Skip to content
Licensed Unlicensed Requires Authentication Published by De Gruyter November 19, 2019

The cGMP system: components and function

  • Franz Hofmann ORCID logo EMAIL logo
From the journal Biological Chemistry

Abstract

The cyclic guanosine monophosphate (cGMP) signaling system is one of the most prominent regulators of a variety of physiological and pathophysiological processes in many mammalian and non-mammalian tissues. Targeting this pathway by increasing cGMP levels has been a very successful approach in pharmacology as shown for nitrates, phosphodiesterase (PDE) inhibitors and stimulators of nitric oxide-guanylyl cyclase (NO-GC) and particulate GC (pGC). This is an introductory review to the cGMP signaling system intended to introduce those readers to this system, who do not work in this area. This article does not intend an in-depth review of this system. Signal transduction by cGMP is controlled by the generating enzymes GCs, the degrading enzymes PDEs and the cGMP-regulated enzymes cyclic nucleotide-gated ion channels, cGMP-dependent protein kinases and cGMP-regulated PDEs. Part A gives a very concise introduction to the components. Part B gives a very concise introduction to the functions modulated by cGMP. The article cites many recent reviews for those who want a deeper insight.

References

Ahsan, M.K., Tchernychev, B., Kessler, M.M., Solinga, R.M., Arthur, D., Linde, C.I., Silos-Santiago, I., Hannig, G., and Ameen, N.A. (2017). Linaclotide activates guanylate cyclase-C/cGMP/protein kinase-II-dependent trafficking of CFTR in the intestine. Physiol. Rep. 5, e13299.10.14814/phy2.13299Search in Google Scholar PubMed PubMed Central

Aicher, A., Heeschen, C., Feil, S., Hofmann, F., Mendelsohn, M.E., Feil, R., and Dimmeler, S. (2009). cGMP-dependent protein kinase I is crucial for angiogenesis and postnatal vasculogenesis. PLoS One 4, e4879.10.1371/journal.pone.0004879Search in Google Scholar PubMed PubMed Central

Airhart, N., Yang, Y.F., Roberts Jr, C.T., and Silberbach, M. (2003). Atrial natriuretic peptide induces natriuretic peptide receptor-cGMP-dependent protein kinase interaction. J. Biol. Chem. 278, 38693–38698.10.1074/jbc.M304098200Search in Google Scholar PubMed

Al-Salama, Z.T. and Syed, Y.Y. (2017). Plecanatide: first global approval. Drugs 77, 593–598.10.1007/s40265-017-0718-0Search in Google Scholar PubMed

Alam, M.M., Solyakov, L., Bottrill, A.R., Flueck, C., Siddiqui, F.A., Singh, S., Mistry, S., Viskaduraki, M., Lee, K., Hopp, C.S., et al. (2015). Phosphoproteomics reveals malaria parasite Protein Kinase G as a signalling hub regulating egress and invasion. Nat. Commun. 6, 7285.10.1038/ncomms8285Search in Google Scholar PubMed PubMed Central

Alam, M.M., Sanchez-Azqueta, A., Janha, O., Flannery, E.L., Mahindra, A., Mapesa, K., Char, A.B., Sriranganadane, D., Brancucci, N.M.B., Antonova-Koch, Y., et al. (2019). Validation of the protein kinase PfCLK3 as a multistage cross-species malarial drug target. Science 365, 6456 Aug 6430.10.1126/science.aau1682Search in Google Scholar

Alioua, A., Tanaka, Y., Wallner, M., Hofmann, F., Ruth, P., Meera, P., and Toro, L. (1998). The large conductance, voltage-dependent, and calcium-sensitive K+ channel, Hslo, is a target of cGMP-dependent protein kinase phosphorylation in vivo. J. Biol. Chem. 273, 32950–32956.10.1074/jbc.273.49.32950Search in Google Scholar PubMed

Allen, A.M., Anreiter, I., Neville, M.C., and Sokolowski, M.B. (2017). Feeding-related traits are affected by dosage of the foraging gene in Drosophila melanogaster. Genetics 205, 761–773.10.1534/genetics.116.197939Search in Google Scholar PubMed PubMed Central

Ammendola, A., Geiselhoringer, A., Hofmann, F., and Schlossmann, J. (2001). Molecular determinants of the interaction between the inositol 1,4,5-trisphosphate receptor-associated cGMP kinase substrate (IRAG) and cGMP kinase Ibeta. J. Biol. Chem. 276, 24153–24159.10.1074/jbc.M101530200Search in Google Scholar PubMed

Andersen, M.J., Ersboll, M., Axelsson, A., Gustafsson, F., Hassager, C., Kober, L., Borlaug, B.A., Boesgaard, S., Skovgaard, L.T., and Moller, J.E. (2013). Sildenafil and diastolic dysfunction after acute myocardial infarction in patients with preserved ejection fraction: the Sildenafil and Diastolic Dysfunction After Acute Myocardial Infarction (SIDAMI) trial. Circulation 127, 1200–1208.10.1161/CIRCULATIONAHA.112.000056Search in Google Scholar PubMed

Angermeier, E., Domes, K., Lukowski, R., Schlossmann, J., Rathkolb, B., Angelis, M.H., and Hofmann, F. (2016). Iron deficiency anemia in cyclic GMP kinase knockout mice. Haematologica 101, e48–51.10.3324/haematol.2015.137026Search in Google Scholar PubMed PubMed Central

Anreiter, I. and Sokolowski, M. (2019). The foraging gene and its behavioral effects: pleiotropy and plasticity. Annu. Rev. Genet. 53. Doi: 10.1146/annurev-genet-112618-043536.10.1146/annurev-genet-112618-043536Search in Google Scholar PubMed

Antl, M., von Bruhl, M.L., Eiglsperger, C., Werner, M., Konrad, I., Kocher, T., Wilm, M., Hofmann, F., Massberg, S., and Schlossmann, J. (2007). IRAG mediates NO/cGMP-dependent inhibition of platelet aggregation and thrombus formation. Blood 109, 552–559.10.1182/blood-2005-10-026294Search in Google Scholar PubMed

Arimoto, H. and Takahashi, D. (2017). 8-Nitro-cGMP: a novel protein-reactive cNMP and its emerging roles in autophagy. Handb. Exp. Pharmacol. 238, 253–268.10.1007/164_2016_5000Search in Google Scholar PubMed

Baker, D.A., Drought, L.G., Flueck, C., Nofal, S.D., Patel, A., Penzo, M., and Walker, E.M. (2017a). Cyclic nucleotide signalling in malaria parasites. Open Biol. 7, 170213.10.1098/rsob.170213Search in Google Scholar PubMed PubMed Central

Baker, D.A., Stewart, L.B., Large, J.M., Bowyer, P.W., Ansell, K.H., Jimenez-Diaz, M.B., El Bakkouri, M., Birchall, K., Dechering, K.J., Bouloc, N.S., et al. (2017b). A potent series targeting the malarial cGMP-dependent protein kinase clears infection and blocks transmission. Nat Commun. 8, 430.10.1038/s41467-017-00572-xSearch in Google Scholar PubMed PubMed Central

Balligand, J.L., Kelly, R.A., Marsden, P.A., Smith, T.W., and Michel, T. (1993). Control of cardiac muscle cell function by an endogenous nitric oxide signaling system. Proc. Natl. Acad. Sci. U.S.A. 90, 347–351.10.1073/pnas.90.1.347Search in Google Scholar PubMed PubMed Central

Behrends, S. and Vehse, K. (2000). The beta(2) subunit of soluble guanylyl cyclase contains a human-specific frameshift and is expressed in gastric carcinoma. Biochem. Biophys. Res. Commun. 271, 64–69.10.1006/bbrc.2000.2596Search in Google Scholar PubMed

Bice, J.S., Keim, Y., Stasch, J.P., and Baxter, G.F. (2014). NO-independent stimulation or activation of soluble guanylyl cyclase during early reperfusion limits infarct size. Cardiovasc Res. 101, 220–228.10.1093/cvr/cvt257Search in Google Scholar PubMed PubMed Central

Biel, M. and Michalakis, S. (2009). Cyclic nucleotide-gated channels. Handb. Exp. Pharmacol. 191, 111–136.10.1007/978-3-540-68964-5_7Search in Google Scholar PubMed

Bisio, H. and Soldati-Favre, D. (2019). Signaling cascades governing entry into and exit from host cells by Toxoplasma gondii. Annu. Rev. Microbiol. 73, 579–599.10.1146/annurev-micro-020518-120235Search in Google Scholar PubMed

Blanton, R.M. (2019). cGMP signaling and modulation in heart failure. J. Cardiovasc. Pharmacol. Aug 27. [Epub ahead of print].10.1097/FJC.0000000000000749Search in Google Scholar

Blanton, R.M., Takimoto, E., Lane, A.M., Aronovitz, M., Piotrowski, R., Karas, R.H., Kass, D.A., and Mendelsohn, M.E. (2012). Protein kinase g ialpha inhibits pressure overload-induced cardiac remodeling and is required for the cardioprotective effect of sildenafil in vivo. J. Am. Heart Assoc. 1, e003731.10.1161/JAHA.112.003731Search in Google Scholar

Bradley, J., Reisert, J., and Frings, S. (2005). Regulation of cyclic nucleotide-gated channels. Curr. Opin. Neurobiol. 15, 343–349.10.1016/j.conb.2005.05.014Search in Google Scholar

Browning, D.D., Kwon, I.K., and Wang, R. (2010). cGMP-dependent protein kinases as potential targets for colon cancer prevention and treatment. Future Medicinal. Chem. 2, 65–80.10.4155/fmc.09.142Search in Google Scholar

Bryant, A.P., Busby, R.W., Bartolini, W.P., Cordero, E.A., Hannig, G., Kessler, M.M., Pierce, C.M., Solinga, R.M., Tobin, J.V., Mahajan-Miklos, S., et al. (2010). Linaclotide is a potent and selective guanylate cyclase C agonist that elicits pharmacological effects locally in the gastrointestinal tract. Life Sci. 86, 760–765.10.1016/j.lfs.2010.03.015Search in Google Scholar

Bubb, K.J., Aubdool, A.A., Moyes, A.J., Lewis, S., Drayton, J.P., Tang, O., Mehta, V., Zachary, I.C., Abraham, D.J., Tsui, J., et al. (2019). Endothelial C-type natriuretic peptide is a critical regulator of angiogenesis and vascular remodeling. Circulation 139, 1612–1628.10.1161/CIRCULATIONAHA.118.036344Search in Google Scholar

Burgoyne, J.R., Madhani, M., Cuello, F., Charles, R.L., Brennan, J.P., Schroder, E., Browning, D.D., and Eaton, P. (2007). Cysteine redox sensor in PKGIa enables oxidant-induced activation.Science 317, 1393–1397.10.1126/science.1144318Search in Google Scholar

Buxton, I.L. and Duan, D. (2008). Cyclic GMP/protein kinase G phosphorylation of Smad3 blocks transforming growth factor-beta-induced nuclear Smad translocation: a key antifibrogenic mechanism of atrial natriuretic peptide. Circ. Res. 102, 151–153.10.1161/CIRCRESAHA.107.170217Search in Google Scholar

Cahill, P.A. and Hassid, A. (1991). Clearance receptor-binding atrial natriuretic peptides inhibit mitogenesis and proliferation of rat aortic smooth-muscle cells. Biochem. Biophys. Res. Commun. 179, 1606–1613.10.1016/0006-291X(91)91758-5Search in Google Scholar

Cawley, S.M., Kolodziej, S., Ichinose, F., Brouckaert, P., Buys, E.S., and Bloch, K.D. (2011). sGC{alpha}1 mediates the negative inotropic effects of NO in cardiac myocytes independent of changes in calcium handling. Am. J. Physiol. Heart Circ. Physiol. 301, H157–H163.10.1152/ajpheart.01273.2010Search in Google Scholar PubMed PubMed Central

Chen, W., Oberwinkler, H., Werner, F., Ganer, B., Nakagawa, H., Feil, R., Hofmann, F., Schlossmann, J., Dietrich, A., Gudermann, T., et al. (2013). Atrial natriuretic peptide-mediated inhibition of microcirculatory endothelial Ca2+ and permeability response to histamine involves cGMP-dependent protein kinase I and TRPC6 channels. Arterioscler. Thromb. Vasc. Biol. 33, 2121–2129.10.1161/ATVBAHA.113.001974Search in Google Scholar

Chen, T., Kocinsky, H.S., Cha, B.Y., Murtazina, R., Yang, J.B., Tse, C.M., Singh, V., Cole, R., Aronson, P.S., de Jonge, H., et al. (2015). Cyclic GMP kinase II (cGKII) inhibits NHE3 by altering its trafficking and phosphorylating NHE3 at three required sites identification of a multifunctional phosphorylation site. J. Biol. Chem. 290, 1952–1965.10.1074/jbc.M114.590174Search in Google Scholar

Chen, Y., Harty, G.J., Huntley, B.K., Iyer, S.R., Heublein, D.M., Harders, G.E., Meems, L., Pan, S., Sangaralingham, S.J., Ichiki, T., et al. (2018). CRRL269: a novel designer and renal-enhancing pGC-A peptide activator. Am. J. Physiol. Regul. Integr. Comp. Physiol. 314, R407–R414.10.1152/ajpregu.00286.2017Search in Google Scholar

Chen, Y., Harty, G.J., Zheng, Y., Iyer, S.R., Sugihara, S., Sangaralingham, S.J., Ichiki, T., Grande, J.P., Lee, H.C., Wang, X., et al. (2019). Crrl269. Circ. Res. 124, 1462–1472.10.1161/CIRCRESAHA.118.314164Search in Google Scholar

Conti, M. and Beavo, J. (2007). Biochemistry and physiology of cyclic nucleotide phosphodiesterases: essential components in cyclic nucleotide signaling. Annu. Rev. Biochem. 76, 481–511.10.1146/annurev.biochem.76.060305.150444Search in Google Scholar

Dason, J.S., Cheung, A., Anreiter, I., Montemurri, V.A., Allen, A.M., and Sokolowski, M.B. (2019). Drosophila melanogaster foraging regulates a nociceptive-like escape behavior through a developmentally plastic sensory circuit. Proc. Natl. Acad. Sci. U.S.A. Doi: 10.1073/pnas.1820840116.10.1073/pnas.1820840116Search in Google Scholar

Dazert, P., Meissner, K., Vogelgesang, S., Heydrich, B., Eckel, L., Bohm, M., Warzok, R., Kerb, R., Brinkmann, U., Schaeffeler, E., et al. (2003). Expression and localization of the multidrug resistance protein 5 (MRP5/ABCC5), a cellular export pump for cyclic nucleotides, in human heart. Am. J. Pathol. 163, 1567–1577.10.1016/S0002-9440(10)63513-4Search in Google Scholar

Degen, C.V., Bishu, K., Zakeri, R., Ogut, O., Redfield, M.M., and Brozovich, F.V. (2015). The emperor’s new clothes: PDE5 and the heart. PLoS One 10, e0118664.10.1371/journal.pone.0118664Search in Google Scholar PubMed PubMed Central

Desai, K.M., Zembowicz, A., Sessa, W.C., and Vane, J.R. (1991). Nitroxergic nerves mediate vagally induced relaxation in the isolated stomach of the guinea pig. Proc. Natl. Acad. Sci. U.S.A. 88, 11490–11494.10.1073/pnas.88.24.11490Search in Google Scholar PubMed PubMed Central

Dryja, T.P., Finn, J.T., Peng, Y.W., McGee, T.L., Berson, E.L., and Yau, K.W. (1995). Mutations in the gene encoding the alpha subunit of the rod cGMP-gated channel in autosomal recessive retinitis pigmentosa. Proc. Natl. Acad. Sci. U.S.A. 92, 10177–10181.10.1073/pnas.92.22.10177Search in Google Scholar PubMed PubMed Central

Dumoulin, A., Ter-Avetisyan, G., Schmidt, H., and Rathjen, F.G. (2018). Molecular analysis of sensory axon branching unraveled a cGMP-dependent signaling cascade. Int. J. Mol. Sci. 19, pii: E1266.10.3390/ijms19051266Search in Google Scholar

Ecker, T., Gobel, C., Hullin, R., Rettig, R., Seitz, G., and Hofmann, F. (1989). Decreased cardiac concentration of cGMP kinase in hypertensive animals. An index for cardiac vascularization? Circ. Res. 65, 1361–1369.10.1161/01.RES.65.5.1361Search in Google Scholar

Edmund, A.B., Walseth, T.F., Levinson, N.M., and Potter, L.R. (2019). The pseudokinase domains of guanylyl cyclase-A and -B allosterically increase the affinity of their catalytic domains for substrate. Sci. Signal. 12, pii: eaau5378.10.1126/scisignal.aau5378Search in Google Scholar

El Bakkouri, M., Kouidmi, I., Wernimont, A.K., Amani, M., Hutchinson, A., Loppnau, P., Kim, J.J., Flueck, C., Walker, J.R., Seitova, A., et al. (2019). Structures of the cGMP-dependent protein kinase in malaria parasites reveal a unique structural relay mechanism for activation. Proc. Natl. Acad. Sci. U.S.A. 116, 14164–14173.10.1073/pnas.1905558116Search in Google Scholar

Emdin, C.A., Khera, A.V., Klarin, D., Natarajan, P., Zekavat, S.M., Nomura, A., Haas, M., Aragam, K., Ardissino, D., Wilson, J.G., et al. (2018). Phenotypic consequences of a genetic predisposition to enhanced nitric oxide signaling. Circulation 137, 222–232.10.1161/CIRCULATIONAHA.117.028021Search in Google Scholar

Erdmann, J., Stark, K., Esslinger, U.B., Rumpf, P.M., Koesling, D., de Wit, C., Kaiser, F.J., Braunholz, D., Medack, A., Fischer, M., et al. (2013). Dysfunctional nitric oxide signalling increases risk of myocardial infarction. Nature 504, 432–6.10.1038/nature12722Search in Google Scholar

Erdmann, J., Kessler, T., Munoz Venegas, L., and Schunkert, H. (2018). A decade of genome-wide association studies for coronary artery disease: the challenges ahead. Cardiovasc. Res. 114, 1241–1257.10.1093/cvr/cvy084Search in Google Scholar

Farber, D.B. and Danciger, M. (1997). Identification of genes causing photoreceptor degenerations leading to blindness. Curr. Opin. Neurobiol. 7, 666–673.10.1016/S0959-4388(97)80087-6Search in Google Scholar

Feil, R., Hartmann, J., Luo, C., Wolfsgruber, W., Schilling, K., Feil, S., Barski, J.J., Meyer, M., Konnerth, A., De Zeeuw, C.I., et al. (2003). Impairment of LTD and cerebellar learning by Purkinje cell-specific ablation of cGMP-dependent protein kinase I. J. Cell Biol. 163, 295–302.10.1083/jcb.200306148Search in Google Scholar PubMed PubMed Central

Feil, S., Zimmermann, P., Knorn, A., Brummer, S., Schlossmann, J., Hofmann, F., and Feil, R. (2005). Distribution of cGMP-dependent protein kinase type I and its isoforms in the mouse brain and retina. Neuroscience 135, 863–868.10.1016/j.neuroscience.2005.06.051Search in Google Scholar PubMed

Francis, S.H. and Corbin, J.D. (2005). Phosphodiesterase-5 inhibition: the molecular biology of erectile function and dysfunction. Urol. Clin. North Am. 32, 419–29.10.1016/j.ucl.2005.08.001Search in Google Scholar PubMed

Francis, S.H., Corbin, J.D., and Bischoff, E. (2009). Cyclic GMP-hydrolyzing phosphodiesterases. Handb. Exp. Pharmacol. 191, 367–408.10.1007/978-3-540-68964-5_16Search in Google Scholar

Frankenreiter, S., Bednarczyk, P., Kniess, A., Bork, N.I., Straubinger, J., Koprowski, P., Wrzosek, A., Mohr, E., Logan, A., Murphy, M.P., et al. (2017). cGMP-elevating compounds and ischemic conditioning provide cardioprotection against ischemia and reperfusion injury via cardiomyocyte-specific BK channels. Circulation 136, 2337–2355.10.1161/CIRCULATIONAHA.117.028723Search in Google Scholar

Frankenreiter, S., Groneberg, D., Kuret, A., Krieg, T., Ruth, P., Friebe, A., and Lukowski, R. (2018). Cardioprotection by ischemic postconditioning and cyclic guanosine monophosphate-elevating agents involves cardiomyocyte nitric oxide-sensitive guanylyl cyclase. Cardiovasc. Res. 114, 822–829.10.1093/cvr/cvy039Search in Google Scholar

Franz, E., Knape, M.J., and Herberg, F.W. (2018). cGMP binding domain D mediates a unique activation mechanism in Plasmodium falciparum PKG. ACS Infect. Dis. 4, 415–423.10.1021/acsinfecdis.7b00222Search in Google Scholar

Frolich, L., Wunderlich, G., Thamer, C., Roehrle, M., Garcia Jr, M., and Dubois, B. (2019). Evaluation of the efficacy, safety and tolerability of orally administered BI 409306, a novel phosphodiesterase type 9 inhibitor, in two randomised controlled phase II studies in patients with prodromal and mild Alzheimer’s disease. Alzheimers Res. Ther. 11, 18.10.1186/s13195-019-0467-2Search in Google Scholar

Galie, N., Ghofrani, H.A., Torbicki, A., Barst, R.J., Rubin, L.J.,Badesch, D., Fleming, T., Parpia, T., Burgess, G., Branzi, A., et al. (2005). Sildenafil citrate therapy for pulmonary arterial hypertension. N. Engl. J. Med. 353, 2148–2157.10.1056/NEJMoa050010Search in Google Scholar

Gambaryan, S., Wagner, C., Smolenski, A., Walter, U., Poller, W., Haase, W., Kurtz, A., and Lohmann, S.M. (1998). Endogenous or overexpressed cGMP-dependent protein kinases inhibit cAMP-dependent renin release from rat isolated perfused kidney, microdissected glomeruli, and isolated juxtaglomerular cells. Proc. Natl. Acad. Sci. U.S.A. 95, 9003–9008.10.1073/pnas.95.15.9003Search in Google Scholar

Garbers, D.L., Koesling, D., and Schultz, G. (1994). Guanylyl cyclase receptors. Mol. Biol. Cell 5, 1–5.10.1091/mbc.5.1.1Search in Google Scholar

Garthwaite, J., Southam, E., Boulton, C.L., Nielsen, E.B., Schmidt, K., and Mayer, B. (1995). Potent and selective inhibition of nitric oxide-sensitive guanylyl cyclase by 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one. Mol. Pharmacol. 48, 184–188.Search in Google Scholar

Geiselhöringer, A., Gaisa, M., Hofmann, F., and Schlossmann, J. (2004). Distribution of IRAG and cGKI-isoforms in murine tissues. FEBS Lett. 575, 19–22.10.1016/j.febslet.2004.08.030Search in Google Scholar

Gerzer, R., Bohme, E., Hofmann, F., and Schultz, G. (1981). Soluble guanylate cyclase purified from bovine lung contains heme and copper. FEBS Lett. 132, 71–74.10.1016/0014-5793(81)80429-2Search in Google Scholar

Ghofrani, H.A., D’Armini, A.M., Grimminger, F., Hoeper, M.M., Jansa, P., Kim, N.H., Mayer, E., Simonneau, G., Wilkins, M.R., Fritsch, A., et al. (2013a). Riociguat for the treatment of chronic thromboembolic pulmonary hypertension. N. Engl. J. Med. 369, 319–329.10.1056/NEJMoa1209657Search in Google Scholar PubMed

Ghofrani, H.A., Galie, N., Grimminger, F., Grunig, E., Humbert, M., Jing, Z.C., Keogh, A.M., Langleben, D., Kilama, M.O., Fritsch, A., et al. (2013b). Riociguat for the treatment of pulmonary arterial hypertension. N. Engl. J. Med. 369, 330–340.10.1056/NEJMoa1209655Search in Google Scholar PubMed

Golombek, D.A., Agostino, P.V., Plano, S.A., and Ferreyra, G.A. (2004). Signaling in the mammalian circadian clock: the NO/cGMP pathway. Neurochem. Int. 45, 929–936.10.1016/j.neuint.2004.03.023Search in Google Scholar PubMed

Gong, K., Xing, D., Li, P., Hilgers, R.H., Hage, F.G., Oparil, S., and Chen, Y.F. (2011). cGMP inhibits TGF-beta signaling by sequestering Smad3 with cytosolic beta2-tubulin in pulmonary artery smooth muscle cells. Mol. Endocrinol. 25, 1794–1803.10.1210/me.2011-1009Search in Google Scholar PubMed PubMed Central

Gotz, K.R., Sprenger, J.U., Perera, R.K., Steinbrecher, J.H., Lehnart, S.E., Kuhn, M., Gorelik, J., Balligand, J.L., and Nikolaev, V.O. (2014). Transgenic mice for real-time visualization of cGMP in intact adult cardiomyocytes. Circ. Res. 114, 1235–1245.10.1161/CIRCRESAHA.114.302437Search in Google Scholar PubMed

Groneberg, D., Lies, B., Konig, P., Jager, R., Seidler, B., Klein, S., Saur, D., and Friebe, A. (2013). Cell-specific deletion of nitric oxide-sensitive guanylyl cyclase reveals a dual pathway for nitrergic neuromuscular transmission in the murine fundus. Gastroenterology 145, 188–196.10.1053/j.gastro.2013.03.042Search in Google Scholar PubMed

Guggino, W.B. and Stanton, B.A. (2006). New insights into cystic fibrosis: molecular switches that regulate CFTR. Nat. Rev. Mol. Cell. Biol. 7, 426–436.10.1038/nrm1949Search in Google Scholar PubMed

Guo, D.C., Regalado, E., Casteel, D.E., Santos-Cortez, R.L., Gong, L., Kim, J.J., Dyack, S., Horne, S.G., Chang, G., Jondeau, G., et al. (2013). Recurrent gain-of-function mutation in PRKG1 causes thoracic aortic aneurysms and acute aortic dissections. Am. J. Hum. Genet. 93, 398–404.10.1016/j.ajhg.2013.06.019Search in Google Scholar PubMed PubMed Central

Gyurko, R., Kuhlencordt, P., Fishman, M.C., and Huang, P.L. (2000). Modulation of mouse cardiac function in vivo by eNOS and ANP. Am. J. Physiol. Heart Circ. Physiol. 278, H971–H981.10.1152/ajpheart.2000.278.3.H971Search in Google Scholar PubMed

Hassid, A., Cahill, P.A., and Patel, T.B. (1991). Inhibition of rat aortic smooth-muscle cell mitogenesis by atrial-natriuretic-peptide is mediated by a cyclic GMP-independent mechanism via the anf clearance receptor. FASEB J. 5, A1608.Search in Google Scholar

Hedlund, P., Aszodi, A., Pfeifer, A., Alm, P., Hofmann, F., Ahmad, M., Fassler, R., and Andersson, K.E. (2000). Erectile dysfunction in cyclic CMP-dependent kinase I-deficient mice. Proc. Natl. Acad. Sci. U.S.A. 97, 2349–2354.10.1073/pnas.030419997Search in Google Scholar PubMed PubMed Central

Hemmrich, M., Distler, O., Allanore, Y., Denton, C., Kuwana, M., Matucci-Cerinic, M., Pope, J., de Oliveira Pena, J., Laapas, K., Yao, Z., et al. (2019). Efficacy and safety of riociguat in patients with early diffuse cutaneous systemic sclerosis: results from the RISE-SSc study, a randomized, double-blind, placebo-controlled phase IIb study. J. Transl. Med. 17, S1–03.10.1164/ajrccm-conference.2019.199.1_MeetingAbstracts.A4086Search in Google Scholar

Higenbottam, T., Siddons, T., and Demoncheaux, E. (2000). A therapeutic role for chronic inhaled nitric oxide? Lancet 356, 446–447.10.1016/S0140-6736(00)02548-4Search in Google Scholar

Hofmann, F. (2018). A concise discussion of the regulatory role of cGMP kinase I in cardiac physiology and pathology. Basic Res. Cardiol. 113, 31.10.1007/s00395-018-0690-1Search in Google Scholar

Hofmann, F. and Sold, G. (1972). A protein kinase activity from rat cerebellum stimulated by guanosine-3′:5′-monophosphate. Biochem. Biophys. Res. Commun. 49, 1100–1107.10.1016/0006-291X(72)90326-9Search in Google Scholar

Hofmann, F., Feil, R., Kleppisch, T., and Schlossmann, J. (2006). Function of cGMP-dependent protein kinases as revealed by gene deletion. Physiol. Rev. 86, 1–23.10.1152/physrev.00015.2005Search in Google Scholar PubMed

Hofmann, F., Bernhard, D., Lukowski, R., and Weinmeister, P. (2009). cGMP regulated protein kinases (cGK). Handb. Exp. Pharmacol. 191, 137–162.10.1007/978-3-540-68964-5_8Search in Google Scholar PubMed

Hollas, M.A., Ben Aissa, M., Lee, S.H., Gordon-Blake, J.M., and Thatcher, G.R.J. (2019). Pharmacological manipulation of cGMP and NO/cGMP in CNS drug discovery. Nitric Oxide 82, 59–74.10.1016/j.niox.2018.10.006Search in Google Scholar PubMed PubMed Central

Horst, B.G. and Marletta, M.A. (2018). Physiological activation and deactivation of soluble guanylate cyclase. Nitric Oxide 77, 65–74.10.1016/j.niox.2018.04.011Search in Google Scholar PubMed PubMed Central

Horst, B.G., Yokom, A.L., Rosenberg, D.J., Morris, K.L., Hammel, M., Hurley, J.H., and Marletta, M.A. (2019). Allosteric activation of the nitric oxide receptor soluble guanylate cyclase mapped by cryo-electron microscopy. eLife 8, pii: e50634.10.7554/eLife.50634.055Search in Google Scholar

Huynh, P. and Chai, Z. (2019). Transforming growth factor beta (TGFbeta) and related molecules in chronic kidney disease (CKD). Clin. Sci. (Lond). 133, 287–313.10.1042/CS20180438Search in Google Scholar PubMed

Inserte, J. and Garcia-Dorado, D. (2015). The cGMP/PKG pathway as a common mediator of cardioprotection: translatability and mechanism. Br. J. Pharmacol. 172, 1996–2009.10.1111/bph.12959Search in Google Scholar PubMed PubMed Central

Irvine, J.C., Ganthavee, V., Love, J.E., Alexander, A.E., Horowitz, J.D., Stasch, J.P., Kemp-Harper, B.K., and Ritchie, R.H. (2012). The soluble guanylyl cyclase activator bay 58-2667 selectively limits cardiomyocyte hypertrophy. PLoS One 7, e44481.10.1371/journal.pone.0044481Search in Google Scholar

Itoh, H., Pratt, R.E., and Dzau, V.J. (1990). Atrial natriuretic polypeptide inhibits hypertrophy of vascular smooth-muscle cells. J. Clin. Invest. 86, 1690–1697.10.1172/JCI114893Search in Google Scholar

Jaumann, M., Dettling, J., Gubelt, M., Zimmermann, U., Gerling, A., Paquet-Durand, F., Feil, S., Wolpert, S., Franz, C., Varakina, K., et al. (2012). cGMP-Prkg1 signaling and Pde5 inhibition shelter cochlear hair cells and hearing function. Nat. Med. 18, 252–259.10.1038/nm.2634Search in Google Scholar

Jin, J.G., Murthy, K.S., Grider, J.R., and Makhlouf, G.M. (1996). Stoichiometry of neurally induced VIP release, NO formation, and relaxation in rabbit and rat gastric muscle. Am. J. Physiol. 271, G357–G369.10.1152/ajpgi.1996.271.2.G357Search in Google Scholar

Juilfs, D.M., Fulle, H.J., Zhao, A.Z., Houslay, M.D., Garbers, D.L., and Beavo, J.A. (1997). A subset of olfactory neurons that selectively express cGMP-stimulated phosphodiesterase (PDE2) and guanylyl cyclase-D define a unique olfactory signal transduction pathway. Proc. Natl. Acad. Sci. U.S.A. 94, 3388–3395.10.1073/pnas.94.7.3388Search in Google Scholar

Kalderon, D. and Rubin, G.M. (1989). cGMP-dependent protein kinase genes in Drosophila. J. Biol. Chem. 264, 10738–10748.10.1016/S0021-9258(18)81684-2Search in Google Scholar

Kalyanaraman, H., Zhuang, S., Pilz, R.B., and Casteel, D.E. (2017). The activity of cGMP-dependent protein kinase Ialpha is not directly regulated by oxidation-induced disulfide formation at cysteine 43. J. Biol. Chem. 292, 8262–8268.10.1074/jbc.C117.787358Search in Google Scholar PubMed PubMed Central

Kang, Y., Liu, R., Wu, J.X., and Chen, L. (2019). Structural insights into the mechanism of human soluble guanylate cyclase. Nature 574, 206–210.10.1038/s41586-019-1584-6Search in Google Scholar PubMed

Kaupp, U.B. and Seifert, R. (2002). Cyclic nucleotide-gated ion channels. Physiol. Rev. 82, 769–824.10.1152/physrev.00008.2002Search in Google Scholar PubMed

Kawasaki, Y., Kugimiya, F., Chikuda, H., Kamekura, S., Ikeda, T., Kawamura, N., Saito, T., Shinoda, Y., Higashikawa, A., Yano, F., et al. (2008). Phosphorylation of GSK-3 beta by cGMP-dependent protein kinase II promotes hypertrophic differentiation of murine chondrocytes (vol 118, pg 2506, 2008). J. Clin. Invest. 118, 2986–2986.10.1172/JCI35243E1Search in Google Scholar

Kemp-Harper, B. and Schmidt, H.H. (2009). cGMP in the vasculature. Handb. Exp. Pharmacol. 191, 447–467.10.1007/978-3-540-68964-5_19Search in Google Scholar PubMed

Kessler, T., Wolf, B., Eriksson, N., Kofink, D., Mahmoodi, B.K., Rai, H., Tragante, V., Akerblom, A., Becker, R.C., Bernlochner, I., et al. (2019). Association of the coronary artery disease risk gene GUCY1A3 with ischaemic events after coronary intervention. Cardiovasc. Res. 115, 1512–1518.10.1093/cvr/cvz015Search in Google Scholar PubMed

Khalil, H., Kanisicak, O., Prasad, V., Correll, R.N., Fu, X., Schips, T., Vagnozzi, R.J., Liu, R., Huynh, T., Lee, S.J., et al. (2017). Fibroblast-specific TGF-beta-Smad2/3 signaling underlies cardiac fibrosis. J. Clin. Invest. 127, 3770–3783.10.1172/JCI94753Search in Google Scholar PubMed PubMed Central

Khavandi, K., Baylie, R.A., Sugden, S.A., Ahmed, M., Csato, V., Eaton, P., Hill-Eubanks, D.C., Bonev, A.D., Nelson, M.T., and Greenstein, A.S. (2016). Pressure-induced oxidative activation of PKG enables vasoregulation by Ca2+ sparks and BK channels. Sci. Signal. 9, ra100.10.1126/scisignal.aaf6625Search in Google Scholar PubMed PubMed Central

Kim, S., Titcombe, R.F., Zhang, H., Khatri, L., Girma, H.K., Hofmann, F., Arancio, O., and Ziff, E.B. (2015). Network compensation of cyclic GMP-dependent protein kinase II knockout in the hippocampus by Ca2+-permeable AMPA receptors. Proc. Natl. Acad. Sci. U.S.A. 112, 3122–3127.10.1073/pnas.1417498112Search in Google Scholar PubMed PubMed Central

Kim, C., Sarma, R., and Casteel, D. (2019). Crystal structure of PKG Ib holoenzyme reveals a trans-inhibiting dimer assembly. J. Transl. Med. 17, S2–06.10.1186/s12967-019-02198-7Search in Google Scholar

Kleppisch, T. (2009). Phosphodiesterases in the central nervous system. Handb. Exp. Pharmacol. 191, 71–92.10.1007/978-3-540-68964-5_5Search in Google Scholar PubMed

Kleppisch, T. and Feil, R. (2009). cGMP signalling in the mammalian brain: role in synaptic plasticity and behaviour. Handb. Exp. Pharmacol. 191, 549–579.10.1007/978-3-540-68964-5_24Search in Google Scholar PubMed

Kleppisch, T., Pfeifer, A., Klatt, P., Ruth, P., Montkowski, A., Fassler, R., and Hofmann, F. (1999). Long-term potentiation in the hippocampal CA1 region of mice lacking cGMP-dependent kinases is normal and susceptible to inhibition of nitric oxide synthase. J. Neurosci. 19, 48–55.10.1523/JNEUROSCI.19-01-00048.1999Search in Google Scholar

Koesling, D., Bohme, E., and Schultz, G. (1991). Guanylyl cyclases, a growing family of signal-transducing enzymes. FASEB J. 5, 2785–2791.10.1096/fasebj.5.13.1680765Search in Google Scholar PubMed

Koesling, D., Mergia, E., and Russwurm, M. (2016). Physiological functions of NO-sensitive guanylyl cyclase isoforms. Curr. Med. Chem. 23, 2653–2665.10.2174/0929867323666160812145050Search in Google Scholar PubMed

Kokkonen, K. and Kass, D.A. (2017). Nanodomain regulation of cardiac cyclic nucleotide signaling by phosphodiesterases. Annu. Rev. Pharmacol. Toxicol. 57, 455–479.10.1146/annurev-pharmtox-010716-104756Search in Google Scholar

Krawutschke, C., Koesling, D., and Russwurm, M. (2015). Cyclic GMP in vascular relaxation: export is of similar importance as degradation. Arterioscler. Thromb. Vasc. Biol. 35, 2011–2019.10.1161/ATVBAHA.115.306133Search in Google Scholar

Kuhn, M. (2003). Structure, regulation, and function of mammalian membrane guanylyl cyclase receptors, with a focus on guanylyl cyclase-A. Circ. Res. 93, 700–709.10.1161/01.RES.0000094745.28948.4DSearch in Google Scholar

Kuhn, M. (2016). Molecular physiology of membrane guanylyl cyclase receptors. Physiol. Rev. 96, 751–804.10.1152/physrev.00022.2015Search in Google Scholar

Kumar, R., Joyner, R.W., Komalavilas, P., and Lincoln, T.M. (1999). Analysis of expression of cGMP-dependent protein kinase in rabbit heart cells. J. Pharmacol. Exp. Ther. 291, 967–975.Search in Google Scholar

Kurtz, A., Gotz, K.H., Hamann, M., and Wagner, C. (1998). Stimulation of renin secretion by nitric oxide is mediated by phosphodiesterase 3. Proc. Natl. Acad. Sci. U.S.A. 95, 4743–4747.10.1073/pnas.95.8.4743Search in Google Scholar

Landgraf, W., Regulla, S., Meyer, H.E., and Hofmann, F. (1991). Oxidation of cysteines activates cGMP-dependent protein kinase. J. Biol. Chem. 266, 16305–16311.10.1016/S0021-9258(18)55297-2Search in Google Scholar

Landgraf, W., Ruth, P., Keilbach, A., May, B., Welling, A., and Hofmann, F. (1992). Cyclic GMP-dependent protein kinase and smooth muscle relaxation. J. Cardiovasc. Pharmacol. 20(Suppl 1), S18–22.10.1097/00005344-199212001-00005Search in Google Scholar

Legeai-Mallet, L. (2016). C-type natriuretic peptide analog as therapy for achondroplasia. Adv. Ther. Pediatr. Endocrinol. Diabetol. 30, 98–105.10.1159/000439334Search in Google Scholar PubMed

Leinders-Zufall, T., Cockerham, R.E., Michalakis, S., Biel, M., Garbers, D.L., Reed, R.R., Zufall, F., and Munger, S.D. (2007). Contribution of the receptor guanylyl cyclase GC-D to chemosensory function in the olfactory epithelium. Proc. Natl. Acad. Sci. U.S.A. 104, 14507–14512.10.1073/pnas.0704965104Search in Google Scholar PubMed PubMed Central

Lembo, A.J., Schneier, H.A., Shiff, S.J., Kurtz, C.B., MacDougall, J.E., Jia, X.D., Shao, J.Z., Lavins, B.J., Currie, M.G., Fitch, D.A., et al. (2011). Two randomized trials of linaclotide for chronic constipation. N. Engl. J. Med. 365, 527–536.10.1056/NEJMoa1010863Search in Google Scholar PubMed

Lev-Ram, V., Makings, L.R., Keitz, P.F., Kao, J.P., and Tsien, R.Y. (1995). Long-term depression in cerebellar Purkinje neurons results from coincidence of nitric oxide and depolarization-induced Ca2+ transients. Neuron 15, 407–415.10.1016/0896-6273(95)90044-6Search in Google Scholar

Lev-Ram, V., Nebyelul, Z., Ellisman, M.H., Huang, P.L., and Tsien, R.Y. (1997). Absence of cerebellar long-term depression in mice lacking neuronal nitric oxide synthase. Learn. Mem. 4, 169–177.10.1101/lm.4.1.169Search in Google Scholar PubMed

Lewin, M.R. and Walters, E.T. (1999). Cyclic GMP pathway is critical for inducing long-term sensitization of nociceptive sensory neurons. Nat. Neurosci. 2, 18–23.10.1038/4520Search in Google Scholar PubMed

Lodish, H., Berk, A., Kaiser, C., Krieger, M., Bretscher, A., Ploegh, H., Amon, A., and Martin, K.C.M. (2016). Molecular Cell Biology, 8th ed. (New York, NY, USA: Macmillan).Search in Google Scholar

Lohmann, S.M., Walter, U., Miller, P.E., Greengard, P., and De Camilli, P. (1981). Immunohistochemical localization of cyclic GMP-dependent protein kinase in mammalian brain. Proc. Natl. Acad. Sci. U.S.A. 78, 653–657.10.1073/pnas.78.1.653Search in Google Scholar PubMed PubMed Central

Loscalzo, J. (2001). Nitric oxide insufficiency, platelet activation, and arterial thrombosis. Circ. Res. 88, 756–762.10.1161/hh0801.089861Search in Google Scholar PubMed

Lukowski, R., Weinmeister, P., Bernhard, D., Feil, S., Gotthardt, M., Herz, J., Massberg, S., Zernecke, A., Weber, C., Hofmann, F., et al. (2008). Role of smooth muscle cGMP/cGKI signaling in murine vascular restenosis. Arterioscler. Thromb. Vasc. Biol. 28, 1244–1250.10.1161/ATVBAHA.108.166405Search in Google Scholar PubMed PubMed Central

Lukowski, R., Rybalkin, S.D., Loga, F., Leiss, V., Beavo, J.A., and Hofmann, F. (2010). Cardiac hypertrophy is not amplified by deletion of cGMP-dependent protein kinase I in cardiomyocytes. Proc. Natl. Acad. Sci. U.S.A. 107, 5646–5651.10.1073/pnas.1001360107Search in Google Scholar PubMed PubMed Central

Lukowski, R., Krieg, T., Rybalkin, S.D., Beavo, J., and Hofmann, F. (2014). Turning on cGMP-dependent pathways to treat cardiac dysfunctions: boom, bust, and beyond. Trends Pharmacol. Sci. 35, 404–413.10.1016/j.tips.2014.05.003Search in Google Scholar PubMed

Luo, C., Gangadharan, V., Bali, K.K., Xie, R.G., Agarwal, N., Kurejova, M., Tappe-Theodor, A., Tegeder, I., Feil, S., Lewin, G., et al. (2012). Presynaptically localized cyclic GMP-dependent protein kinase 1 is a key determinant of spinal synaptic potentiation and pain hypersensitivity. PLoS Biol. 10, e1001283.10.1371/journal.pbio.1001283Search in Google Scholar PubMed PubMed Central

Makhoul, S., Walter, E., Pagel, O., Walter, U., Sickmann, A., Gambaryan, S., Smolenski, A., Zahedi, R.P., and Jurk, K. (2018). Effects of the NO/soluble guanylate cyclase/cGMP system on the functions of human platelets. Nitric Oxide 76, 71–80.10.1016/j.niox.2018.03.008Search in Google Scholar PubMed

Markert, T., Vaandrager, A.B., Gambaryan, S., Pohler, D., Hausler, C., Walter, U., De Jonge, H.R., Jarchau, T., and Lohmann, S.M. (1995). Endogenous expression of type II cGMP-dependent protein kinase mRNA and protein in rat intestine. Implications for cystic fibrosis transmembrane conductance regulator. J. Clin. Invest. 96, 822–830.10.1172/JCI118128Search in Google Scholar

Massberg, S., Sausbier, M., Klatt, P., Bauer, M., Pfeifer, A., Siess, W., Fassler, R., Ruth, P., Krombach, F., and Hofmann, F. (1999). Increased adhesion and aggregation of platelets lacking cyclic guanosine 3′,5′-monophosphate kinase I. J. Exp. Med. 189, 1255–1264.10.1084/jem.189.8.1255Search in Google Scholar

Matei, A.E., Beyer, C., Gyorfi, A.H., Soare, A., Chen, C.W., Dees, C., Bergmann, C., Ramming, A., Friebe, A., Hofmann, F., et al. (2018). Protein kinases G are essential downstream mediators of the antifibrotic effects of sGC stimulators. Ann. Rheum. Dis. 77, 459.10.1136/annrheumdis-2017-212489Search in Google Scholar

Matsue, Y., Kagiyama, N., Yoshida, K., Kume, T., Okura, H., Suzuki, M., Matsumura, A., Yoshida, K., and Hashimoto, Y. (2015). Carperitide is associated with increased in-hospital mortality in acute heart failure: a propensity score-matched analysis. J. Card. Fail. 21, 859–864.10.1016/j.cardfail.2015.05.007Search in Google Scholar

McMurray, J.J., Packer, M., Desai, A.S., Gong, J., Lefkowitz, M.P., Rizkala, A.R., Rouleau, J.L., Shi, V.C., Solomon, S.D., Swedberg, K., et al. (2014). Angiotensin-neprilysin inhibition versus enalapril in heart failure. N. Engl. J. Med. 371, 993–1004.10.1056/NEJMoa1409077Search in Google Scholar

Meng, X.M., Nikolic-Paterson, D.J., and Lan, H.Y. (2016). TGF-beta: the master regulator of fibrosis. Nat. Rev. Nephrol. 12, 325–338.10.1038/nrneph.2016.48Search in Google Scholar

Menges, L., Krawutschke, C., Fuchtbauer, E.M., Fuchtbauer, A., Sandner, P., Koesling, D., and Russwurm, M. (2019). Mind the gap [junction]: NO-induced cGMP in cardiac myocytes originates from fibroblasts. Br J Pharmacol. Doi: 10.1111/bph.14835.10.1111/bph.14835Search in Google Scholar

Menniti, F.S., Faraci, W.S., and Schmidt, C.J. (2006). Phosphodiesterases in the CNS: targets for drug development. Nat. Rev. Drug Discov. 5, 660–670.10.1038/nrd2058Search in Google Scholar

Mergia, E., Russwurm, M., Zoidl, G., and Koesling, D. (2003). Major occurrence of the new alpha2beta1 isoform of NO-sensitive guanylyl cyclase in brain. Cell Signal. 15, 189–195.10.1016/S0898-6568(02)00078-5Search in Google Scholar

Methner, C., Buonincontri, G., Hu, C.H., Vujic, A., Kretschmer, A., Sawiak, S., Carpenter, A., Stasch, J.P., and Krieg, T. (2013). Riociguat reduces infarct size and post-infarct heart failure in mouse hearts: insights from MRI/PET imaging. PLoS One 8, e83910.10.1371/journal.pone.0083910Search in Google Scholar PubMed PubMed Central

Michalakis, S., Becirovic, E., and Biel, M. (2018). Retinal cyclic nucleotide-gated channels: from pathophysiology to therapy. Int. J. Mol. Sci. 19.10.3390/ijms19030749Search in Google Scholar

Milewicz, D. and Regalado, E. (2017). Heritable thoracic aortic disease overview. GeneReviews® [Internet]. Book.Search in Google Scholar

Milewicz, D.M., Guo, D.C., Tran-Fadulu, V., Lafont, A.L., Papke, C.L., Inamoto, S., Kwartler, C.S., and Pannu, H. (2008). Genetic basis of thoracic aortic aneurysms and dissections: focus on smooth muscle cell contractile dysfunction. Annu. Rev. Genomics Hum. Genet. 9, 283–302.10.1146/annurev.genom.8.080706.092303Search in Google Scholar

Moyes, A.J., Khambata, R.S., Villar, I., Bubb, K.J., Baliga, R.S., Lumsden, N.G., Xiao, F., Gane, P.J., Rebstock, A.S., Worthington, R.J., et al. (2014). Endothelial C-type natriuretic peptide maintains vascular homeostasis. J. Clin. Invest. 124, 4039–4051.10.1172/JCI74281Search in Google Scholar

Murrel, W. (1879). Nitro-glycerin as a remedy for angina pectoris. Lancet 113, 80–81.10.1016/S0140-6736(02)46032-1Search in Google Scholar

Neitz, A., Mergia, E., Eysel, U.T., Koesling, D., and Mittmann, T. (2011). Presynaptic nitric oxide/cGMP facilitates glutamate release via hyperpolarization-activated cyclic nucleotide-gated channels in the hippocampus. Eur. J. Neurosci. 33, 1611–1621.10.1111/j.1460-9568.2011.07654.xSearch in Google Scholar PubMed

Neitz, A., Mergia, E., Imbrosci, B., Petrasch-Parwez, E., Eysel, U.T., Koesling, D., and Mittmann, T. (2014). Postsynaptic NO/cGMP increases NMDA receptor currents via hyperpolarization-activated cyclic nucleotide-gated channels in the hippocampus. Cereb Cortex. 24, 1923–1936.10.1093/cercor/bht048Search in Google Scholar PubMed

Neitz, A., Mergia, E., Neubacher, U., Koesling, D., and Mittmann, T. (2015). NO regulates the strength of synaptic inputs onto hippocampal CA1 neurons via NO-GC1/cGMP signalling. Pflüger’s Arch. 467, 1383–1394.10.1007/s00424-014-1571-6Search in Google Scholar PubMed

Nishida, M., Nishimura, A., Matsunaga, T., Motohashi, H., Kasamatsu, S., and Akaike, T. (2017). Redox regulation of electrophilic signaling by reactive persulfides in cardiac cells. Free Radic. Biol. Med. 109, 132–140.10.1016/j.freeradbiomed.2017.01.024Search in Google Scholar PubMed

O’Connor, C.M., Starling, R.C., Hernandez, A.F., Armstrong, P.W., Dickstein, K., Hasselblad, V., Heizer, G.M., Komajda, M., Massie, B.M., McMurray, J.J., et al. (2011). Effect of nesiritide in patients with acute decompensated heart failure. N. Engl. J. Med. 365, 32–43.10.1056/NEJMoa1100171Search in Google Scholar PubMed

O’Dell, T.J., Hawkins, R.D., Kandel, E.R., and Arancio, O. (1991). Tests of the roles of two diffusible substances in long-term potentiation: evidence for nitric oxide as a possible early retrograde messenger. Proc. Natl. Acad. Sci. U.S.A. 88, 11285–11289.10.1073/pnas.88.24.11285Search in Google Scholar PubMed PubMed Central

Oldenburg, O., Qin, Q., Krieg, T., Yang, X.M., Philipp, S., Critz, S.D., Cohen, M.V., and Downey, J.M. (2004). Bradykinin induces mitochondrial ROS generation via NO, cGMP, PKG, and mitoKATP channel opening and leads to cardioprotection. Am. J. Physiol. Heart Circ. Physiol. 286, H468–H476.10.1152/ajpheart.00360.2003Search in Google Scholar

Osborne, K.A., Robichon, A., Burgess, E., Butland, S., Shaw, R.A., Coulthard, A., Pereira, H.S., Greenspan, R.J., and Sokolowski, M.B. (1997). Natural behavior polymorphism due to a cGMP-dependent protein kinase of Drosophila. Science 277, 834–836.10.1126/science.277.5327.834Search in Google Scholar

Osborne, B.W., Wu, J., McFarland, C.J., Nickl, C.K., Sankaran, B., Casteel, D.E., Woods, V.L., Jr., Kornev, A.P., Taylor, S.S., and Dostmann, W.R. (2011). Crystal structure of cGMP-dependent protein kinase reveals novel site of interchain communication. Structure 19, 1317–1327.10.1016/j.str.2011.06.012Search in Google Scholar

Oster, H., Werner, C., Magnone, M.C., Mayser, H., Feil, R., Seeliger, M.W., Hofmann, F., and Albrecht, U. (2003). cGMP-dependent protein kinase II modulates mPer1 and mPer2 gene induction and influences phase shifts of the circadian clock. Curr. Biol. 13, 725–733.10.1016/S0960-9822(03)00252-5Search in Google Scholar

Patrucco, E., Domes, K., Sbroggio, M., Blaich, A., Schlossmann, J., Desch, M., Rybalkin, S.D., Beavo, J.A., Lukowski, R., and Hofmann, F. (2014). Roles of cGMP-dependent protein kinase I (cGKI) and PDE5 in the regulation of Ang II-induced cardiac hypertrophy and fibrosis. Proc. Natl. Acad. Sci. U.S.A. 111, 12925–12929.10.1073/pnas.1414364111Search in Google Scholar PubMed PubMed Central

Penzo, M., de Las Heras-Duena, L., Mata-Cantero, L., Diaz-Hernandez, B., Vazquez-Muniz, M.J., Ghidelli-Disse, S., Drewes, G., Fernandez-Alvaro, E., and Baker, D.A. (2019). High-throughput screening of the Plasmodium falciparum cGMP-dependent protein kinase identified a thiazole scaffold which kills erythrocytic and sexual stage parasites. Sci. Rep. 9, 7005.10.1038/s41598-019-42801-xSearch in Google Scholar PubMed PubMed Central

Pfeifer, A., Aszodi, A., Seidler, U., Ruth, P., Hofmann, F., and Fassler, R. (1996). Intestinal secretory defects and dwarfism in mice lacking cGMP-dependent protein kinase II. Science 274, 2082–2086.10.1126/science.274.5295.2082Search in Google Scholar PubMed

Pfeifer, A., Klatt, P., Massberg, S., Ny, L., Sausbier, M., Hirneiss, C., Wang, G.X., Korth, M., Aszodi, A., Andersson, K.E., et al. (1998). Defective smooth muscle regulation in cGMP kinase I-deficient mice. EMBO J. 17, 3045–3051.10.1093/emboj/17.11.3045Search in Google Scholar PubMed PubMed Central

Polimanti, R., Kaufman, J., Zhao, H., Kranzler, H.R., Ursano, R.J., Kessler, R.C., Gelernter, J., and Stein, M.B. (2018). A genome-wide gene-by-trauma interaction study of alcohol misuse in two independent cohorts identifies PRKG1 as a risk locus. Mol. Psychiatry. 23, 154–160.10.1038/mp.2017.24Search in Google Scholar PubMed PubMed Central

Potter, L.R. (2011). Guanylyl cyclase structure, function and regulation. Cell Signal. 23, 1921–1926.10.1016/j.cellsig.2011.09.001Search in Google Scholar PubMed PubMed Central

Potter, L.R. and Garbers, D.L. (1992). Dephosphorylation of the guanylyl cyclase-A receptor causes desensitization. J. Biol. Chem. 267, 14531–14534.10.1016/S0021-9258(18)42069-8Search in Google Scholar

Potter, L.R. and Hunter, T. (1998). Phosphorylation of the kinase homology domain is essential for activation of the A-type natriuretic peptide receptor. Mol. Cell. Biol. 18, 2164–2172.10.1128/MCB.18.4.2164Search in Google Scholar PubMed PubMed Central

Potter, L.R., Abbey-Hosch, S., and Dickey, D.M. (2006). Natriuretic peptides, their receptors, and cyclic guanosine monophosphate-dependent signaling functions. Endocr. Rev. 27, 47–72.10.1210/er.2005-0014Search in Google Scholar PubMed

Power, M., Das, S., Schutze, K., Marigo, V., Ekstrom, P., and Paquet-Durand, F. (2019). Cellular mechanisms of hereditary photoreceptor degeneration – focus on cGMP. Prog. Retin. Eye Res. 100772. Doi: 10.1016/j.preteyeres.2019.07.005. [Epub ahead of print]10.1016/j.preteyeres.2019.07.005Search in Google Scholar PubMed

Prysyazhna, O., Rudyk, O., and Eaton, P. (2012). Single atom substitution in mouse protein kinase G eliminates oxidant sensing to cause hypertension. Nat. Med. 18, 286–290.10.1038/nm.2603Search in Google Scholar PubMed PubMed Central

Rainer, P.P. and Kass, D.A. (2016). Old dog, new tricks: novel cardiac targets and stress regulation by protein kinase G. Cardiovasc. Res. 111, 154-162.10.1093/cvr/cvw107Search in Google Scholar PubMed PubMed Central

Rainer, P.P., Hao, S., Vanhoutte, D., Lee, D.I., Koitabashi, N., Molkentin, J.D., and Kass, D.A. (2014). Cardiomyocyte-specific transforming growth factor beta suppression blocks neutrophil infiltration, augments multiple cytoprotective cascades, and reduces early mortality after myocardial infarction. Circ. Res. 114, 1246–1257.10.1161/CIRCRESAHA.114.302653Search in Google Scholar PubMed PubMed Central

Rappaport, J.A. and Waldman, S.A. (2018). The guanylate cyclase C-cGMP signaling axis opposes intestinal epithelial injury and neoplasia. Front. Oncol. 8, 299.10.3389/fonc.2018.00299Search in Google Scholar PubMed PubMed Central

Reaume, C.J. and Sokolowski, M.B. (2009). cGMP-dependent protein kinase as a modifier of behaviour. Handb. Exp. Pharmacol. 191, 423–443.10.1007/978-3-540-68964-5_18Search in Google Scholar PubMed

Redfield, M.M., Chen, H.H., Borlaug, B.A., Semigran, M.J., Lee, K.L., Lewis, G., LeWinter, M.M., Rouleau, J.L., Bull, D.A., Mann, D.L., et al. (2013). Effect of phosphodiesterase-5 inhibition on exercise capacity and clinical status in heart failure with preserved ejection fraction: a randomized clinical trial. J. Am. Med. Assoc. 309, 1268–1277.10.1001/jama.2013.2024Search in Google Scholar PubMed PubMed Central

Reinke, Y., Gross, S., Eckerle, L.G., Hertrich, I., Busch, M., Busch, R., Riad, A., Rauch, B.H., Stasch, J.P., Dorr, M., et al. (2015). The soluble guanylate cyclase stimulator riociguat and the soluble guanylate cyclase activator cinaciguat exert no direct effects on contractility and relaxation of cardiac myocytes from normal rats. Eur. J. Pharmacol. 767, 1–9.10.1016/j.ejphar.2015.09.022Search in Google Scholar PubMed

Rosenwasser, A.M. and Turek, F.W. (2015). Neurobiology of circadian rhythm regulation. Sleep Med. Clin. 10, 403–412.10.1016/j.jsmc.2015.08.003Search in Google Scholar PubMed

Rothstein, R.D. and Friedenberg, F.K. (2013). Linaclotide: a novel compound for the treatment of irritable bowel syndrome with constipation. Expert Opin. Pharmacother. 14, 2125–2132.10.1517/14656566.2013.833605Search in Google Scholar PubMed

Ruth, P., Pfeifer, A., Kamm, S., Klatt, P., Dostmann, W.R., and Hofmann, F. (1997). Identification of the amino acid sequences responsible for high affinity activation of cGMP kinase Ialpha. J. Biol. Chem. 272, 10522–10528.10.1074/jbc.272.16.10522Search in Google Scholar PubMed

Rybalkin, S.D., Rybalkina, I.G., Feil, R., Hofmann, F., and Beavo, J.A. (2002). Regulation of cGMP-specific phosphodiesterase (PDE5) phosphorylation in smooth muscle cells. J. Biol. Chem. 277, 3310–3317.10.1074/jbc.M106562200Search in Google Scholar PubMed

Sanders, K.M. and Ward, S.M. (1992). Nitric-oxide as a mediator of nonadrenergic noncholinergic neurotransmission. Am. J. Physiol. 262, G379–G392.10.1152/ajpgi.1992.262.3.G379Search in Google Scholar PubMed

Sandner, P. (2018). From molecules to patients: exploring the therapeutic role of soluble guanylate cyclase stimulators. Biol. Chem. 399, 679–690.10.1515/hsz-2018-0155Search in Google Scholar PubMed

Sandner, P. and Stasch, J.P. (2017). Anti-fibrotic effects of soluble guanylate cyclase stimulators and activators: a review of the preclinical evidence. Respir. Med. 122(Suppl 1), S1–S9.10.1016/j.rmed.2016.08.022Search in Google Scholar PubMed

Sandner, P., Zimmer, D.P., Milne, G.T., Follmann, M., Hobbs, A., and Stasch, J.-P. (2019). Soluble guanylate cyclase stimulators and activators. Handb Exp Pharmacol. Doi: 10.1007/164_2019_249.10.1007/164_2019_249Search in Google Scholar PubMed

Sausbier, M., Schubert, R., Voigt, V., Hirneiss, C., Pfeifer, A., Korth, M., Kleppisch, T., Ruth, P., and Hofmann, F. (2000). Mechanisms of NO/cGMP-dependent vasorelaxation. Circ. Res. 87, 825–830.10.1161/01.RES.87.9.825Search in Google Scholar PubMed

Savarirayan, R., Irving, M., Bacino, C.A., Bostwick, B., Charrow, J., Cormier-Daire, V., Le Quan Sang, K.H., Dickson, P., Harmatz, P., Phillips, J., et al. (2019). C-type natriuretic peptide analogue therapy in children with achondroplasia. N. Engl. J. Med. 381, 25–35.10.1056/NEJMoa1813446Search in Google Scholar PubMed

Schermuly, R.T., Ghofrani, H.A., Wilkins, M.R., and Grimminger, F. (2011). Mechanisms of disease: pulmonary arterial hypertension. Nat. Rev. Cardiol. 8, 443–455.10.1038/nrcardio.2011.87Search in Google Scholar PubMed PubMed Central

Schinner, E., Wetzl, V., Schramm, A., Kees, F., Sandner, P., Stasch, J.P., Hofmann, F., and Schlossmann, J. (2017). Inhibition of the TGFbeta signalling pathway by cGMP and cGMP-dependent kinase I in renal fibrosis. FEBS Open Bio. 7, 550–561.10.1002/2211-5463.12202Search in Google Scholar PubMed PubMed Central

Schlossmann, J., Ammendola, A., Ashman, K., Zong, X., Huber, A., Neubauer, G., Wang, G.X., Allescher, H.D., Korth, M., Wilm, M., et al. (2000). Regulation of intracellular calcium by a signalling complex of IRAG, IP3 receptor and cGMP kinase Ibeta. Nature 404, 197–201.10.1038/35004606Search in Google Scholar PubMed

Schmidt, H., Werner, M., Heppenstall, P.A., Henning, M., More, M.I., Kuhbandner, S., Lewin, G.R., Hofmann, F., Feil, R., and Rathjen, F.G. (2002). cGMP-mediated signaling via cGKIalpha is required for the guidance and connectivity of sensory axons. J. Cell. Biol. 159, 489–498.10.1083/jcb.200207058Search in Google Scholar PubMed PubMed Central

Schmidt, H., Stonkute, A., Juttner, R., Koesling, D., Friebe, A., and Rathjen, F.G. (2009). C-type natriuretic peptide (CNP) is a bifurcation factor for sensory neurons. Proc. Natl. Acad. Sci. U.S.A. 106, 16847–16852.10.1073/pnas.0906571106Search in Google Scholar PubMed PubMed Central

Schmidtko, A., Tegeder, I., and Geisslinger, G. (2009). No NO, no pain? The role of nitric oxide and cGMP in spinal pain processing. Trends Neurosci. 32, 339–346.10.1016/j.tins.2009.01.010Search in Google Scholar PubMed

Schon, C., Biel, M., and Michalakis, S. (2013). Gene replacement therapy for retinal CNG channelopathies. Mol. Genet. Genomics 288, 459–467.10.1007/s00438-013-0766-4Search in Google Scholar PubMed

Schramm, A., Schweda, F., Sequeira-Lopez, M.L.S., Hofmann, F., Sandner, P., and Schlossmann, J. (2019). Protein kinase G is involved in acute but not in long-term regulation of renin secretion. Front. Pharmacol. 10, 800.10.3389/fphar.2019.00800Search in Google Scholar PubMed PubMed Central

Schrammel, A., Behrends, S., Schmidt, K., Koesling, D., and Mayer, B. (1996). Characterization of 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one as a heme-site inhibitor of nitric oxide-sensitive guanylyl cyclase. Mol. Pharmacol. 50, 1–5.Search in Google Scholar

Schulz, S., Wedel, B.J., Matthews, A., and Garbers, D.L. (1998). The cloning and expression of a new guanylyl cyclase orphan receptor. J. Biol. Chem. 273, 1032–1037.10.1074/jbc.273.2.1032Search in Google Scholar PubMed

Seftel, A.D. (2004). Phosphodiesterase type 5 inhibitor differentiation based on selectivity, pharmacokinetic, and efficacy profiles. Clin. Cardiol. 27, I14–I19.10.1002/clc.4960271305Search in Google Scholar PubMed PubMed Central

Serulle, Y., Zhang, S., Ninan, I., Puzzo, D., McCarthy, M., Khatri, L., Arancio, O., and Ziff, E.B. (2007). A GluR1-cGKII interaction regulates AMPA receptor trafficking. Neuron 56, 670–688.10.1016/j.neuron.2007.09.016Search in Google Scholar PubMed PubMed Central

Sheehe, J.L., Bonev, A.D., Schmoker, A.M., Ballif, B.A., Nelson, M.T., Moon, T.M., and Dostmann, W.R. (2018). Oxidation of cysteine 117 stimulates constitutive activation of the type I alpha cGMP-dependent protein kinase. J. Biol. Chem. 293, 16791–16802.10.1074/jbc.RA118.004363Search in Google Scholar PubMed PubMed Central

Singh, A.K., Spiessberger, B., Zheng, W., Xiao, F., Lukowski, R., Wegener, J.W., Weinmeister, P., Saur, D., Klein, S., Schemann, M., et al. (2012). Neuronal cGMP kinase I is essential for stimulation of duodenal bicarbonate secretion by luminal acid. FASEB J. 26, 1745–1754.10.1096/fj.11-200394Search in Google Scholar PubMed

Somlyo, A.P. and Somlyo, A.V. (2003). Ca2+ sensitivity of smooth muscle and nonmuscle myosin II: modulated by G proteins, kinases, and myosin phosphatase. Physiol. Rev. 83, 1325–1358.10.1152/physrev.00023.2003Search in Google Scholar PubMed

Stasch, J.P., Becker, E.M., Alonso-Alija, C., Apeler, H., Dembowsky, K., Feurer, A., Gerzer, R., Minuth, T., Perzborn, E., Pleiss, U., et al. (2001). NO-independent regulatory site on soluble guanylate cyclase. Nature 410, 212–215.10.1038/35065611Search in Google Scholar PubMed

Stasch, J.P., Schlossmann, J., and Hocher, B. (2015). Renal effects of soluble guanylate cyclase stimulators and activators: a review of the preclinical evidence. Curr. Opin. Pharmacol. 21, 95–104.10.1016/j.coph.2014.12.014Search in Google Scholar PubMed

Struk, A.A., Mugon, J., Huston, A., Scholer, A.A., Stadler, G., Higgins, E.T., Sokolowski, M.B., and Danckert, J. (2019). Self-regulation and the foraging gene (PRKG1) in humans. Proc. Natl. Acad. Sci. U.S.A. 116, 4434–4439.10.1073/pnas.1809924116Search in Google Scholar PubMed PubMed Central

Su, T., Zhang, T., Xie, S., Yan, J., Wu, Y., Li, X., Huang, L., and Luo, H.B. (2016). Discovery of novel PDE9 inhibitors capable of inhibiting Abeta aggregation as potential candidates for the treatment of Alzheimer’s disease. Sci. Rep. 6, 21826.10.1038/srep21826Search in Google Scholar PubMed PubMed Central

Suda, M., Ogawa, Y., Tanaka, K., Tamura, N., Yasoda, A., Takigawa, T., Uehira, M., Nishimoto, H., Itoh, H., Saito, Y., et al. (1998). Skeletal overgrowth in transgenic mice that overexpress brain natriuretic peptide. Proc. Natl. Acad. Sci. U.S.A. 95, 2337–2342.10.1073/pnas.95.5.2337Search in Google Scholar PubMed PubMed Central

Surks, H.K., Mochizuki, N., Kasai, Y., Georgescu, S.P., Tang, K.M., Ito, M., Lincoln, T.M., and Mendelsohn, M.E. (1999). Regulation of myosin phosphatase by a specific interaction with cGMP- dependent protein kinase Ialpha. Science 286, 1583–1587.10.1126/science.286.5444.1583Search in Google Scholar

Takimoto, E. (2012). Cyclic GMP-dependent signaling in cardiac myocytes. Circ. J. 76, 1819–1825.10.1253/circj.CJ-12-0664Search in Google Scholar

Takimoto, E., Champion, H.C., Li, M., Belardi, D., Ren, S., Rodriguez, E.R., Bedja, D., Gabrielson, K.L., Wang, Y., and Kass, D.A. (2005). Chronic inhibition of cyclic GMP phosphodiesterase 5A prevents and reverses cardiac hypertrophy. Nat. Med. 11, 214–222.10.1038/nm1175Search in Google Scholar

Tamura, N., Ogawa, Y., Chusho, H., Nakamura, K., Nakao, K., Suda, M., Kasahara, M., Hashimoto, R., Katsuura, G., Mukoyama, M., et al. (2000). Cardiac fibrosis in mice lacking brain natriuretic peptide. Proc. Natl. Acad. Sci. U.S.A. 97, 4239–4244.10.1073/pnas.070371497Search in Google Scholar

Tang, K.M., Wang, G.R., Lu, P., Karas, R.H., Aronovitz, M., Heximer, S.P., Kaltenbronn, K.M., Blumer, K.J., Siderovski, D.P., Zhu, Y., et al. (2003). Regulator of G-protein signaling-2 mediates vascular smooth muscle relaxation and blood pressure. Nat. Med. 9, 1506–1512.10.1038/nm958Search in Google Scholar

Tegeder, I., Del Turco, D., Schmidtko, A., Sausbier, M., Feil, R., Hofmann, F., Deller, T., Ruth, P., and Geisslinger, G. (2004). Reduced inflammatory hyperalgesia with preservation of acute thermal nociception in mice lacking cGMP-dependent protein kinase I. Proc. Natl. Acad. Sci. U.S.A. 101, 3253–3257.10.1073/pnas.0304076101Search in Google Scholar

Thomas, M.K., Francis, S.H., and Corbin, J.D. (1990). Substrate- and kinase-directed regulation of phosphorylation of a cGMP-binding phosphodiesterase by cGMP. J. Biol. Chem. 265, 14971–14978.10.1016/S0021-9258(18)77211-6Search in Google Scholar

Thunemann, M., Wen, L., Hillenbrand, M., Vachaviolos, A., Feil, S., Ott, T., Han, X., Fukumura, D., Jain, R.K., Russwurm, M., et al. (2013). Transgenic mice for cGMP imaging. Circ. Res. 113, 365–371.10.1161/CIRCRESAHA.113.301063Search in Google Scholar PubMed PubMed Central

Tischkau, S.A., Weber, E.T., Abbott, S.M., Mitchell, J.W., and Gillette, M.U. (2003). Circadian clock-controlled regulation of cGMP-protein kinase G in the nocturnal domain. J. Neurosci. 23, 7543–7550.10.1523/JNEUROSCI.23-20-07543.2003Search in Google Scholar

Turunen, T. and Koskolainen, A. (2019). Determination of basal phosphodiesterase activity in mouse rod photoreceptors with cGMP clamp. Sci. Rep. 9, 1183.10.1038/s41598-018-37661-wSearch in Google Scholar PubMed PubMed Central

Uckert, S. and Oelke, M. (2011). Phosphodiesterase (PDE) inhibitors in the treatment of lower urinary tract dysfunction. Br. J. Clin. Pharmacol. 72, 197–204.10.1111/j.1365-2125.2010.03828.xSearch in Google Scholar PubMed PubMed Central

Vaandrager, A.B., Smolenski, A., Tilly, B.C., Houtsmuller, A.B., Ehlert, E.M., Bot, A.G., Edixhoven, M., Boomaars, W.E., Lohmann, S.M., and de Jonge, H.R. (1998). Membrane targeting of cGMP-dependent protein kinase is required for cystic fibrosis transmembrane conductance regulator Cl- channel activation. Proc. Natl. Acad. Sci. U.S.A. 95, 1466–1471.10.1073/pnas.95.4.1466Search in Google Scholar PubMed PubMed Central

Vandecasteele, G., Eschenhagen, T., Scholz, H., Stein, B., Verde, I., and Fischmeister, R. (1999). Muscarinic and beta-adrenergic regulation of heart rate, force of contraction and calcium current is preserved in mice lacking endothelial nitric oxide synthase. Nat. Med. 5, 331–334.10.1038/6553Search in Google Scholar PubMed

von Euler, U. and Liljestrand, G. (1946). Observations on the pulmonary arterial blood pressure in the cat. Acta Physiol. Scand. 12, 301–320.10.1111/j.1748-1716.1946.tb00389.xSearch in Google Scholar

Wagner, C., Pfeifer, A., Ruth, P., Hofmann, F., and Kurtz, A. (1998). Role of cGMP-kinase II in the control of renin secretion and renin expression. J. Clin. Invest. 102, 1576–1582.10.1172/JCI4044Search in Google Scholar PubMed PubMed Central

Walter, U. and Gambaryan, S. (2009). cGMP and cGMP-dependent protein kinase in platelets and blood cells. Handb. Exp. Pharmacol. 191, 533–548.10.1007/978-3-540-68964-5_23Search in Google Scholar PubMed

Wang, R., Kwon, I.K., Thangaraju, M., Singh, N., Liu, K., Jay, P., Hofmann, F., Ganapathy, V., and Browning, D.D. (2012). Type 2 cGMP-dependent protein kinase regulates proliferation and differentiation in the colonic mucosa. Am. J. Physiol. Gastrointest. Liver Physiol. 303, G209–219.10.1152/ajpgi.00500.2011Search in Google Scholar PubMed

Wang, R., Kwon, I.K., Singh, N., Islam, B., Liu, K., Sridhar, S., Hofmann, F., and Browning, D.D. (2014). Type 2 cGMP-dependent protein kinase regulates homeostasis by blocking c-Jun N-terminal kinase in the colon epithelium. Cell Death Differ. 21, 427–437.10.1038/cdd.2013.163Search in Google Scholar PubMed PubMed Central

Weber, S., Bernhard, D., Lukowski, R., Weinmeister, P., Worner, R., Wegener, J.W., Valtcheva, N., Feil, S., Schlossmann, J., Hofmann, F., et al. (2007). Rescue of cGMP kinase I knockout mice by smooth muscle specific expression of either isozyme. Circ. Res. 101, 1096–1103.10.1161/CIRCRESAHA.107.154351Search in Google Scholar PubMed

Weber, S., Zeller, M., Guan, K., Wunder, F., Wagner, M., and El-Armouche, A. (2017). PDE2 at the crossway between cAMP and cGMP signalling in the heart. Cell Signal. 38, 76–84.10.1016/j.cellsig.2017.06.020Search in Google Scholar PubMed

Wedel, B., Humbert, P., Harteneck, C., Foerster, J., Malkewitz, J., Bohme, E., Schultz, G., and Koesling, D. (1994). Mutation of His-105 in the beta 1 subunit yields a nitric oxide-insensitive form of soluble guanylyl cyclase. Proc. Natl. Acad. Sci. U.S.A. 91, 2592–2596.10.1073/pnas.91.7.2592Search in Google Scholar PubMed PubMed Central

Wegener, J.W., Schulla, V., Koller, A., Klugbauer, N., Feil, R., and Hofmann, F. (2006). Control of intestinal motility by the Ca(v)1.2 L-type calcium channel in mice. FASEB J. 20, 1260–1262.10.1096/fj.05-5292fjeSearch in Google Scholar PubMed

Wei, F., Wang, W., and Liu, Q. (2013). Protein kinases of Toxoplasma gondii: functions and drug targets. Parasitol. Res. 112, 2121–2129.10.1007/s00436-013-3451-ySearch in Google Scholar PubMed

Wen, X.H., Dizhoor, A.M., and Makino, C.L. (2014). Membrane guanylyl cyclase complexes shape the photoresponses of retinal rods and cones. Front. Mol. Neurosci. 7, 45.10.3389/fnmol.2014.00045Search in Google Scholar PubMed PubMed Central

Wen, L., Feil, S., Wolters, M., Thunemann, M., Regler, F., Schmidt, K., Friebe, A., Olbrich, M., Langer, H., Gawaz, M., et al. (2018). A shear-dependent NO-cGMP-cGKI cascade in platelets acts as an auto-regulatory brake of thrombosis. Nat. Commun. 9, 4301.10.1038/s41467-018-06638-8Search in Google Scholar PubMed PubMed Central

Wincott, C.M., Abera, S., Vunck, S.A., Tirko, N., Choi, Y., Titcombe, R.F., Antoine, S.O., Tukey, D.S., DeVito, L.M., Hofmann, F., et al. (2014). cGMP-dependent protein kinase type II knockout mice exhibit working memory impairments, decreased repetitive behavior, and increased anxiety-like traits. Neurobiol. Learn. Mem. 114, 32–39.10.1016/j.nlm.2014.04.007Search in Google Scholar PubMed PubMed Central

Wobst, J., Schunkert, H., and Kessler, T. (2018). Genetic alterations in the NO-cGMP pathway and cardiovascular risk. Nitric Oxide 76, 105–112.10.1016/j.niox.2018.03.019Search in Google Scholar PubMed

Wu, Y., Cai, Q., Li, W., Cai, Z., Liu, Y., Li, H., Pang, J., and Chen, Y. (2019). Active PKG II inhibited the growth and migration of ovarian cancer cells through blocking Raf/MEK and PI3K/Akt signaling pathways. Biosci. Rep. 39, pii: BSR20190405.10.1042/BSR20190405Search in Google Scholar PubMed PubMed Central

Yamahara, K., Itoh, H., Chun, T.H., Ogawa, Y., Yamashita, J., Sawada, N., Fukunaga, Y., Sone, M., Yurugi-Kobayashi, T., Miyashita, K., et al. (2003). Significance and therapeutic potential of the natriuretic peptides/cGMP/cGMP-dependent protein kinase pathway in vascular regeneration. Proc. Natl. Acad. Sci. U.S.A. 100, 3404–3409.10.1073/pnas.0538059100Search in Google Scholar PubMed PubMed Central

Yang, H.M., Jin, S., Jang, H., Kim, J.Y., Lee, J.E., Kim, J., and Kim, H.S. (2019). Sildenafil reduces neointimal hyperplasia after angioplasty and inhibits platelet aggregation via activation of cGMP-dependent protein kinase. Sci Rep. 9, 7769.10.1038/s41598-019-44190-7Search in Google Scholar PubMed PubMed Central

Yasoda, A. and Nakao, K. (2010). Translational research of C-type natriuretic peptide (CNP) into skeletal dysplasias. Endocr. J. 57, 659–666.10.1507/endocrj.K10E-164Search in Google Scholar

Zaccolo, M. and Movsesian, M.A. (2007). cAMP and cGMP signaling cross-talk: role of phosphodiesterases and implications for cardiac pathophysiology. Circ. Res. 100, 1569–1578.10.1161/CIRCRESAHA.106.144501Search in Google Scholar PubMed

Zamanillo, D., Sprengel, R., Hvalby, O., Jensen, V., Burnashev, N., Rozov, A., Kaiser, K.M., Koster, H.J., Borchardt, T., Worley, P., et al. (1999). Importance of AMPA receptors for hippocampal synaptic plasticity but not for spatial learning. Science 284, 1805–1811.10.1126/science.284.5421.1805Search in Google Scholar PubMed

Zhang, Y.E. (2009). Non-Smad pathways in TGF-beta signaling. Cell Res. 19, 128–139.10.1038/cr.2008.328Search in Google Scholar PubMed PubMed Central

Zhang, L., Lukowski, R., Gaertner, F., Lorenz, M., Legate, K.R., Domes, K., Angermeier, E., Hofmann, F., and Massberg, S. (2013). Thrombocytosis as a response to high interleukin-6 levels in cGMP-dependent protein kinase I mutant mice. Arterioscler. Thromb. Vasc. Biol. 33, 1820–1828.10.1161/ATVBAHA.113.301507Search in Google Scholar PubMed

Zhuo, M., Hu, Y., Schultz, C., Kandel, E.R., and Hawkins, R.D. (1994). Role of guanylyl cyclase and cGMP-dependent protein kinase in long-term potentiation. Nature 368, 635–639.10.1038/368635a0Search in Google Scholar PubMed

Received: 2019-10-07
Accepted: 2019-10-30
Published Online: 2019-11-19
Published in Print: 2020-03-26

©2020 Walter de Gruyter GmbH, Berlin/Boston

Downloaded on 5.6.2024 from https://www.degruyter.com/document/doi/10.1515/hsz-2019-0386/html
Scroll to top button