Next Article in Journal
Enrichment of Bread with Lycium barbarum (Goji) Puree
Previous Article in Journal
Burnout Syndrome and Associated Sociodemographic Factors in Medical Students: A Cross-Sectional Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Proceeding Paper

GPCRs of Diverse Physiologic and Pathologic Effects with Fingerprints in COVID-19 †

by
Reza Nejat
1,2,*,
Ahmad Shahir Sadr
3,4,5,
Maziar Fayaz Torshizi
6 and
David J. Najafi
7
1
Department of Anesthesiology and Critical Care Medicine, Shahid Beheshti University of Medical Sciences, Tehran 1971983314, Iran
2
Bazarganan Hospital, Tehran 1173814411, Iran
3
Institute for Research in Fundamental Sciences (IPM), School of Biological Sciences, Tehran 9613937932, Iran
4
Department of Computer and Data Sciences, Faculty of Mathematical Sciences, Shahid Beheshti University, Tehran 1983969411, Iran
5
Bioinformatics Research Center, Cheragh Medical Institute and Hospital, Kabul 0093, Afghanistan
6
Department of Chemical Engineering, Imperial College London, London SW7 2BB, UK
7
Alliance Retina Consultants, La Mesa, CA 91942, USA
*
Author to whom correspondence should be addressed.
Presented at the 1st International Electronic Conference on Biomedicine, 1–26 June 2021; Available online: https://ecb2021.sciforum.net/.
Biol. Life Sci. Forum 2021, 7(1), 19; https://doi.org/10.3390/ECB2021-10261
Published: 31 May 2021
(This article belongs to the Proceedings of The 1st International Electronic Conference on Biomedicine)

Abstract

:
G-protein-coupled receptors (GPCR) belong to a large family of molecules eliciting different responses to a variety of signaling molecules. These receptors participate in various physiological functions such as metabolism, growth, immune responses, inflammation, vision, taste, olfaction, and neurotransmission as well as in pathologic responses including chronic inflammatory and vascular diseases. Receptors contributing to the biological responses of the renin–angiotensin system (RAS) are members of the GPCR family. COVID-19-induced inflammatory cascade has been attributed to acute ACE2 downregulation and imbalance of proinflammatory ACE/AngII/AT1R and anti-inflammatory ACE2/angiotensin (1–7)/Mas axes in favor of the former. Some of the receptors contributing to activities of proteins in RAS, including AT1R, AT2R, Mas receptors, and Mas-related GPCR-member D receptor are members of the GPCR family. It is notable that these receptors induce their effects both through G-protein and β-arrestin pathways; the former exerts temporary and the latter more sustained effects. In addition to the imbalance of GPCR responses contributing to RAS activities, it has been suggested that SARS-CoV2 pathogenesis might be attributed to the activation of GPCRs or modulating G proteins involved in the adenosine–CFTR regulation system and epithelial Na-channel function. This article includes a minireview of the physiological functions of GPCRs and their contribution to COVID-19.

1. Introduction

Cell-signaling, an adaptive strategy of cells in interaction with the environment, involves cellular chemical interaction to provoke appropriate responses to increase their survival. Cells release signaling molecules to bind to the receptors expressed in and on themselves or on neighboring or remote cells to initiate, integrate, or coordinate their activities [1]. Receptors are of different families. G-protein-coupled receptors (GPCR), as a large family of proteins encoded in the mammalian genome, contribute to many physiologic functions including vision, olfaction, taste, neurotransmission, metabolism, cell differentiation, immunity, inflammation, and prohormone signaling and, if dysregulated, can lead to several pathologic processes including hypersensitivity to angiotensin II and inflammatory and vascular diseases [2,3,4]. These receptors share a common counter-clockwise bundle structure composed of a single polypeptide with an extracellular N-terminus, an intracellular C-terminus and a hydrophobic seven-transmembrane domain (TM) connected by three intracellular and three extracellular loops [4,5] (Figure 1).
However, despite this common structure, the functional diversity and heterogenous amino acid sequence of GPCRs make it difficult to propose a comprehensive classification method of this receptor superfamily [6]. Once, GPCRs were classified based on ligand specificity or chemical properties of their primary residues, independent of the amino acid sequence [7,8]. Later, both characterization of sequence alignment and motifs as well as computational methods using machine learning algorithms such as those obtained from support vector machines (SVMs) associated with statistical models provided a rather robust way of prediction and classification of GPCRs [9,10]. Focusing on integration of the structure, amino acid sequence, and function of this receptor superfamily, the GPCR database (GPCRdb), a molecular class-specific information system, has made possible a reliable approach to classifying GPCRs. This database encompasses all human non-olfactory GPCRs along with G proteins, human β-arrestins and more than 28,000 orthologs updated continuously [11,12]. Recently, a novel GPCR data base, the GPCR ensemble database (GPCREnDB) containing 3129 confirmed GPCR and 3575 non-GPCR sequences has been introduced [13]. Based on sequence homology and functional similarity, the most widely used A–F classification of the GPCR family and subfamily in both vertebrates and invertebrates was suggested—Class A, rhodopsin-like; Class B1, secretin-like; Class B2, adhesion receptors; Class C, metabotropic glutamate receptors; Class D, pheromone receptors; Class E, cAMP receptors; and Class F, frizzled smoothened family [4,14,15]. Vertebrates do not possess D and E classes. Alternatively, GRAFS classification phylogenetically groups GPCRs into five families—glutamate (class C), rhodopsin (class A), adhesion (class B2), frizzled (class F), and secretin (class B1) [16,17,18]. These groups are hierarchically further divided into subfamilies, sub-subfamilies, and types, based on their specific ligands and function [12,19,20]. As phylogenetic classification fails to fully support receptor family structure in diverse species due to continuous novel genome characterization, a modified classification method, the level-based system, has been suggested [21].
Upon binding with agonistic ligands, GPCRs adopt a proton-transport-dependent conformational change and activate cytoplasmic heterotrimeric G proteins (containing Gα/Gβγ subunits) through dissociation or conformational rearrangement (rotation of 130°) of Gα from the Gβγ complex triggered by exchange of GTP for GDP in the Gα subunit [22,23,24,25]. Human cells contain 16 Gα, 5 Gβ, and 13 Gγ subunit combinations which yield a great number of heterotrimeric G proteins [15]. Depending on the family of Gα subunit activated by GPCR, a second messenger including cAMP, inositol phosphates (IP), Ca2+, and diacylglycerol (DAG) is induced which stimulates some intracellular pathways such as MAPK, PI3K-Akt, and Ras and Rho GTPases [26,27].
Based on the structure and function of the Gα subunit, G proteins are divided into four main families—Gαs, Gαo/i, Gαq/11, and Gα12/13 [28]. The Gαs family consists of Gαs and Gαolf, while the Gαo/i family consists of Gαi1, Gαi2, Gαi3, Gαo, Gαt1, Gαt2, Gαt3, and Gα. The Gαq/11 family encompasses Gαq,11, Gα14, and Gα16, and the Gα12/13 family comprises Gα12 and Gα13 [29]. The Gαs family contributes to activation of adenyl cyclase, Gαo/i inhibits adenyl cyclase and induces ion transport, Gαq/11 activates phospholipase C (PLC), and Gα12/13 is involved in the Na+/H+ exchange pathway [30].
It is of paramount importance that ligand–GPCR interaction is not a simple on/off switch. In fact, GPCR stimulation, depending on the energy transferred between ligand and G protein, is a complex phenomenon and may lead to multiple responses so that specific ligands may induce different pathways. Ligands may function as full, partial, or inverse agonist, or a neutral antagonist [31]. Even GPCR signaling system has recently been shown to be capable of responding to different concentrations of the cognate ligand with distinct responses [32].
After propagation of the response, hydrolysis of GTP to GDP by the GTPase intrinsic activity of the Gα subunit ends G-protein signaling transduction. GTPase-activating proteins (GAPs) and regulators of G-protein signaling (RGS) accelerate this reaction. This results in recreation of a Gα/Gβγ complex for the next signal transduction [32]. Alternatively, after activation of GPCRs, phosphorylation of serine and threonine residues on the C-terminal end or third intracellular loop of the receptors by GPCR kinases (GRK) creates a binding site for β-arrestins. Binding with β-arrestins shuts down G-dependent signaling cascades through desensitization of the receptor to the next ligand or internalization of the receptor and promotion of multiple G-independent pathways by inducing different protein-kinase-dependent pathways [33,34,35]. For instance, internalization of the complex of ligand activated GPCRs and β-arrestin promotes prolonged MAPK signaling predominantly in the cytoplasm and independent of G-protein signaling [33].
Intriguingly, GPCRs can also be found within the cells residing on membranous organelles such as endoplasmic reticulum, Golgi apparatus, and endosomes with the ability to translocate to the nucleus. After internalization, AT1R, apelin, and bradykinin2 receptors as well as receptors for chemokines may use canonical nuclear localization signals (NLSs) recognized by the karyopherin superfamily for nuclear import [36,37].

2. COVID-19 and GPCRs

SARS-CoV2, one of the members of β-family of coronaviruses and the cause of COVID-19 has resulted in a pandemic with a huge social economical and healthcare burden [38]. The resulting insult leads to acute inflammation in the lungs similar to the pathology seen in acute respiratory distress syndrome. It is worth mentioning that SARS-CoV2, like SARS-CoV, evades the immune system [39,40]. Accordingly, failure to activate a well-orchestrated effective innate immunity leads to eruption of an abortive adaptive immunity with the inability to resolve the pathology and clearance of infected cells. This results in sequential uninterrupted production of inflammatory mediators and eventually, a cytokine storm [41,42].
GPCRs contribute to inflammation with their presence as receptors for chemoattractants and chemokines on the cells such as polymorphonuclears, monocytes, macrophages, and endothelial cells and by mediating the production of cytokines and other inflammatory molecules [43]. Based on a great amount of data, the elicited profound inflammatory responses in COVID-19 has been attributed to an imbalance in the local renin angiotensin system (RAS) [44]. SARS-CoV2 on entry the host cell uses ACE2 as its receptor which is itself a transmembrane carboxy-monopeptidase of RAS converting Ang II to angiotensin (1–7) [45,46]. Acute downregulation of ACE2 resulting from internalization of virus–ACE2 complexes to the infected cells, as may occur in showering of a tremendous number of SARS-CoV2 virions to the lower airways, leads to an acute imbalance of the two major arms of local RAS, the ACE/Ang II/AT1R and ACE2/angiotensin (1–7)/Mas receptor pathways, in favor of the former [47]. ACE/Ang II/AT1R signaling elicits pro-inflammatory, pro-thrombotic, pro-apoptotic, and oxidative responses while the ACE2/angiotensin (1–7)/Mas receptor pathway predominantly provokes anti-inflammatory, anti-thrombotic, and anti-apoptotic effects [48]. Ang II-mediated AT1R stimulation upregulates phosphorylation of Yes-associated protein (YAP) and activates macrophages and their adhesion to endothelial cells [49]. Intriguingly, AT1R, AT2R, and the Mas receptor are members of GPCR family [50,51,52,53] although AT2R may also promote ERK1/2 and nitric oxide synthase (NOS) through a GPCR- or arrestin-independent pathway [54]. AngII activation of AT1R induces inflammatory responses through various G proteins—Gαo/i, Gαq/11, and Gα12/13 [55]. Gαq activation of PLC produces DAG and IP3. The latter promotes intracellular calcium mobilization which, in collaboration with DAG activates various protein kinase C involved in inducing innate and adaptive immunity components such as TL4, inducible NOS, and NF-kB signaling as well as gene expression and release of an array of cytokines such as TNF-α in tissues including the lungs [55,56,57,58]. Furthermore, PKC contributes to oxidative stress as a mediator of NADPH-oxidase activation and plays a role in neutrophil extracellular-trap (NET) formation [59]. In addition, calcium influx leads to disassembly of intercellular adhesion and cell retraction in endothelial cells resulting in increased permeability of the vascular wall [60]. Similar to other GPCRs, AT1R, after being complexed with β-arrestin, is internalized and the signaling transduction ends. The receptor may either be recycled to the cell membrane or degraded intracellularly. Alternatively, AT1R activation induces β-arrestin regulation of various MAPK-cascade members including p38MAPK, ERK, and JNK [55]. Angiotensin (1–7) promotes anti-inflammatory effects both via Mas/GPCR receptors and the AT1R/GPCR-mediated β-arrestin pathway [61]. It is implicated that in COVID-19 sustained imbalance of the two GPCR-mediated pathways in local RAS with predominance of AT1R over Mas-receptor signaling may ultimately result in severe inflammatory responses and multiple organ failure with utmost similarity to macrophage activation syndrome or other kinds of cytokine storms [47,62].
To make it more complex, homomerization and heteromerization of GPCRs, recognized as a constitutive property of this receptor superfamily that modulates receptor-evoked responses [63,64,65], may affect the adverse outcome of the hypothetical imbalance of counteracting arms of RAS in the pathogenesis of COVID-19. Receptor homomerization and heteromerization may lead to altered ligand binding, plasma-membrane diffusion, receptor endocytosis, and signaling transduction which may potentiate or attenuate pathobiological changes in the diseases [43,66]. For example, AT1R-bradykinin type 2 receptor (AT1R-B2R) complex may induce hypersensitivity to Ang II, while AT1R-AT2R complex reduces responses elicited by AT1R. Moreover, Ang (1–7)/Mas receptor in heteromerization with AT1R reduces the functional activities of the latter [67].
SARS-CoV2 has been suggested to induce lung edema through the cystic fibrosis transmembrane-conductance regulator (CFTR) system and epithelial Na+ channel (ENaC) function [68,69]. CFTR predominantly regulated by cAMP/PKA, the second messenger of the GPCR system, inhibits ENaC function, is yet to be elucidated meticulously [70,71]. ENaC as a member of voltage-independent Na+-selective ENaC/DEG ion channels is involved in airway and alveolar-space fluid clearance by epithelial and alveolar cells of the lungs [72,73]. SARS-CoV and some other viruses infecting the lungs, including the influenza virus, have been shown to decrease the activity of ENaC through activation of phosphokinase C (PKC) [74,75]. PKC, upregulated by extracellular purinergic receptors, which are themselves of or intricately interact with the GPCR family, may exert negative influence on ENaC cell-membrane trafficking and expression [76,77]. In addition, GPCR affects ENaC activity through phosphatidylinositol 4,5-biphosphate (PI4,5-P) [78].
Furthermore, complement 5a receptor1 (C5aR1), a member of the GPCR family, has recently been proposed to be involved in COVID-19 pathogenesis [79]. GPCR4, which regulates vascular permeability and leukocyte recruitment, has also been hypothesized to play a part in SARS-CoV2 infection [80].

3. Conclusions and Future Perspectives

GPCRs, as a large family of receptors in cell-signaling systems, contribute to a huge number of broad physiologic and pathologic responses. The effects of these receptors can be traced in a vast array of infections. As for the complex pathogenesis of SARS-CoV2 infection which requires the contribution of GPCRs and the sustained responses that these receptors may exert at intracellular scale, they might be a good target for production of drugs that could restrict this infection and improve the long-term sequelae of the disease through restoring cell homeostasis.

Author Contributions

R.N., A.S.S., M.F.T. and D.J.N. have contributed to the article equally. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

The authors would like to extend their gratitude to all healthcare personnel around the world who sacrifice their lives to take care of COVID-19 patients.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ACEangiotensin converting enzyme
ACE2angiotensin converting enzyme2
Ang IIangiotensin II
AT1Rtype I angiotensin II receptor
AT2Rtype II angiotensin II receptor
CFTRcystic fibrosis transmembrane conductance regulator
ENaCepithelial Na Channel
GPCRG-protein-coupled receptor
NOSnitric oxide synthase
RASrenin angiotensin system

References

  1. Cooper, G.M. Cell Signaling. In The Cell—A Molecular Approach; Oxford University Press.: New York, NY, USA, 2019; p. 565. [Google Scholar]
  2. Quitterer, U.; AbdAlla, S. Discovery of pathologic GPCR aggregation. Front. Med. 2019, 6, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Zalewska, M.; Siara, M.; Sajewicz, W. G protein-coupled receptors: Abnormalities in signal transmission, disease states and pharmacotherapy. Acta Pol. Pharm. 2014, 71, 229–243. [Google Scholar]
  4. Hu, G.-M.; Mai, T.-L.; Chen, C.-M. Visualizing the GPCR network: Classification and evolution. Sci. Rep. 2017, 7, 1–15. [Google Scholar] [CrossRef] [PubMed]
  5. Alexander, S.P.; Christopoulos, A.; Davenport, A.P.; Kelly, E.; Mathie, A.; Peters, J.A.; Veale, E.L.; Armstrong, J.F.; Faccenda, E.; Harding, S.D. The Concise Guide to PHARMACOLOGY 2019/20: G protein-coupled receptors. Br. J. Pharmacol. 2019, 176, S21–S141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Davies, M.N.; Gloriam, D.E.; Secker, A.; Freitas, A.A.; Mendao, M.; Timmis, J.; Flower, D.R. Proteomic applications of automated GPCR classification. Proteomics 2007, 7, 2800–2814. [Google Scholar] [CrossRef] [PubMed]
  7. Shrivastava, S.; Pardasani, K.; Malik, M. SVM Model for Identification of human GPCRs. arXiv 2010, arXiv:1002.3983. [Google Scholar]
  8. Lapinsh, M.; Gutcaits, A.; Prusis, P.; Post, C.; Lundstedt, T.; Wikberg, J.E. Classification of G-protein coupled receptors by alignment-independent extraction of principal chemical properties of primary amino acid sequences. Protein Sci. 2002, 11, 795–805. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Li, Z.; Zhou, X.; Dai, Z.; Zou, X. Classification of G-protein coupled receptors based on support vector machine with maximum relevance minimum redundancy and genetic algorithm. BMC Bioinform. 2010, 11, 1–15. [Google Scholar] [CrossRef] [Green Version]
  10. Li, M.; Ling, C.; Gao, J. An efficient CNN-based classification on G-protein Coupled Receptors using TF-IDF and N-gram. In Proceedings of the 2017 IEEE Symposium on Computers and Communications (ISCC), Heraklion, Greece, 3–6 July 2017; pp. 924–931. [Google Scholar]
  11. Kooistra, A.J.; Mordalski, S.; Pándy-Szekeres, G.; Esguerra, M.; Mamyrbekov, A.; Munk, C.; Keserű, G.M.; Gloriam, D.E. GPCRdb in 2021: Integrating GPCR sequence, structure and function. Nucleic Acids Res. 2021, 49, D335–D343. [Google Scholar] [CrossRef]
  12. Davies, M.N.; Secker, A.; Halling-Brown, M.; Moss, D.S.; Freitas, A.A.; Timmis, J.; Clark, E.; Flower, D.R. GPCRTree: Online hierarchical classification of GPCR function. BMC Res. Notes 2008, 1, 1–5. [Google Scholar] [CrossRef] [Green Version]
  13. Begum, K.; Mohl, J.E.; Ayivor, F.; Perez, E.E.; Leung, M.-Y. GPCR-PEnDB: A database of protein sequences and derived features to facilitate prediction and classification of G protein-coupled receptors. Database 2020, 2020, baaa087. [Google Scholar]
  14. Davies, M.N.; Secker, A.; Freitas, A.A.; Mendao, M.; Timmis, J.; Flower, D.R. On the hierarchical classification of G protein-coupled receptors. Bioinformatics 2007, 23, 3113–3118. [Google Scholar] [CrossRef] [Green Version]
  15. Wootten, D.; Christopoulos, A.; Marti-Solano, M.; Babu, M.M.; Sexton, P.M. Mechanisms of signalling and biased agonism in G protein-coupled receptors. Nat. Rev. Mol. Cell Biol. 2018, 19, 638–653. [Google Scholar] [CrossRef]
  16. Basith, S.; Cui, M.; Macalino, S.J.; Park, J.; Clavio, N.A.; Kang, S.; Choi, S. Exploring G protein-coupled receptors (GPCRs) ligand space via cheminformatics approaches: Impact on rational drug design. Front. Pharmacol. 2018, 9, 128. [Google Scholar] [CrossRef]
  17. De Francesco, E.M.; Sotgia, F.; Clarke, R.B.; Lisanti, M.P.; Maggiolini, M. G protein-coupled receptors at the crossroad between physiologic and pathologic angiogenesis: Old paradigms and emerging concepts. Int. J. Mol. Sci. 2017, 18, 2713. [Google Scholar] [CrossRef] [Green Version]
  18. Schiöth, H.B.; Fredriksson, R. The GRAFS classification system of G-protein coupled receptors in comparative perspective. Gen. Comp. Endocrinol. 2005, 142, 94–101. [Google Scholar] [CrossRef]
  19. Southan, C.; Sharman, J.L.; Benson, H.E.; Faccenda, E.; Pawson, A.J.; Alexander, S.P.; Buneman, O.P.; Davenport, A.P.; McGrath, J.C.; Peters, J.A.; et al. The IUPHAR/BPS Guide to PHARMACOLOGY in 2016: Towards curated quantitative interactions between 1300 protein targets and 6000 ligands. Nucleic Acids Res. 2016, 44, D1054–D1068. [Google Scholar] [CrossRef] [Green Version]
  20. Beigi, M.; Zell, A. A novel method for classifying subfamilies and sub-subfamilies of G-protein coupled receptors. In Proceedings of the International Symposium on Biological and Medical Data Analysis, Thessaloniki, Greece, 7–8 December 2006; pp. 25–36. [Google Scholar]
  21. Wittlake, A.; Prömel, S.; Schöneberg, T. The Evolutionary History of Vertebrate Adhesion GPCRs and Its Implication on Their Classification. Int. J. Mol. Sci. 2021, 22, 11803. [Google Scholar] [CrossRef]
  22. Rajagopal, S.; Rajagopal, K.; Lefkowitz, R.J. Teaching old receptors new tricks: Biasing seven-transmembrane receptors. Nat. Rev. Drug Discov. 2010, 9, 373–386. [Google Scholar] [CrossRef] [Green Version]
  23. Zhang, X.C.; Cao, C.; Zhou, Y.; Zhao, Y. Proton transfer-mediated GPCR activation. Protein Cell 2015, 6, 12–17. [Google Scholar] [CrossRef] [Green Version]
  24. Ganguly, S.; Saxena, R.; Chattopadhyay, A. Reorganization of the actin cytoskeleton upon G-protein coupled receptor signaling. Biochim. Et Biophys. Acta (BBA) Biomembr. 2011, 1808, 1921–1929. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Audet, M.; Bouvier, M. Restructuring G-protein-coupled receptor activation. Cell 2012, 151, 14–23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Walsh, C.T.; Stupack, D.; Brown, J.H. G Protein–Coupled Receptors Go Extracellular: RhoA Integrates the Integrins. Mol. Interv. 2008, 8, 165. [Google Scholar] [CrossRef] [PubMed]
  27. Otto, J.C.; Kelly, P.; Chiou, S.-T.; York, J.D. Alterations in an inositol phosphate code through synergistic activation of a G protein and inositol phosphate kinases. Proc. Natl. Acad. Sci. USA 2007, 104, 15653–15658. [Google Scholar] [CrossRef] [Green Version]
  28. Kamato, D.; Thach, L.; Bernard, R.; Chan, V.; Zheng, W.; Kaur, H.; Brimble, M.; Osman, N.; Little, P.J. Structure, function, pharmacology, and therapeutic potential of the G protein, Gα/q, 11. Front. Cardiovasc. Med. 2015, 2, 14. [Google Scholar] [CrossRef] [Green Version]
  29. Kim, H.R.; Xu, J.; Maeda, S.; Duc, N.M.; Ahn, D.; Du, Y.; Chung, K.Y. Structural mechanism underlying primary and secondary coupling between GPCRs and the Gi/o family. Nat. Commun. 2020, 11, 1–12. [Google Scholar] [CrossRef]
  30. Li, Z.; Zhou, X.; Dai, Z.; Zou, X. Classification of G proteins and prediction of GPCRs-G proteins coupling specificity using continuous wavelet transform and information theory. Amino Acids 2012, 43, 793–804. [Google Scholar] [CrossRef]
  31. Rosenbaum, D.M.; Rasmussen, S.G.; Kobilka, B.K. The structure and function of G-protein-coupled receptors. Nature 2009, 459, 356–363. [Google Scholar] [CrossRef] [Green Version]
  32. Keshelava, A.; Solis, G.P.; Hersch, M.; Koval, A.; Kryuchkov, M.; Bergmann, S.; Katanaev, V.L. High capacity in G protein-coupled receptor signaling. Nat. Commun. 2018, 9, 1–8. [Google Scholar] [CrossRef]
  33. Tilley, D.G. G protein–dependent and G protein–independent signaling pathways and their impact on cardiac function. Circ. Res. 2011, 109, 217–230. [Google Scholar] [CrossRef] [Green Version]
  34. Peterson, Y.K.; Luttrell, L.M. The diverse roles of arrestin scaffolds in G protein–coupled receptor signaling. Pharmacol. Rev. 2017, 69, 256–297. [Google Scholar] [CrossRef] [Green Version]
  35. Turu, G.; Balla, A.; Hunyady, L. The role of β-arrestin proteins in organization of signaling and regulation of the AT1 angiotensin receptor. Front. Endocrinol. 2019, 10, 519. [Google Scholar] [CrossRef]
  36. Jong, Y.J.I.; Harmon, S.K.; O’Malley, K.L. GPCR signalling from within the cell. Br. J. Pharmacol. 2018, 175, 4026–4035. [Google Scholar] [CrossRef] [Green Version]
  37. Eichel, K.; von Zastrow, M. Subcellular organization of GPCR signaling. Trends Pharmacol. Sci. 2018, 39, 200–208. [Google Scholar] [CrossRef]
  38. Andronico, A.; Kiem, C.T.; Paireau, J.; Succo, T.; Bosetti, P.; Lefrancq, N.; Nacher, M.; Djossou, F.; Sanna, A.; Flamand, C.; et al. Evaluating the impact of curfews and other measures on SARS-CoV-2 transmission in French Guiana. Nat. Commun. 2021, 12, 1–8. [Google Scholar] [CrossRef]
  39. Molaei, S.; Dadkhah, M.; Asghariazar, V.; Karami, C.; Safarzadeh, E. The immune response and immune evasion characteristics in SARS-CoV, MERS-CoV, and SARS-CoV-2: Vaccine design strategies. Int. Immunopharmacol. 2020, 92, 107051. [Google Scholar] [CrossRef]
  40. Nejat, R.; Sadr, A.S. SARS virus papain-like protease: A mysterious weapon. J. Biostat. Epidemiol. 2019, 5, 288–295. [Google Scholar] [CrossRef]
  41. Tufet, M. T cells calm the storm. Nat. Rev. Immunol. 2007, 7, 834–835. [Google Scholar] [CrossRef]
  42. Henderson, L.A.; Canna, S.W.; Schulert, G.S.; Volpi, S.; Lee, P.Y.; Kernan, K.F.; Caricchio, R.; Mahmud, S.; Hazen, M.M.; Halyabar, O.; et al. On the alert for cytokine storm: Immunopathology in COVID-19. Arthritis Rheumatol. 2020, 72, 1059–1063. [Google Scholar] [CrossRef] [Green Version]
  43. Barnes, P.J. Receptor heterodimerization: A new level of cross-talk. J. Clin. Investig. 2006, 116, 1210–1212. [Google Scholar] [CrossRef]
  44. Nejat, R.; Sadr, A.S. Are Losartan and Imatinib Effective Against SARS-CoV2 Pathogenesis? A Pathophysiologic-Based In Silico Study. Silico Pharmacol. 2021, 9, 1. [Google Scholar] [CrossRef] [PubMed]
  45. Hamming, I.; Cooper, M.E.; Haagmans, B.L.; Hooper, N.M.; Korstanje, R.; Osterhaus, A.D.; Timens, W.; Turner, A.; Navis, G.; van Goor, H. The emerging role of ACE2 in physiology and disease. J. Pathol. A J. Pathol. Soc. Great Br. Irel. 2007, 212, 1–11. [Google Scholar] [CrossRef] [PubMed]
  46. Shang, J.; Wan, Y.; Luo, C.; Ye, G.; Geng, Q.; Auerbach, A.; Li, F. Cell entry mechanisms of SARS-CoV-2. Proc. Natl. Acad. Sci. USA 2020, 117, 11727–11734. [Google Scholar] [CrossRef]
  47. Banu, N.; Panikar, S.S.; Leal, L.R.; Leal, A.R. Protective role of ACE2 and its downregulation in SARS-CoV-2 infection leading to Macrophage Activation Syndrome: Therapeutic implications. Life Sci. 2020, 256, 117905. [Google Scholar] [CrossRef] [PubMed]
  48. Reddy Gaddam, R.; Chambers, S.; Bhatia, M. ACE and ACE2 in inflammation: A tale of two enzymes. Inflamm. Allergy-Drug Targets (Former. Curr. Drug Targets-Inflamm. Allergy) (Discontin.) 2014, 13, 224–234. [Google Scholar]
  49. Wang, X.; Zhang, H.; Ge, Y.; Cao, L.; He, Y.; Sun, G.; Jia, S.; Ma, A.; Liu, J.; Rong, D.; et al. AT1R regulates macrophage polarization through YAP and regulates aortic dissection incidence. Front. Physiol. 2021, 12, 644903. [Google Scholar] [CrossRef] [PubMed]
  50. Magnani, F.; Pappas, C.G.; Crook, T.; Magafa, V.; Cordopatis, P.; Ishiguro, S.; Ohta, N.; Selent, J.; Bosnyak, S.; Jones, E.S.; et al. Electronic sculpting of ligand-GPCR subtype selectivity: The case of angiotensin II. ACS Chem. Biol. 2014, 9, 1420–1425. [Google Scholar] [CrossRef] [Green Version]
  51. Lu, S.; He, X.; Yang, Z.; Chai, Z.; Zhou, S.; Wang, J.; Rehman, A.U.; Ni, D.; Pu, J.; Sun, J.; et al. Activation pathway of a G protein-coupled receptor uncovers conformational intermediates as targets for allosteric drug design. Nat. Commun. 2021, 12, 1–15. [Google Scholar] [CrossRef]
  52. Burghi, V.; Echeverría, E.B.; Sosa, M.H.; Quiroga, D.T.; Muñoz, M.C.; Davio, C.; Monczor, F.; Fernández, N.C.; Dominici, F.P. Participation of Gαi-Adenylate Cyclase and ERK1/2 in Mas Receptor Signaling Pathways. Front. Pharmacol. 2019, 10, 146. [Google Scholar] [CrossRef]
  53. Zhang, H.; Han, G.W.; Batyuk, A.; Ishchenko, A.; White, K.L.; Patel, N.; Sadybekov, A.; Zamlynny, B.; Rudd, M.T.; Hollenstein, K.; et al. Structural basis for selectivity and diversity in angiotensin II receptors. Nature 2017, 544, 327–332. [Google Scholar] [CrossRef]
  54. Sadybekov, A.; Katritch, V. Breaking the Enigma Code of angiotensin II type 2 receptor signaling. Structure 2020, 28, 390–392. [Google Scholar] [CrossRef]
  55. Tóth, A.D.; Turu, G.; Hunyady, L.; Balla, A. Novel mechanisms of G-protein-coupled receptors functions: AT1 angiotensin receptor acts as a signaling hub and focal point of receptor cross-talk. Best Pract. Res. Clin. Endocrinol. Metab. 2018, 32, 69–82. [Google Scholar] [CrossRef] [Green Version]
  56. Thillaiappan, N.B.; Chakraborty, P.; Hasan, G.; Taylor, C.W. IP3 receptors and Ca2+ entry. Biochim. Biophys. Acta (BBA) Mol. Cell Res. 2019, 1866, 1092–1100. [Google Scholar] [CrossRef]
  57. Leppänen, T.; Tuominen, R.K.; Moilanen, E. Protein kinase C and its inhibitors in the regulation of inflammation: Inducible nitric oxide synthase as an example. Basic Clin. Pharmacol. Toxicol. 2014, 114, 37–43. [Google Scholar] [CrossRef]
  58. Kim, H.; Zamel, R.; Bai, X.-H.; Liu, M. PKC activation induces inflammatory response and cell death in human bronchial epithelial cells. PLoS ONE 2013, 8, e64182. [Google Scholar] [CrossRef] [Green Version]
  59. Gray, R.D.; Lucas, C.D.; MacKellar, A.; Li, F.; Hiersemenzel, K.; Haslett, C.; Davidson, D.J.; Rossi, A.G. Activation of conventional protein kinase C (PKC) is critical in the generation of human neutrophil extracellular traps. J. Inflamm. 2013, 10, 1–8. [Google Scholar] [CrossRef] [Green Version]
  60. Dalal, P.J.; Muller, W.A.; Sullivan, D.P. Endothelial cell calcium signaling during barrier function and inflammation. Am. J. Pathol. 2020, 190, 535–542. [Google Scholar] [CrossRef] [Green Version]
  61. Manglik, A.; Wingler, L.M.; Rockman, H.A.; Lefkowitz, R.J. β-Arrestin–Biased Angiotensin II Receptor Agonists for COVID-19. Circulation 2020, 142, 318–320. [Google Scholar] [CrossRef]
  62. Iwasaki, M.; Saito, J.; Zhao, H.; Sakamoto, A.; Hirota, K.; Ma, D. Inflammation triggered by SARS-CoV-2 and ace2 augment drives multiple organ failure of severe COVID-19: Molecular mechanisms and implications. Inflammation 2021, 44, 13–34. [Google Scholar] [CrossRef]
  63. Angers, S.; Salahpour, A.; Bouvier, M. Dimerization: An emerging concept for G protein–coupled receptor ontogeny and function. Annu. Rev. Pharmacol. Toxicol. 2002, 42, 409–435. [Google Scholar] [CrossRef] [Green Version]
  64. Faron-Górecka, A.; Szlachta, M.; Kolasa, M.; Solich, J.; Górecki, A.; Kuśmider, M.; Żurawek, D.; Dziedzicka-Wasylewska, M. Understanding GPCR dimerization. Methods Cell Biol. 2019, 149, 155–178. [Google Scholar]
  65. Bulenger, S.; Marullo, S.; Bouvier, M. Emerging role of homo-and heterodimerization in G-protein-coupled receptor biosynthesis and maturation. Trends Pharmacol. Sci. 2005, 26, 131–137. [Google Scholar] [CrossRef]
  66. Gahbauer, S.; Böckmann, R.A. Membrane-mediated oligomerization of G protein coupled receptors and its implications for GPCR function. Front. Physiol. 2016, 7, 494. [Google Scholar] [CrossRef] [Green Version]
  67. Rukavina Mikusic, N.L.; Silva, M.G.; Pineda, A.M.; Gironacci, M.M. Angiotensin Receptors Heterodimerization and Trafficking: How Much Do They Influence Their Biological Function? Front. Pharmacol. 2020, 11, 1179. [Google Scholar] [CrossRef] [PubMed]
  68. Abdel Hameid, R.; Cormet-Boyaka, E.; Kuebler, W.M.; Uddin, M.; Berdiev, B.K. SARS-CoV-2 may hijack GPCR signaling pathways to dysregulate lung ion and fluid transport. Am. J. Physiol. Lung Cell. Mol. Physiol. 2021, 320, L430–L435. [Google Scholar] [CrossRef] [PubMed]
  69. Yu, L.; Al-Khalili, O.; Duke, B.J.; Stockand, J.D.; Eaton, D.C.; Bao, H.-F. The inhibitory effect of Gβγ and Gβ isoform specificity on ENaC activity. Am. J. Physiol. -Ren. Physiol. 2013, 305, F1365–F1373. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Saint-Criq, V.; Gray, M.A. Role of CFTR in epithelial physiology. Cell. Mol. Life Sci. 2017, 74, 93–115. [Google Scholar] [CrossRef] [Green Version]
  71. Berdiev, B.K.; Qadri, Y.J.; Benos, D.J. Assessment of the CFTR and ENaC association. Mol. BioSyst. 2009, 5, 123–127. [Google Scholar] [CrossRef] [Green Version]
  72. Noreng, S.; Bharadwaj, A.; Posert, R.; Yoshioka, C.; Baconguis, I. Structure of the human epithelial sodium channel by cryo-electron microscopy. Elife 2018, 7, e39340. [Google Scholar] [CrossRef]
  73. Planès, C.; Randrianarison, N.H.; Charles, R.P.; Frateschi, S.; Cluzeaud, F.; Vuagniaux, G.; Soler, P.; Clerici, C.; Rossier, B.C.; Hummler, E. ENaC-mediated alveolar fluid clearance and lung fluid balance depend on the channel-activating protease 1. EMBO Mol. Med. 2010, 2, 26–37. [Google Scholar] [CrossRef]
  74. Ji, H.-L.; Song, W.; Gao, Z.; Su, X.-F.; Nie, H.-G.; Jiang, Y.; Peng, J.-B.; He, Y.-X.; Liao, Y.; Zhou, Y.-J.; et al. SARS-CoV proteins decrease levels and activity of human ENaC via activation of distinct PKC isoforms. Am. J. Physiol. Lung Cell. Mol. Physiol. 2009, 296, L372–L383. [Google Scholar] [CrossRef] [Green Version]
  75. Chen, X.-J.; Seth, S.; Yue, G.; Kamat, P.; Compans, R.W.; Guidot, D.; Brown, L.A.; Eaton, D.C.; Jain, L. Influenza virus inhibits ENaC and lung fluid clearance. Am. J. Physiol. Lung Cell. Mol. Physiol. 2004, 287, L366–L373. [Google Scholar] [CrossRef]
  76. Bhalla, V.; Hallows, K.R. Mechanisms of ENaC regulation and clinical implications. J. Am. Soc. Nephrol. 2008, 19, 1845–1854. [Google Scholar] [CrossRef] [Green Version]
  77. Bilbao, P.S.; Katz, S.; Boland, R. Interaction of purinergic receptors with GPCRs, ion channels, tyrosine kinase and steroid hormone receptors orchestrates cell function. Purinergic Signal. 2012, 8, 91–103. [Google Scholar] [CrossRef] [Green Version]
  78. Pochynyuk, O.; Tong, Q.; Staruschenko, A.; Stockand, J.D. Binding and direct activation of the epithelial Na+ channel (ENaC) by phosphatidylinositides. J. Physiol. 2007, 580, 365–372. [Google Scholar] [CrossRef]
  79. Carvelli, J.; Demaria, O.; Vély, F.; Batista, L.; Benmansour, N.C.; Fares, J.; Carpentier, S.; Thibult, M.-L.; Morel, A.; Remark, R.; et al. Association of COVID-19 inflammation with activation of the C5a–C5aR1 axis. Nature 2020, 588, 146–150. [Google Scholar] [CrossRef]
  80. Yang, L.V.; Oppelt, K.A.; Thomassen, M.J.; Marie, M.A.; Nik Akhtar, S.; McCallen, J.D. Can GPR4 Be a Potential Therapeutic Target for COVID-19? Front. Med. 2021, 7, 1150. [Google Scholar] [CrossRef]
Figure 1. A typical beta2 adrenergic G protein coupled protein (GPCR) coupled to Gs family of Gα protein.
Figure 1. A typical beta2 adrenergic G protein coupled protein (GPCR) coupled to Gs family of Gα protein.
Blsf 07 00019 g001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Nejat, R.; Sadr, A.S.; Torshizi, M.F.; Najafi, D.J. GPCRs of Diverse Physiologic and Pathologic Effects with Fingerprints in COVID-19. Biol. Life Sci. Forum 2021, 7, 19. https://doi.org/10.3390/ECB2021-10261

AMA Style

Nejat R, Sadr AS, Torshizi MF, Najafi DJ. GPCRs of Diverse Physiologic and Pathologic Effects with Fingerprints in COVID-19. Biology and Life Sciences Forum. 2021; 7(1):19. https://doi.org/10.3390/ECB2021-10261

Chicago/Turabian Style

Nejat, Reza, Ahmad Shahir Sadr, Maziar Fayaz Torshizi, and David J. Najafi. 2021. "GPCRs of Diverse Physiologic and Pathologic Effects with Fingerprints in COVID-19" Biology and Life Sciences Forum 7, no. 1: 19. https://doi.org/10.3390/ECB2021-10261

Article Metrics

Back to TopTop