Generic placeholder image

Current Pharmacogenomics and Personalized Medicine

Editor-in-Chief

ISSN (Print): 1875-6921
ISSN (Online): 1875-6913

Research Article

The Comparative Genomics and Network Analysis of eNOS by Using Different Bioinformatics Approaches

Author(s): Arpita Banerjee, Randeep Singh, Nymphaea Arora, Tania Arora, Vikash Prashar, Priya Godara, Arti Sharma*, Harish Changotra and Jyoti Parkash*

Volume 20, Issue 1, 2023

Published on: 15 February, 2023

Page: [37 - 56] Pages: 20

DOI: 10.2174/1875692120666230126110252

Price: $65

conference banner
Abstract

Background: Nitric oxide synthase (NOS) is an enzyme that catalyzes the synthesis of nitric oxide (NO) from L-arginine. It has three isoforms- (i) neuronal NOS (nNOS or NOS1), which participates in neural transmission; (ii) inducible NOS (iNOS or NOS2), which produces NO in macrophages; and (iii) endothelial NOS (eNOS or NOS3) that regulates blood pressure. The eNOS is mainly expressed in blood vessels and is a crucial regulator of endothelial homeostasis.

Objective: The present study aimed to unravel the role of eNOS in different signaling pathways and its involvement as a therapeutic target in various neurodegenerative disorders.

Methods: This study used various in silico methods for comprehensive genomic analysis of eNOS in 16 organisms from 7 different phyla. Prediction of conserved domains and evolutionary relationship for eNOS among 16 organisms was made. Various physical and chemical parameters, signal peptides, and transmembrane regions that helped understand its functional relevance were also studied.

Results: Three transcription factor binding sites (TFBS), i.e., CP2, AR, and LDSPOLYA, were identified in human eNOS, while ATF1, T3R, and STAT1 were predicted in mouse eNOS. Transcription factors were identified for each regulatory region in human as well as mouse eNOS. eNOS protein was predicted to harbor 14 different post-translational modification (PTM) sites, most of which have phosphorylation (serine followed by threonine and tyrosine phosphorylation) followed by sumoylation and palmitoylation among all the organisms used in the current study. However, human eNOS has a relatively lower number of PTM sites for tyrosine phosphorylation.

Conclusion: Structures of eNOS isoform, consistent with available biochemical and structural data, provide substantial insight into the NOS conformational changes, which give in-depth knowledge of the mechanism of eNOS, and will be helpful for better understanding the role of eNOS in pathophysiology.

Graphical Abstract

[1]
Chen Z, D S, Oliveira S, Zimnicka AM, et al. Reciprocal regulation of eNOS and caveolin-1 functions in endothelial cells. Mol Biol Cell 2018; 29(10): 1190-202.
[http://dx.doi.org/10.1091/mbc.E17-01-0049] [PMID: 29563255]
[2]
Bredt DS, Snyder SH. Isolation of nitric oxide synthetase, a calmodulin-requiring enzyme. Proc Natl Acad Sci 1990; 87(2): 682-5.
[http://dx.doi.org/10.1073/pnas.87.2.682] [PMID: 1689048]
[3]
Li Q, Youn JY, Cai H. Mechanisms and consequences of endothelial nitric oxide synthase dysfunction in hypertension. J Hypertens 2015; 33(6): 1128-36.
[http://dx.doi.org/10.1097/HJH.0000000000000587] [PMID: 25882860]
[4]
Huang PL, Dawson TM, Bredt DS, Snyder SH, Fishman MC. Targeted disruption of the neuronal nitric oxide synthase gene. Cell 1993; 75(7): 1273-86.
[http://dx.doi.org/10.1016/0092-8674(93)90615-W] [PMID: 7505721]
[5]
Keilhoff G, Seidel B, Reiser M, et al. Lack of neuronal NOS has consequences for the expression of POMC and POMC-derived peptides in the mouse pituitary. Acta Histochem 2001; 103(4): 397-412.
[http://dx.doi.org/10.1078/0065-1281-00611] [PMID: 11700945]
[6]
Töpel I, Stanarius A, Wolf G. Distribution of the endothelial constitutive nitric oxide synthase in the developing rat brain: an immunohistochemical study. Brain Res 1998; 788(1-2): 43-8.
[http://dx.doi.org/10.1016/S0006-8993(97)01506-0] [PMID: 9554947]
[7]
Stanarius A, Töpel I, Schulz S, Noack H. wolf G. Immunocytochemistry of endothelial nitric oxide synthase in the rat brain: a light and electron microscopical study using the tyramide signal amplification technique. Acta Histochem 1997; 99(4): 411-29.
[http://dx.doi.org/10.1016/S0065-1281(97)80034-7] [PMID: 9429601]
[8]
Umar S, van der Laarse A. Nitric oxide and nitric oxide synthase isoforms in the normal, hypertrophic, and failing heart. Mol Cell Biochem 2010; 333(1-2): 191-201.
[http://dx.doi.org/10.1007/s11010-009-0219-x] [PMID: 19618122]
[9]
Feron O, Dessy C, Desager JP, Balligand JL. Hydroxy-methylglutaryl-coenzyme A reductase inhibition promotes endothelial nitric oxide synthase activation through a decrease in caveolin abundance. Circulation 2001; 103(1): 113-8.
[http://dx.doi.org/10.1161/01.CIR.103.1.113] [PMID: 11136695]
[10]
Feron O, Kelly RA. The caveolar paradox: suppressing, inducing, and terminating eNOS signaling. Circ Res 2001; 88(2): 129-31.
[http://dx.doi.org/10.1161/01.RES.88.2.129] [PMID: 11157661]
[11]
Feron O, Belhassen L, Kobzik L, Smith TW, Kelly RA, Michel T. Endothelial nitric oxide synthase targeting to caveolae. Specific interactions with caveolin isoforms in cardiac myocytes and endothelial cells. J Biol Chem 1996; 271(37): 22810-4.
[http://dx.doi.org/10.1074/jbc.271.37.22810] [PMID: 8798458]
[12]
Michel JB, Feron O, Sacks D, Michel T. Reciprocal regulation of endothelial nitric-oxide synthase by Ca2+-calmodulin and caveolin. J Biol Chem 1997; 272(25): 15583-6.
[http://dx.doi.org/10.1074/jbc.272.25.15583] [PMID: 9188442]
[13]
Fulton D, Gratton JP, McCabe TJ, et al. Regulation of endothelium-derived nitric oxide production by the protein kinase Akt. Nature 1999; 399(6736): 597-601.
[http://dx.doi.org/10.1038/21218] [PMID: 10376602]
[14]
Bełtowski J. Leptin and the regulation of endothelial function in physiological and pathological conditions. Clin Exp Pharmacol Physiol 2012; 39(2): 168-78.
[http://dx.doi.org/10.1111/j.1440-1681.2011.05623.x] [PMID: 21973116]
[15]
Zhao Y, Vanhoutte PM, Leung SWS. Vascular nitric oxide: Beyond eNOS. J Pharmacol Sci 2015; 129(2): 83-94.
[http://dx.doi.org/10.1016/j.jphs.2015.09.002] [PMID: 26499181]
[16]
Virdis A, Bacca A, Colucci R, et al. Endothelial dysfunction in small arteries of essential hypertensive patients: role of cyclooxygenase-2 in oxidative stress generation. Hypertension 2013; 62(2): 337-–344.
[http://dx.doi.org/10.1161/HYPERTENSIONAHA.111.00995]
[17]
Wong WT, Tian XY, Huang Y. Endothelial dysfunction in diabetes and hypertension: Cross talk in RAS, BMP4, and ROS-dependent COX-2-derived prostanoids. J Cardiovasc Pharmacol 2013; 61(3): 204-14.
[http://dx.doi.org/10.1097/FJC.0b013e31827fe46e] [PMID: 23232839]
[18]
Austin SA, d’Uscio LV, Katusic ZS. Supplementation of nitric oxide attenuates AβPP and BACE1 protein in cerebral microcirculation of eNOS-deficient mice. J Alzheimers Dis 2012; 33(1): 29-33.
[http://dx.doi.org/10.3233/JAD-2012-121351] [PMID: 22886025]
[19]
Hariharan A, Jing Y, Collie ND, Zhang H, Liu P. P3-204: Endothelial nitric oxide synthase deficiency alters neurovascular coupling and brain glutamine metabolism. 2018; 14(7S_Part_21): 1146.
[http://dx.doi.org/10.1016/j.jalz.2018.06.1563]
[20]
Tan XL, Xue YQ, Ma T, et al. Partial eNOS deficiency causes spontaneous thrombotic cerebral infarction, amyloid angiopathy and cognitive impairment. Mol Neurodegener 2015; 10(1): 24.
[http://dx.doi.org/10.1186/s13024-015-0020-0] [PMID: 26104027]
[21]
ALrefai AA Habib MSE, Yaseen RI, Gabr MK, Habeeb RM. Association of endothelial nitric oxide synthase (eNOS) gene G894T polymorphism with hypertension risk and complications. Mol Cell Biochem 2016; 421(1-2): 103-10.
[http://dx.doi.org/10.1007/s11010-016-2790-2] [PMID: 27557897]
[22]
Nishank SS. Endothelial Nitric Oxide Synthase (eNOS) gene polymorphism is associated with age onset of menarche in sickle cell disease females of India. Mediterr J Hematol Infect Dis 2013; 5(1): e2013036.
[http://dx.doi.org/10.4084/mjhid.2013.036] [PMID: 23795274]
[23]
Bhanoori M. Endothelial nitric oxide synthase (eNOS) variants in cardiovascular disease: Pharmacogenomic implications. Indian J Med Res 2011; 133(5): 464-6.
[PMID: 21623028]
[24]
Gibney G, Baxevanis AD. Baxevanis, Searching NCBI Databases Using Entrez. In: Current protocols in human genetics. 2011.
[http://dx.doi.org/10.1002/0471142905.hg0610s71]
[25]
Lemoine F, Correia D, Lefort V, et al. NGPhylogeny.fr: new generation phylogenetic services for non-specialists. Nucleic Acids Res 2019; 47(W1): W260-5.
[http://dx.doi.org/10.1093/nar/gkz303] [PMID: 31028399]
[26]
El-Gebali S, Mistry J, Bateman A, et al. The Pfam protein families database in 2019. Nucleic Acids Res 2019; 47(D1): D427-32.
[http://dx.doi.org/10.1093/nar/gky995] [PMID: 30357350]
[27]
Bock JR, Gough DA. Predicting protein-protein interactions from primary structure. Bioinformatics 2001; 17(5): 455-60.
[http://dx.doi.org/10.1093/bioinformatics/17.5.455] [PMID: 11331240]
[28]
Gasteiger E, Hoogland C, Gattiker A, et al. Protein identification and analysis tools on the ExPASy server.In: Walker JM, Ed The Proteomics Protocols Handbook. Totowa, NJ: Humana Press 2005; pp. 571-607.
[http://dx.doi.org/10.1385/1-59259-890-0:571]
[29]
Kyte J, Doolittle RF. A simple method for displaying the hydropathic character of a protein. J Mol Biol 1982; 157(1): 105-32.
[http://dx.doi.org/10.1016/0022-2836(82)90515-0] [PMID: 7108955]
[30]
Vij A, Randhawa R, Parkash J, Changotra H. Investigating regulatory signatures of human autophagy related gene 5 (ATG5) through functional in silico analysis. Meta Gene 2016; 9: 237-48.
[http://dx.doi.org/10.1016/j.mgene.2016.07.012] [PMID: 27617225]
[31]
Gotea V, Ovcharenko I. DiRE: identifying distant regulatory elements of co-expressed genes. Nucleic acids research 2008; 36: W133–W139.
[http://dx.doi.org/10.1093/nar/gkn300]
[32]
Kwon AT, Arenillas DJ, Hunt RW, Wasserman WW. oPOSSUM-3: Advanced analysis of regulatory motif over-representation across genes or ChIP-Seq datasets. G3 2012; 2(9): 987-1002.
[http://dx.doi.org/10.1534/g3.112.003202] [PMID: 22973536]
[33]
Vaser R, Adusumalli S, Leng SN, Sikic M, Ng PC. SIFT missense predictions for genomes. Nat Protoc 2016; 11(1): 1-9.
[http://dx.doi.org/10.1038/nprot.2015.123] [PMID: 26633127]
[34]
Singh N, Upadhyay S, Jaiswar A, Mishra N. In silico analysis of protein. J Bioinform Genomics Proteomics 2016; 1(2): 1007.
[35]
Xie Y, Zheng Y, Li H, et al. GPS-Lipid: a robust tool for the prediction of multiple lipid modification sites. Sci Rep 2016; 6(1): 28249.
[http://dx.doi.org/10.1038/srep28249] [PMID: 27306108]
[36]
Arshad M, Bhatti A, John P. Identification and in silico analysis of functional SNPs of human TAGAP protein: A comprehensive study. PLoS One 2018; 13(1): e0188143.
[http://dx.doi.org/10.1371/journal.pone.0188143] [PMID: 29329296]
[37]
Muniz L, Luizon MR, Palei ACT, et al. eNOS tag SNP haplotypes in hypertensive disorders of pregnancy. DNA Cell Biol 2012; 31(12): 1665-70.
[http://dx.doi.org/10.1089/dna.2012.1768] [PMID: 23062210]
[38]
Hornbeck PV, Kornhauser JM, Latham V, et al. 15 years of PhosphoSitePlus®: integrating post-translationally modified sites, disease variants and isoforms. Nucleic Acids Res 2019; 47(D1): D433-41.
[http://dx.doi.org/10.1093/nar/gky1159] [PMID: 30445427]
[39]
Barandun J, Delley CL, Weber-Ban E. The pupylation pathway and its role in mycobacteria. BMC Biol 2012; 10(1): 95.
[http://dx.doi.org/10.1186/1741-7007-10-95] [PMID: 23198822]
[40]
Kone BC. Protein-protein interactions controlling nitric oxide synthases. Acta Physiol Scand 2000; 168(1): 27-31.
[http://dx.doi.org/10.1046/j.1365-201x.2000.00629.x] [PMID: 10691776]
[41]
Clapauch R, Mourão AF, Mecenas AS, Maranhão PA, Rossini A, Bouskela E. Endothelial function and insulin resistance in early postmenopausal women with cardiovascular risk factors: importance of ESR1 and NOS3 polymorphisms. PLoS One 2014; 9(7): e103444.
[http://dx.doi.org/10.1371/journal.pone.0103444] [PMID: 25077953]
[42]
Sud N, Sharma S, Wiseman DA, et al. Nitric oxide and superoxide generation from endothelial NOS: modulation by HSP90. Am J Physiol Lung Cell Mol Physiol 2007; 293(6): L1444-53.
[http://dx.doi.org/10.1152/ajplung.00175.2007] [PMID: 17827253]
[43]
Schilling K, Opitz N, Wiesenthal A, et al. Translocation of endothelial nitric-oxide synthase involves a ternary complex with caveolin-1 and NOSTRIN. Mol Biol Cell 2006; 17(9): 3870-80.
[http://dx.doi.org/10.1091/mbc.e05-08-0709] [PMID: 16807357]
[44]
Sousa MSA, Latini FRM, Monteiro HP, Cerutti JM. Arginase 2 and nitric oxide synthase: Pathways associated with the pathogenesis of thyroid tumors. Free Radic Biol Med 2010; 49(6): 997-1007.
[http://dx.doi.org/10.1016/j.freeradbiomed.2010.06.006] [PMID: 20542107]
[45]
Somanath PR, Razorenova OV, Chen J, Byzova TV. Akt1 in endothelial cell and angiogenesis. Cell Cycle 2006; 5(5): 512-8.
[http://dx.doi.org/10.4161/cc.5.5.2538] [PMID: 16552185]
[46]
Nickel W, Seedorf M. Unconventional mechanisms of protein transport to the cell surface of eukaryotic cells. Annu Rev Cell Dev Biol 2008; 24(1): 287-308.
[http://dx.doi.org/10.1146/annurev.cellbio.24.110707.175320] [PMID: 18590485]
[47]
Heijnen HFG, Waaijenborg S, Crapo JD, Bowler RP, Akkerman JWN, Slot JW. Colocalization of eNOS and the catalytic subunit of PKA in endothelial cell junctions: a clue for regulated NO production. J Histochem Cytochem 2004; 52(10): 1277-85.
[http://dx.doi.org/10.1177/002215540405201004] [PMID: 15385574]
[48]
Davis ME, Grumbach IM, Fukai T, Cutchins A, Harrison DG. Shear stress regulates endothelial nitric-oxide synthase promoter activity through nuclear factor kappaB binding. J Biol Chem 2004; 279(1): 163-8.
[http://dx.doi.org/10.1074/jbc.M307528200] [PMID: 14570928]
[49]
Nakamura T, Prikhodko OA, Pirie E, et al. Aberrant protein S-nitrosylation contributes to the pathophysiology of neurodegenerative diseases. Neurobiol Dis 2015; 84: 99-108.
[http://dx.doi.org/10.1016/j.nbd.2015.03.017] [PMID: 25796565]
[50]
Mount PF, Kemp BE, Power DA. Regulation of endothelial and myocardial NO synthesis by multi-site eNOS phosphorylation. J Mol Cell Cardiol 2007; 42(2): 271-9.
[http://dx.doi.org/10.1016/j.yjmcc.2006.05.023] [PMID: 16839566]
[51]
Fulton DJR. Transcriptional and Posttranslational Regulation of eNOS in the Endothelium. Adv Pharmacol 2016; 77: 29-64.
[http://dx.doi.org/10.1016/bs.apha.2016.04.001] [PMID: 27451094]
[52]
Chen HH, Liu P, Auger P, et al. Calpain-mediated tau fragmentation is altered in Alzheimer’s disease progression. Sci Rep 2018; 8(1): 16725.
[http://dx.doi.org/10.1038/s41598-018-35130-y] [PMID: 30425303]
[53]
del Carmen Lafita-Navarro M, Conacci-Sorrell M. Identification of calpain-activated protein functions. Methods Mol Biol 2019; 1915: 149-60.
[http://dx.doi.org/10.1007/978-1-4939-8988-1_12] [PMID: 30617802]
[54]
Josa-Prado F, Luo J, Rubin P, Henley JM, Wilkinson KA. Developmental profiles of SUMOylation pathway proteins in rat cerebrum and cerebellum. PLoS One 2019; 14(2): e0212857.
[http://dx.doi.org/10.1371/journal.pone.0212857] [PMID: 30794696]
[55]
Antonelli M, Fadda A, Loi E, et al. Integrated DNA methylation analysis identifies topographical and tumoral biomarkers in pilocytic astrocytomas. Oncotarget 2018; 9(17): 13807-21.
[http://dx.doi.org/10.18632/oncotarget.24480] [PMID: 29568396]
[56]
Pearce MJ, Mintseris J, Ferreyra J, Gygi SP, Darwin KH. Ubiquitin-like protein involved in the proteasome pathway of Mycobacterium tuberculosis. Science 2008; 322(5904): 1104-7.
[http://dx.doi.org/10.1126/science.1163885] [PMID: 18832610]
[57]
Davies MN, Kjalarsdottir L, Thompson JW, et al. The acetyl group buffering action of carnitine acetyltransferase offsets macronutrient-induced lysine acetylation of mitochondrial proteins. Cell Rep 2016; 14(2): 243-54.
[http://dx.doi.org/10.1016/j.celrep.2015.12.030] [PMID: 26748706]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy