D. Disease-associated mutations in CNGB3 produce gain of function alterations in cone cyclic nucleotide-gated channels. Mol. Vis. 2005, 11, 1141–1150. [Google Scholar]
  19. Grunwald, M.E.; Yu, W.P.; Yu, H.H.; Yau, K.W. Identification of a domain on the beta-subunit of the rod cGMP-gated cation channel that mediates inhibition by calcium-calmodulin. J. Biol. Chem. 1998, 273, 9148–9157. [Google Scholar] [CrossRef] [Green Version]
  20. Zheng, X.; Hu, Z.; Li, H.; Yang, J. Structure of the human cone photoreceptor cyclic nucleotide-gated channel. Nat. Struct. Mol. Biol. 2020, 29, 40–46. [Google Scholar] [CrossRef]
  21. Kohl, S.; Baumann, B.; Broghammer, M.; Jägle, H.; Sieving, P.; Kellner, U.; Spegal, R.; Anastasi, M.; Zrenner, E.; Sharpe, L.T.; et al. Mutations in the CNGB3 gene encoding the beta-subunit of the cone photoreceptor cGMP-gated channel are responsible for achromatopsia (ACHM3) linked to chromosome 8q21. Hum. Mol. Genet. 2000, 9, 2107–2116. [Google Scholar] [CrossRef]
  22. Sundin, O.H.; Yang, J.-M.; Li, Y.; Zhu, D.; Hurd, J.N.; Mitchell, T.N.; Silva, E.; Maumenee, I.H. Genetic basis of total colourblindness among the Pingelapese islanders. Nat. Genet. 2000, 25, 289–293. [Google Scholar] [CrossRef] [PubMed]
  23. Jumper, J.; Evans, R.; Pritzel, A.; Green, T.; Figurnov, M.; Ronneberger, O.; Tunyasuvunakool, K.; Bates, R.; Žídek, A.; Potapenko, A.; et al. Highly accurate protein structure prediction with AlphaFold. Nature 2021, 596, 583–589. [Google Scholar] [CrossRef] [PubMed]
  24. Baylor, D. How photons start vision. Proc. Natl. Acad. Sci. USA 1996, 93, 560–565. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Stryer, L. Visual excitation and recovery. J. Biol. Chem. 1991, 266, 10711–10714. [Google Scholar] [CrossRef]
  26. Yau, K.W. Phototransduction mechanism in retinal rods and cones. Investig. Ophthalmol. Vis. Sci. 1994, 35, 9–32. [Google Scholar]
  27. Cote, R.H. Photoreceptor phosphodiesterase (PDE6): Activation and inactivation mechanisms during visual transduction in rods and cones. Pflügers Arch.-Eur. J. Physiol. 2021, 473, 1377–1391. [Google Scholar] [CrossRef]
  28. Arshavsky, V.Y.; Burns, M.E. Current understanding of signal amplification in phototransduction. Cell. Logist. 2014, 4, e29390. [Google Scholar] [CrossRef] [Green Version]
  29. Koch, K.-W.; Dell’Orco, D. Protein and Signaling Networks in Vertebrate Photoreceptor Cells. Front. Mol. Neurosci. 2015, 8, 67. [Google Scholar] [CrossRef] [Green Version]
  30. Pugh, E.; Nikonov, S.; Lamb, T. Molecular mechanisms of vertebrate photoreceptor light adaptation. Curr. Opin. Neurobiol. 1999, 9, 410–418. [Google Scholar] [CrossRef]
  31. Gray-Keller, M.P.; Detwiler, P. The calcium feedback signal in the phototransduction cascade of vertebrate rods. Neuron 1994, 13, 849–861. [Google Scholar] [CrossRef]
  32. Schnetkamp, P.P. How does the retinal rod Na-Ca+K exchanger regulate cytosolic free Ca2+? J. Biol. Chem. 1995, 270, 13231–13239. [Google Scholar] [CrossRef] [PubMed]
  33. Fain, G.L.; Matthews, H.R.; Cornwall, M.C.; Koutalos, Y. Adaptation in Vertebrate Photoreceptors. Physiol. Rev. 2001, 81, 117–151. [Google Scholar] [CrossRef] [Green Version]
  34. Hsu, Y.T.; Molday, R.S. Modulation of the cGMP-gated channel of rod photoreceptor cells by calmodulin. Nature 1993, 361, 76–79. [Google Scholar] [CrossRef] [PubMed]
  35. Dizhoor, A. The human photoreceptor membrane guanylyl cyclase, RetGC, is present in outer segments and is regulated by calcium and a soluble activator. Neuron 1994, 12, 1345–1352. [Google Scholar] [CrossRef]
  36. Koch, K.-W.; Stryer, L. Highly cooperative feedback control of retinal rod guanylate cyclase by calcium ions. Nature 1988, 334, 64–66. [Google Scholar] [CrossRef] [PubMed]
  37. Palczewski, K.; Subbaraya, I.; Gorczyca, W.A.; Helekar, B.S.; Ruiz, C.C.; Ohguro, H.; Huang, J.; Zhao, X.; Crabb, J.W.; Johnson, R.S.; et al. Molecular cloning and characterization of retinal photoreceptor guanylyl cyclase-activating protein. Neuron 1994, 13, 395–404. [Google Scholar] [CrossRef]
  38. Bradley, J.; Reisert, J.; Frings, S. Regulation of cyclic nucleotide-gated channels. Curr. Opin. Neurobiol. 2005, 15, 343–349. [Google Scholar] [CrossRef]
  39. Koutalos, Y.; Yau, K.W. Regulation of sensitivity in vertebrate rod photoreceptors by calcium. Trends Neurosci. 1996, 19, 73–81. [Google Scholar] [CrossRef]
  40. Chen, T.Y.; Illing, M.; Molday, L.L.; Hsu, Y.T.; Yau, K.W.; Molday, R.S. Subunit 2 (or beta) of retinal rod cGMP-gated cation channel is a component of the 240-kDa channel-associated protein and mediates Ca(2+)-calmodulin modulation. Proc. Natl. Acad. Sci. USA 1994, 91, 11757–11761. [Google Scholar] [CrossRef] [Green Version]
  41. Weitz, D.; Zoche, M.; Müller, F.; Beyermann, M.; Körschen, H.G.; Kaupp, U.B.; Koch, K.W. Calmodulin controls the rod photoreceptor CNG channel through an unconventional binding site in the N-terminus of the β-subunit. EMBO J. 1998, 17, 2273–2284. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Peng, C.; Rich, E.D.; Thor, C.A.; Varnum, M.D. Functionally Important Calmodulin-binding Sites in Both NH2- and COOH-terminal Regions of the Cone Photoreceptor Cyclic Nucleotide-gated Channel CNGB3 Subunit. J. Biol. Chem. 2003, 278, 24617–24623. [Google Scholar] [CrossRef] [PubMed]
  43. Rebrik, T.I.; Botchkina, I.; Arshavsky, V.Y.; Craft, C.M.; Korenbrot, J.I. CNG-modulin: A novel Ca-dependent modulator of ligand sensitivity in cone photoreceptor cGMP-gated ion channels. J. Neurosci. 2012, 32, 3142–3153. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Rebrik, T.I.; Korenbrot, J.I. In intact mammalian photoreceptors, Ca2+-dependent modulation of cGMP-gated ion channels is detectable in cones but not in rods. J. Gen. Physiol. 2004, 123, 63–75. [Google Scholar] [CrossRef] [Green Version]
  45. Bonigk, W.; Müller, F.; Middendorff, R.; Weyand, I.; Kaupp, U.B. Two alternatively spliced forms of the cGMP-gated channel alpha-subunit from cone photoreceptor are expressed in the chick pineal organ. J. Neurosci. 1996, 16, 7458–7468. [Google Scholar] [CrossRef] [Green Version]
  46. Moncrief, N.; Kretsinger, R.H.; Goodman, M. Evolution of EF-hand calcium-modulated proteins. I. Relationships based on amino acid sequences. J. Mol. Evol. 1990, 30, 522–562. [Google Scholar] [CrossRef]
  47. Babu, Y.; Bugg, C.E.; Cook, W.J. Structure of calmodulin refined at 2.2 Å resolution. J. Mol. Biol. 1988, 204, 191–204. [Google Scholar] [CrossRef]
  48. Gilli, R.; Lafitte, D.; Lopez, C.; Kilhoffer, M.-C.; Makarov, A.; Briand, A.C.; Haiech, J. Thermodynamic Analysis of Calcium and Magnesium Binding to Calmodulin. Biochemistry 1998, 37, 5450–5456. [Google Scholar] [CrossRef]
  49. Ikura, M. Calcium binding and conformational response in EF-hand proteins. Trends Biochem. Sci. 1996, 21, 14–17. [Google Scholar] [CrossRef]
  50. Trudeau, M.C.; Zagotta, W.N. Dynamics of Ca2+-Calmodulin–dependent Inhibition of Rod Cyclic Nucleotide-gated Channels Measured by Patch-clamp Fluorometry. J. Gen. Physiol. 2004, 124, 211–223. [Google Scholar] [CrossRef] [Green Version]
  51. Korenbrot, J.I.; Mehta, M.; Tserentsoodol, N.; Postlethwait, J.H.; Rebrik, T.I. EML1 (CNG-modulin) controls light sensitivity in darkness and under continuous illumination in zebrafish retinal cone photoreceptors. J. Neurosci. 2013, 33, 17763–17776. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Poria, D.; Sun, C.; Santeford, A.; Kielar, M.; Apte, R.S.; Kisselev, O.G.; Chen, S.; Kefalov, V.J. EML1 is essential for retinal photoreceptor migration and survival. Sci. Rep. 2022, 12, 2897. [Google Scholar] [CrossRef] [PubMed]
  53. Bej, A.; Ames, J.B. NMR Structures of Calmodulin Bound to Two Separate Regulatory Sites in the Retinal Cyclic Nucleotide-Gated Channel. Biochemistry 2022, 61, 1955–1965. [Google Scholar] [CrossRef]
  54. Grunwald, M.E.; Yau, K.W. Modulation of rod cGMP-gated cation channel by calmodulin. Met. Enzymol. 2000, 315, 817–828. [Google Scholar]
  55. Sprenger, J.; Trifan, A.; Patel, N.; Vanderbeck, A.; Bredfelt, J.; Tajkhorshid, E.; Rowlett, R.; Leggio, L.L.; Åkerfeldt, K.S.; Linse, S. Calmodulin complexes with brain and muscle creatine kinase peptides. Curr. Res. Struct. Biol. 2021, 3, 121–132. [Google Scholar] [CrossRef]
  56. Ames, J.B. Structural Insights into Retinal Guanylate Cyclase Activator Proteins (GCAPs). Int. J. Mol. Sci. 2021, 22, 8731. [Google Scholar] [CrossRef] [PubMed]
  57. Bartels, P.; Salveson, I.C.; Coleman, A.M.; Anderson, D.; Jeng, G.; Estrada-Tobar, Z.M.; Mimi Man, K.N.; Yu, Q.; Kuzmenkina, E.; Nieves-Cintron, M.; et al. Half-calcified Calmodulin Promotes Basal Activity and Inactivation of the L-type Calcium Channel CaV1.2. bioRxiv 2022. [Google Scholar] [CrossRef]
  58. Spratt, D.; Taiakina, V.; Guillemette, J.G. Calcium-deficient calmodulin binding and activation of neuronal and inducible nitric oxide synthases. Biochim. Biophys. Acta 2007, 1774, 1351–1358. [Google Scholar] [CrossRef]
  59. Zheng, X.; Fu, Z.; Su, D.; Zhang, Y.; Li, M.; Pan, Y.; Li, H.; Li, S.; Grassucci, R.A.; Ren, Z.; et al. Mechanism of ligand activation of a eukaryotic cyclic nucleotide−gated channel. Nat. Struct. Mol. Biol. 2020, 27, 625–634. [Google Scholar] [CrossRef]
  60. Wu, X.; Bers, D.M. Free and bound intracellular calmodulin measurements in cardiac myocytes. Cell Calcium 2007, 41, 353–364. [Google Scholar] [CrossRef]
Figure 2. CNG channel function in visual phototransduction. (A) Schematic model of visual excitation regulated by intracellular Ca2+: Light-excited rhodopsin (R*), GTP-bound transducin (Tα-GTP), phosphodiesterase (PDE6), retinal guanylate cyclase (RetGC), guanylate cyclase activating proteins (GCAPs), phosphorylated rhodopsin (R*-P), arrestin (Arr). (B) CNG channel gating by intracellular cGMP in the plasma membrane of rod photoreceptors.
Figure 2. CNG channel function in visual phototransduction. (A) Schematic model of visual excitation regulated by intracellular Ca2+: Light-excited rhodopsin (R*), GTP-bound transducin (Tα-GTP), phosphodiesterase (PDE6), retinal guanylate cyclase (RetGC), guanylate cyclase activating proteins (GCAPs), phosphorylated rhodopsin (R*-P), arrestin (Arr). (B) CNG channel gating by intracellular cGMP in the plasma membrane of rod photoreceptors.
Ijms 23 14143 g002
Figure 3. CaM binding sites in rod CNGB1 vs. cone CNGB3. (A) Domain architecture and amino acid sequence of CaM binding sites (CaM1 and CaM2) in CNGB1. Sequence alignment of CaM1 and CaM2 with corresponding residues in CNGA2, creatine kinase (CK), cone CNGB3 and vertebrate homologs of CNGB1. Conserved residues are highlighted in color. (B) NMR structure of the Ca2+-bound CaM N-lobe (light green) bound to CaM1 peptide (red) [53]. (C) Cryo-EM structure of CaM C-lobe (cyan) bound to the D-helix of CNGB1 (red) [13]. (D) NMR structure of the Ca2+-bound CaM C-lobe (dark green) bound to CaM2 peptide (pink) [53].
Figure 3. CaM binding sites in rod CNGB1 vs. cone CNGB3. (A) Domain architecture and amino acid sequence of CaM binding sites (CaM1 and CaM2) in CNGB1. Sequence alignment of CaM1 and CaM2 with corresponding residues in CNGA2, creatine kinase (CK), cone CNGB3 and vertebrate homologs of CNGB1. Conserved residues are highlighted in color. (B) NMR structure of the Ca2+-bound CaM N-lobe (light green) bound to CaM1 peptide (red) [53]. (C) Cryo-EM structure of CaM C-lobe (cyan) bound to the D-helix of CNGB1 (red) [13]. (D) NMR structure of the Ca2+-bound CaM C-lobe (dark green) bound to CaM2 peptide (pink) [53].
Ijms 23 14143 g003
Figure 4. Two structural models of CNG channel regulation controlled by CaM. (A) Two-site model proposes that CaM binds to two separate sites (CaM1 and CaM2). Adapted from [53]. CNGB1 is highlighted in red. Bound Ca2+ are magenta spheres. The CNG channel at low Ca2+ concentration in light-activated photoreceptors has the Ca2+-bound CaM C-lobe (dark green) bound to CaM2 (light red) and Ca2+-free CaM N-lobe (light green) does not bind to the channel (right panel). The CNG channel at high Ca2+ concentration in the dark-adapted photoreceptor has the Ca2+-bound CaM C-lobe and N-lobe bound to CaM2 and CaM1, respectively (left panel). (B) A one-site model proposes that CaM binds functionally to only one site (CaM2). At high Ca2+ levels, the Ca2+-bound CaM C-lobe binds to CaM2 and is proposed to stabilize the structure of the S6 helix that causes CNGB1 residues (F872, I876 and R880) to point inward to block the channel pore in the desensitized state (see inset). At low Ca2+ levels, the Ca2+-free CaM dissociates from CaM2, which could allow for the channel to switch to a proposed fully open state, as seen in the homomeric CNGA1 [59]. (C) Proposed model for Ca2+-dependent gating of the rod CNG channel. The closed apo state (in the light-activated photoreceptor) has CNGB1 gate residues (L872, I876 and R880) and CNGA1 residues (F389 and V393), each pointing inward to block the ion conduction pathway (left panel). The binding of cGMP causes the formation of a partially open channel (middle panel) in which CNGB1 gate residues are pointing inward to partially block the ion conduction pathway, while CNGA2 residues (F389 and V393) are pointing outward. We propose that the partial open state (middle panel) may represent the desensitized channel state in dark-adapted photoreceptors (high Ca2+ and cGMP). A hypothetical fully open channel (right panel) is proposed to exist at low Ca2+ levels, in which the CNGB1 gate residues are pointing outward to fully unblock the ion conduction pathway. A similar fully open state was observed for the homomeric CNGA1 channel [59] that has each CNGA1 gate residues pointing outward to fully unblock the ion conduction pathway.
Figure 4. Two structural models of CNG channel regulation controlled by CaM. (A) Two-site model proposes that CaM binds to two separate sites (CaM1 and CaM2). Adapted from [53]. CNGB1 is highlighted in red. Bound Ca2+ are magenta spheres. The CNG channel at low Ca2+ concentration in light-activated photoreceptors has the Ca2+-bound CaM C-lobe (dark green) bound to CaM2 (light red) and Ca2+-free CaM N-lobe (light green) does not bind to the channel (right panel). The CNG channel at high Ca2+ concentration in the dark-adapted photoreceptor has the Ca2+-bound CaM C-lobe and N-lobe bound to CaM2 and CaM1, respectively (left panel). (B) A one-site model proposes that CaM binds functionally to only one site (CaM2). At high Ca2+ levels, the Ca2+-bound CaM C-lobe binds to CaM2 and is proposed to stabilize the structure of the S6 helix that causes CNGB1 residues (F872, I876 and R880) to point inward to block the channel pore in the desensitized state (see inset). At low Ca2+ levels, the Ca2+-free CaM dissociates from CaM2, which could allow for the channel to switch to a proposed fully open state, as seen in the homomeric CNGA1 [59]. (C) Proposed model for Ca2+-dependent gating of the rod CNG channel. The closed apo state (in the light-activated photoreceptor) has CNGB1 gate residues (L872, I876 and R880) and CNGA1 residues (F389 and V393), each pointing inward to block the ion conduction pathway (left panel). The binding of cGMP causes the formation of a partially open channel (middle panel) in which CNGB1 gate residues are pointing inward to partially block the ion conduction pathway, while CNGA2 residues (F389 and V393) are pointing outward. We propose that the partial open state (middle panel) may represent the desensitized channel state in dark-adapted photoreceptors (high Ca2+ and cGMP). A hypothetical fully open channel (right panel) is proposed to exist at low Ca2+ levels, in which the CNGB1 gate residues are pointing outward to fully unblock the ion conduction pathway. A similar fully open state was observed for the homomeric CNGA1 channel [59] that has each CNGA1 gate residues pointing outward to fully unblock the ion conduction pathway.
Ijms 23 14143 g004
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Bej, A.; Ames, J.B. Retinal Cyclic Nucleotide-Gated Channel Regulation by Calmodulin. Int. J. Mol. Sci. 2022, 23, 14143. https://doi.org/10.3390/ijms232214143

AMA Style

Bej A, Ames JB. Retinal Cyclic Nucleotide-Gated Channel Regulation by Calmodulin. International Journal of Molecular Sciences. 2022; 23(22):14143. https://doi.org/10.3390/ijms232214143

Chicago/Turabian Style

Bej, Aritra, and James B. Ames. 2022. "Retinal Cyclic Nucleotide-Gated Channel Regulation by Calmodulin" International Journal of Molecular Sciences 23, no. 22: 14143. https://doi.org/10.3390/ijms232214143

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop