Introduction

Regulator of G protein signaling (RGS) proteins, which modulate G protein-coupled receptors (GPCRs, located in cellular membrane and transmit outer signals into the intra-cellular environment) function, facilitate various downstream cellular signaling through regulating kinds of heterotrimeric G proteins by the acceleration of the intrinsic guanosine triphosphatase (GTPase) activity of their Gα subunits [1, 2]. In most cases, regulation of RGS proteins gives rise to the inhibition of multiple downstream G protein signaling pathways [3], and RGS proteins are thereby recognized by many researchers as the important downstream nodes of those GPCRs [4]. Since the discovery of RGS proteins in different species including yeast, Caenorhabditis elegans as well as mammalian cells in the 1990s [5,6,7,8], their pivotal role in altering cell proliferation, survival and death via controlling downstream cellular signaling activities has provided with the evidence that RGS proteins are potentially involved in sustaining normal physiological functions and that dysregulation of RGS proteins is closely associated with pathologies of many diseases such as cancer. In this review, we summarize the history and structure of RGS, and its role in cancer, and further discuss the molecular mechanisms governing the expression of RGS proteins, offering implications of these new discoveries for novel targeted drug development and related cancer therapy in the future.

A brief description for history of RGS protein discovery

The discovery of RGS proteins is achieved through a series of studies by different experimental systems. In the period of 1995 to 1997, experiments in Saccharomyces cerevisiae revealed the novel factor Sst2 that modulates Gpa1 (a G subunit in yeast) is involved in the regulation of pheromone sensitivity [9, 10]. Additional work performed in 1996 using the nematode C. elegans detected the mutations in the gene egl-10, which reflected mutations in GOA1 that participates in other signalings and in mammals are analogous to G proteins [7]. Later in 1997, egl-10 and Sst2 were genes found to share similar sequences to each other, and then several groups proposed that they could be a potentially new class of GPCR regulators in mammals [1, 7, 8]. Subsequently, the importance of these novel findings were proved by various experiments in rapid publication of papers from several independent research groups, and the main results and conclusions are as follows (Fig. 1): (1) RGS proteins bound with the ∝ subunits of G-protein directly; (2) the interaction of RGS with these subunits potentiated the GTP hydrolysis rate by G∝ (referring to GAP activity); (3) different RGS proteins specifically recognized their targeted G subunits, respectively [2, 6, 11]. The mechanisms underlying RGS protein activity regulation was further deciphered by a report later, showing how those proteins catalytically promoted GTP hydrolysis by G∝ subunits through stabilizing the transition state for GTP hydrolysis, and this finding established the canonical functions of RGS proteins, including GTPase-activating or GAP activity [5].

Fig. 1
figure 1

The schematic graph showing canonical regulation of GPCR signaling by RGS proteins. Upon bound with some agonist, GPCRs undergo a conformation change that facilitates the exchange of GDP for GTP on the a subunit of the heterotrimeric complex. Both GTP-bound Ga in the active form and the released Gbg dimer can subsequently stimulate the corresponding downstream signaling. RGS proteins are GAPs for Ga, which function to terminate signalling through GPCRs by accelerating the intrinsic GTPase activity of Ga and promoting reassociation of the heterotrimeric complex with the receptor at the cell membrane

RGS protein family and RGS protein structure

In mammals, members of the R4 family of RGS proteins were the first ones clarified and studied, which are now typified by RGS4. Among all the RGS proteins, the RGS4 family represents the least structurally and functionally complex. To date, members of RGS proteins have been divided into different families, based on their varied structures and functions. The different RGS proteins were established and were named after their prototypical members, including A/RZ family, B/R4 family, C/R7 family, D/R12 family, E/RA family, F/GEF family and G/GRK family, among which, the A/RZ, B/R4, C/R7 and D/R12 families constitute the canonical RGS proteins [12] (Fig. 2). All the canonical RGS proteins possess the conserved RGS domain with a length of approximately 120 amino acids (aa), which is consisted of nine α-helices structures that can be subdivided further into two subdomains [4]: (1) the first subdomain that forms a smaller helix bundle and is consisted of series of helices including αI, II, III, VIII and IX; and (2) the larger bundle subdomain comprises four helices including αIV, V, VI and VII [13]. Different from the B/R4 family, which is with RGS4 as its prototypical member, the other RGS proteins contain multiple domains that participate in the interaction with various proteins beyond the G∝ and possess more complex domains of cellular function, such as the domain from presence in proteins PSD-95, Dlg and ZO-1/2 domains (PDZ domains), G-protein ∝-like domains (GGL), domain present in disheveled and axin (DAX) domains, kinase domains, Dbl homology/pleckstrin homology domains (DH), G-protein regulatory motif (GoLoco) domains, ∝-catenin–binding domains as well as Ras-binding domains (RBD). To date, there have been at least 20 distinct RGS proteins classified, which play various regulatory roles and can be divided into seven families [14]: A/RZ family includes RGS17, RGS19 and RGS20; B/R4 family includes RGS1, RGS2, RGS3, RGS4, RGS5, RGS8, RGS13, RGS16, RGS18, RGS21; C/R7 family includes RGS6, RGS7, RGS9 and RGS11; D/R12 family includes RGS10, RGS12 and RGS14; E/RA family includes Axin and Axin2; F/GEF family includes P115-RhoGEF, GRK2 and RGS22; G/GRK family includes GRK1, GRK4, GRK5, GRK6 and GRK7. However, as some RGS proteins with a number of genetic variations continue to be revealed, the number of new RGS proteins discovered is still increasing, such as the RGS6 protein that possesses several splicing variants with varied functions and cellular localization [15], and the RGS14 protein with genetic variants that disrupt downstream signaling activation [16].

Fig. 2
figure 2

The schematic lists of the family of RGS proteins. The different RGS proteins were established and were named after their prototypical members, and the A/RZ, B/R4, C/R7 and D/R12 families constitute the canonical RGS proteins

A previous study in 1997 has already identified the structure of RGS protein with the classification of RH (RGS homology) domain [13]. In that study, the crucial structural determinants of the interaction of RGS protein with G determinants of the interaction of RGS protein with G∝ have also been revealed, which has established the structural basis for its GAP activity. As their function of negative mediators in G-protein signaling, RGS proteins are found to mainly exert their effects on regulating GAP activity on α subunits of G-proteins, particularly the Gi/o and Gq families of G-proteins. Although there are yet no reports confirming the GAP activity of any RGS domain against Gαs, emerging evidence has come out that RGS proteins are able to indirectly regulate Gαs downstream signaling through their interaction with subtypes of adenylate cyclase (AC) [17]. Despite the functions of the non-RH domains in RGS proteins, the RH domain is still most studied today, which is attracting the attention of researchers around the world for identifying and develo** novel inhibitors to suppress RGS activity to control kinds of downstream cellular signaling and to further help provide interventions of related diseases.

Mechanisms regulating RGS expression

Previous studies have provided evidence that the levels of RGS proteins are initially associated with the mechanisms that mediate the local concentration of those proteins at the site of a cell signaling. In addition, RGSexpression is also affected by other factors, including its regulation of protein stability, regulation at transcriptional levels, epigenetic regulation, regulation of subcellular localization as well as the environmental conditions such as hypoxia [3, 18,19,20,21,22,23,24,25] (Fig. 3), which allow RGS protein levels to be altered at both an acute and a chronic manner.

Fig. 3
figure 3

Summary of mechanisms regulating RGS proteins expression. RGS protein expression is affected by different factors, including regulation of protein stability (degradation regulation and post-translational modification), transcriptional regulation, epigenetic regulation (DNA methylation and histone deacetylation), and other factors such as hypoxia

Regulation of RGS protein stability

Protein degradation is a dynamic but essential process employed by all of the cells to efficiently and precisely modulate the levels of stable proteins, resulting in the proper functions of those proteins for cells [26, 27]. The degradation commonly undergoes through either of the two ways, including (1) lysosomal proteolysis pathway; and (2) ubiquitin proteasome pathway [28]. Among the two pathways, the lysosomal proteolysis is triggered by proteins such as the lysosomal engulf proteins and the associated digestive enzymes, while the ubiquitin proteasome pathway degrades proteins through regulating poly-ubiquitination of the targeted proteins. During this process, the proteins that complete poly-ubiquitination can be recognized by a large and complex molecular machine, the proteasome complex, which subsequently binds to and degrades the targeted proteins eventually. Evidence has emerged that multiple enzymes participate in regulating the ubiquitin proteasome-dependent protein degradation, including the ubiquitin-activating enzymes (E1), the ubiquitin-conjugating enzymes (E2) and the ubiquitin ligases (E3), and compared to the lysosomal degradation pathway, ubiquitin proteasome pathway requires more energy.

The expression of RGS proteins is affected by their protein stability. In the past decade, previous studies have demonstrated the potential role of RGS4 as a target for degradation by proteasome [29]. The mechanisms underlying RGS4 protein degradation is due the regulation by the N-end rule pathway, a subset of the ubiquitin-mediated proteolytic pathway. The N-end rule pathway potentiates the targeted protein degradation by its recognition of the certain amino acid residues at the N-termini of those proteins. Based on the N-termini residue, the N-end rule pathway can be further subdivided into three types in eukaryotes, including the Arg/N-end rule pathway, the Ac/N-end rule pathway and the Pro/N-end rule pathway, which correspondingly recognizes the basic, acidic, amidated, and bulky hydrophobic N-termini residues, the Nα-terminally acetylated N-termini residues, and the N-termini-Pro residue or a Pro residue at position 2 in the presence of adjoining sequence motifs [30, 31]. A previous report has indicated that blocking of the N-end rule pathway efficiently suppresses the degradation and ubiquitination of RGS4 proteins in the reticulocyte lysate system [29], while the existence of MG132, a kind of proteasome inhibitor, inhibits RGS4 protein degradation but concurrently increased the protein levels of RGS4 that were poly-ubiquitinated, which further provides with the evidence that RGS4 protein is degraded through the N-end rule pathway. Interestingly, RGS4 protein degradation is also observed to be regulated by nitric oxide, which oxidizes the N-termini-cysteine residue that is necessary for the subsequent arginylation [32]. In addition to RGS4 protein, some other RGS proteins are also involved in the regulation by the N-end rule pathway, including RGS16, RGS5 as well as RGS2 [32,33,34]. Particularly, unlike RGS4, the proteasomal degradation of RGS2 protein requires a protein complex that includes DNA damage binding protein 1 (DDB1), F-box only protein 44 (FBXO44) and cullin 4B (CUL4B) [35]. Recently, the expression of RGS proteins has also been found to be affected by their stability through other mechanisms, such as the RGS9-2 protein that is specifically expressed in striatal neurons and functions mainly in the brain [36]. It is reported that, different from many other RGS proteins, RGS9-2 protein is mainly mediated by the lysosomal degradation pathway, and the proteolytic stability of RGS9-2 protein is controlled by R7 family binding protein (R7BP), which is determined as a newly discovered partner for RGS9-2. Another example is the RGS7 protein that belongs to the R7 family of RGS proteins, whose stabilization is specifically modulated by the binding partner Gβ5 [37]. Post-translational modifications, such as protein phosphorylation, are also confirmed to be strongly associated with RGS protein stabilization. Phosphorylation plays a key role in regulating the activities of a variety of cellular signaling pathways by affecting the expression, localization and stabilization of the targeted proteins, which consequently leads to alterations in cell proliferation, apoptosis, survival, mobility and possible malignancy as well [38]. To date, multiple phosphorylating sites have been figured out in different RGS proteins, which give rise to alterations in stability and activity of the RGS proteins. One example is the RGS16 protein that is phosphorylated constitutively at serine 194 (Ser 194) site and undergoes dynamical phosphorylation at the Ser 53 site induced by the activation of α2A-adrenoceptor. The altered phosphorylation levels of RGS16 protein contribute to the inhibition of GAP activity in RGS16, whereas RG16 phosphorylation at Tyr 168 potentiates not only the GAP activity but also the stability of RGS16 protein [39, 40], suggesting RGS16 levels and functions are closely associated with its phosphorylation status. One phenomenon that attracts many researchers’ attention is that, during the phosphorylation regulation, different RGS proteins can be affected by one same kinase. One famous example is the protein kinase A (PKA), a cAMP-dependent protein kinase that is involved in affecting various diseases including cancers in human [41, 42]. PKA is demonstrated to enhance RGS13 activity to negatively regulate CREB-induced transcription of target genes by facilitating the nuclear localization of RGS13, and concurrently, PKA blocks the proteasome degradation of RGS13 protein by its induction in phosphorylation at Thr 41 of RGS13 [43]. Similarly, PKA activation is also involved in promoting the nuclear trans-location of RGS10 protein. In addition to phosphorylation, other post-translational modifications have also been shown to be associated with RGS protein stability, such as protein palmitoylation [

Availability of data and materials

Not applicable.

References

  1. Koelle MR. A new family of G-protein regulators—the RGS proteins. Curr Opin Cell Biol. 1997;9(2):143–7.

    Article  CAS  PubMed  Google Scholar 

  2. Watson N, Linder ME, Druey KM, Kehrl JH, Blumer KJ. RGS family members: GTPase-activating proteins for heterotrimeric G-protein alpha-subunits. Nature. 1996;383(6596):172–5.

    Article  CAS  PubMed  Google Scholar 

  3. Alqinyah M, Hooks SB. Regulating the regulators: epigenetic, transcriptional, and post-translational regulation of RGS proteins. Cell Signal. 2018;42:77–87.

    Article  CAS  PubMed  Google Scholar 

  4. Masuho I, Balaji S, Muntean BS, Skamangas NK, Chavali S, Tesmer JJG, Babu MM, Martemyanov KA. A global map of G protein signaling regulation by RGS proteins. Cell. 2020;183(2):503–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Berman DM, Kozasa T, Gilman AG. The GTPase-activating protein RGS4 stabilizes the transition state for nucleotide hydrolysis. J Biol Chem. 1996;271(44):27209–12.

    Article  CAS  PubMed  Google Scholar 

  6. Berman DM, Wilkie TM, Gilman AG. GAIP and RGS4 are GTPase-activating proteins for the Gi subfamily of G protein alpha subunits. Cell. 1996;86(3):445–52.

    Article  CAS  PubMed  Google Scholar 

  7. Koelle MR, Horvitz HR. EGL-10 regulates G protein signaling in the C. elegans nervous system and shares a conserved domain with many mammalian proteins. Cell. 1996;84(1):115–25.

    Article  CAS  PubMed  Google Scholar 

  8. Siderovski DP, Hessel A, Chung S, Mak TW, Tyers M. A new family of regulators of G-protein-coupled receptors? Curr Biol. 1996;6(2):211–2.

    Article  CAS  PubMed  Google Scholar 

  9. Dohlman HG, Song J, Ma D, Courchesne WE, Thorner J. Sst2, a negative regulator of pheromone signaling in the yeast Saccharomyces cerevisiae: expression, localization, and genetic interaction and physical association with Gpa1 (the G-protein alpha subunit). Mol Cell Biol. 1996;16(9):5194–209.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Dohlman HG, Apaniesk D, Chen Y, Song J, Nusskern D. Inhibition of G-protein signaling by dominant gain-of-function mutations in Sst2p, a pheromone desensitization factor in Saccharomyces cerevisiae. Mol Cell Biol. 1995;15(7):3635–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Rimler A, Jockers R, Lupowitz Z, Zisapel N. Gi and RGS proteins provide biochemical control of androgen receptor nuclear exclusion. J Mol Neurosci. 2007;31(1):1–12.

    Article  CAS  PubMed  Google Scholar 

  12. Almutairi F, Lee JK, Rada B. Regulator of G protein signaling 10: structure, expression and functions in cellular physiology and diseases. Cell Signal. 2020;75:109765.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Tesmer JJ, Berman DM, Gilman AG, Sprang SR. Structure of RGS4 bound to AlF4–activated G(i alpha1): stabilization of the transition state for GTP hydrolysis. Cell. 1997;89(2):251–61.

    Article  CAS  PubMed  Google Scholar 

  14. Willars GB. Mammalian RGS proteins: multifunctional regulators of cellular signalling. Semin Cell Dev Biol. 2006;17(3):363–76.

    Article  CAS  PubMed  Google Scholar 

  15. Ahlers KE, Chakravarti B, Fisher RA. RGS6 as a novel therapeutic target in CNS diseases and cancer. AAPS J. 2016;18(3):560–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Friedman PA, Sneddon WB, Mamonova T, Montanez-Miranda C, Ramineni S, Harbin NH, Squires KE, Gefter JV, Magyar CE, Emlet DR, Hepler JR. RGS14 regulates PTH- and FGF23-sensitive NPT2A-mediated renal phosphate uptake via binding to the NHERF1 scaffolding protein. J Biol Chem. 2022;298(5):101836.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Roy AA, Baragli A, Bernstein LS, Hepler JR, Hebert TE, Chidiac P. RGS2 interacts with Gs and adenylyl cyclase in living cells. Cell Signal. 2006;18(3):336–48.

    Article  CAS  PubMed  Google Scholar 

  18. Song L, Zmijewski JW, Jope RS. RGS2: regulation of expression and nuclear localization. Biochem Biophys Res Commun. 2001;283(1):102–6.

    Article  CAS  PubMed  Google Scholar 

  19. Kim Y, Ghil S. Regulators of G-protein signaling, RGS2 and RGS4, inhibit protease-activated receptor 4-mediated signaling by forming a complex with the receptor and Galpha in live cells. Cell Commun Signal. 2020;18(1):86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Heximer SP, Lim H, Bernard JL, Blumer KJ. Mechanisms governing subcellular localization and function of human RGS2. J Biol Chem. 2001;276(17):14195–203.

    Article  CAS  PubMed  Google Scholar 

  21. Stoveken HM, Fernandez-Vega V, Muntean BS, Patil DN, Shumate J, Bannister TD, Scampavia L, Spicer TP, Martemyanov KA. Identification of potential modulators of the RGS7/Gbeta5/R7BP complex. SLAS Discov. 2021;26(9):1177–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Bosch DE, Jeck WR, Siderovski DP. Self-activating G protein alpha subunits engage seven-transmembrane regulator of G protein signaling (RGS) proteins and a Rho guanine nucleotide exchange factor effector in the amoeba Naegleria fowleri. J Biol Chem. 2022;298(8):102167.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Ehses J, Fernandez-Moya SM, Schroger L, Kiebler MA. Synergistic regulation of Rgs4 mRNA by HuR and miR-26/RISC in neurons. RNA Biol. 2021;18(7):988–98.

    Article  CAS  PubMed  Google Scholar 

  24. Almutairi F, Tucker SL, Sarr D, Rada B. PI3K/ NF-kappaB-dependent TNF-alpha and HDAC activities facilitate LPS-induced RGS10 suppression in pulmonary macrophages. Cell Signal. 2021;86:110099.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. ** M, Xu S, Cao B, Xu Q, Yan Z, Ren Q, Lin C, Tang C. Regulator of G protein signaling 2 is inhibited by hypoxia-inducible factor-1alpha/E1A binding protein P300 complex upon hypoxia in human preeclampsia. Int J Biochem Cell Biol. 2022;147:106211.

    Article  CAS  PubMed  Google Scholar 

  26. Hanzl A, Winter GE. Targeted protein degradation: current and future challenges. Curr Opin Chem Biol. 2020;56:35–41.

    Article  CAS  PubMed  Google Scholar 

  27. Barghout SH. Targeted protein degradation: an emerging therapeutic strategy in cancer. Anticancer Agents Med Chem. 2021;21(2):214–30.

    Article  CAS  PubMed  Google Scholar 

  28. Schapira M, Calabrese MF, Bullock AN, Crews CM. Targeted protein degradation: expanding the toolbox. Nat Rev Drug Discov. 2019;18(12):949–63.

    Article  CAS  PubMed  Google Scholar 

  29. Davydov IV, Varshavsky A. RGS4 is arginylated and degraded by the N-end rule pathway in vitro. J Biol Chem. 2000;275(30):22931–41.

    Article  CAS  PubMed  Google Scholar 

  30. Dougan DA, Varshavsky A. Understanding the Pro/N-end rule pathway. Nat Chem Biol. 2018;14(5):415–6.

    Article  CAS  PubMed  Google Scholar 

  31. Kim JM, Seok OH, Ju S, Heo JE, Yeom J, Kim DS, Yoo JY, Varshavsky A, Lee C, Hwang CS. Formyl-methionine as an N-degron of a eukaryotic N-end rule pathway. Science. 2018;362(6418):0174.

    Article  Google Scholar 

  32. Hu RG, Sheng J, Qi X, Xu Z, Takahashi TT, Varshavsky A. The N-end rule pathway as a nitric oxide sensor controlling the levels of multiple regulators. Nature. 2005;437(7061):981–6.

    Article  CAS  PubMed  Google Scholar 

  33. Lee MJ, Tasaki T, Moroi K, An JY, Kimura S, Davydov IV, Kwon YT. RGS4 and RGS5 are in vivo substrates of the N-end rule pathway. Proc Natl Acad Sci USA. 2005;102(42):15030–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Bodenstein J, Sunahara RK, Neubig RR. N-terminal residues control proteasomal degradation of RGS2, RGS4, and RGS5 in human embryonic kidney 293 cells. Mol Pharmacol. 2007;71(4):1040–50.

    Article  CAS  PubMed  Google Scholar 

  35. Sjogren B, Swaney S, Neubig RR. FBXO44-mediated degradation of RGS2 protein uniquely depends on a Cullin 4B/DDB1 complex. PLoS ONE. 2015;10(5):e0123581.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Anderson GR, Semenov A, Song JH, Martemyanov KA. The membrane anchor R7BP controls the proteolytic stability of the striatal specific RGS protein, RGS9-2. J Biol Chem. 2007;282(7):4772–81.

    Article  CAS  PubMed  Google Scholar 

  37. Chen CK, Eversole-Cire P, Zhang H, Mancino V, Chen YJ, He W, Wensel TG, Simon MI. Instability of GGL domain-containing RGS proteins in mice lacking the G protein beta-subunit Gbeta5. Proc Natl Acad Sci USA. 2003;100(11):6604–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Taracha A, Kotarba G, Wilanowski T. Methods of analysis of protein phosphorylation. Postepy Biochem. 2017;63(2):137–42.

    PubMed  Google Scholar 

  39. Derrien A, Druey KM. RGS16 function is regulated by epidermal growth factor receptor-mediated tyrosine phosphorylation. J Biol Chem. 2001;276(51):48532–8.

    Article  CAS  PubMed  Google Scholar 

  40. Derrien A, Zheng B, Osterhout JL, Ma YC, Milligan G, Farquhar MG, Druey KM. Src-mediated RGS16 tyrosine phosphorylation promotes RGS16 stability. J Biol Chem. 2003;278(18):16107–16.

    Article  CAS  PubMed  Google Scholar 

  41. Cuello F, Herberg FW, Stathopoulou K, Henning P, Diering S. Regulation of cardiac PKA signaling by cAMP and oxidants. Antioxidants. 2021;10(5):663.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Zhang H, Kong Q, Wang J, Jiang Y, Hua H. Complex roles of cAMP-PKA-CREB signaling in cancer. Exp Hematol Oncol. 2020;9(1):32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Lee HK, Park DW, Bae JH, Kim HJ, Shin DG, Park JS, Lee JG, Lee SJ, Bae YS, Baek SH. RGS2 is a negative regulator of STAT3-mediated Nox1 expression. Cell Signal. 2012;24(3):803–9.

    Article  CAS  PubMed  Google Scholar 

  44. **e Z, Yang Z, Druey KM. Phosphorylation of RGS13 by the cyclic AMP-dependent protein kinase inhibits RGS13 degradation. J Mol Cell Biol. 2010;2(6):357–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Osei-Owusu P, Sun X, Drenan RM, Steinberg TH, Blumer KJ. Regulation of RGS2 and second messenger signaling in vascular smooth muscle cells by cGMP-dependent protein kinase. J Biol Chem. 2007;282(43):31656–65.

    Article  CAS  PubMed  Google Scholar 

  46. **e Z, Liu D, Liu S, Calderon L, Zhao G, Turk J, Guo Z. Identification of a cAMP-response element in the regulator of G-protein signaling-2 (RGS2) promoter as a key cis-regulatory element for RGS2 transcriptional regulation by angiotensin II in cultured vascular smooth muscles. J Biol Chem. 2011;286(52):44646–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Yang J, Huang J, Chatterjee TK, Twait E, Fisher RA. A novel mechanism involving coordinated regulation of nuclear levels and acetylation of NF-YA and Bcl6 activates RGS4 transcription. J Biol Chem. 2010;285(39):29760–9.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Seidl MD, Steingraber AK, Wolf CT, Sur TM, Hildebrandt I, Witten A, Stoll M, Fischer JW, Schmitz W, Muller FU. Transcription factor cAMP response element modulator (Crem) restrains Pdgf-dependent proliferation of vascular smooth muscle cells in mice. Pflugers Arch. 2015;467(10):2165–77.

    Article  CAS  PubMed  Google Scholar 

  49. Li F, Murthy KS, Khalili K, Hu W. Cloning and characterization of rabbit Rgs4 promoter in gut smooth muscle. Gene. 2010;451(1–2):45–53.

    Article  CAS  PubMed  Google Scholar 

  50. Zhang Y, Li F, Liu S, Wang H, Mahavadi S, Murthy KS, Khalili K, Hu W. MEKK1-MKK4-JNK-AP1 pathway negatively regulates Rgs4 expression in colonic smooth muscle cells. PLoS ONE. 2012;7(4):e35646.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Iwaki S, Lu Y, **e Z, Druey KM. p53 negatively regulates RGS13 protein expression in immune cells. J Biol Chem. 2011;286(25):22219–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Hu W, Li F, Mahavadi S, Murthy KS. Interleukin-1beta up-regulates RGS4 through the canonical IKK2/IkappaBalpha/NF-kappaB pathway in rabbit colonic smooth muscle. Biochem J. 2008;412(1):35–43.

    Article  CAS  PubMed  Google Scholar 

  53. Zhang Y, Li F, **ao X, Deng W, Yin C, Zhang T, Murthy KS, Hu W. Regulator of G protein signaling 4 is a novel target of GATA-6 transcription factor. Biochem Biophys Res Commun. 2017;483(3):923–9.

    Article  CAS  PubMed  Google Scholar 

  54. **e S, Li J, Wang JH, Wu Q, Yang P, Hsu HC, Smythies LE, Mountz JD. IL-17 activates the canonical NF-kappaB signaling pathway in autoimmune B cells of BXD2 mice to upregulate the expression of regulators of G-protein signaling 16. J Immunol. 2010;184(5):2289–96.

    Article  CAS  PubMed  Google Scholar 

  55. Li J, Peet GW, Balzarano D, Li X, Massa P, Barton RW, Marcu KB. Novel NEMO/IkappaB kinase and NF-kappa B target genes at the pre-B to immature B cell transition. J Biol Chem. 2001;276(21):18579–90.

    Article  CAS  PubMed  Google Scholar 

  56. Buckbinder L, Velasco-Miguel S, Chen Y, Xu N, Talbott R, Gelbert L, Gao J, Seizinger BR, Gutkind JS, Kley N. The p53 tumor suppressor targets a novel regulator of G protein signaling. Proc Natl Acad Sci USA. 1997;94(15):7868–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Yue H, Li W, Desnoyer R, Karnik SS. Role of nuclear unphosphorylated STAT3 in angiotensin II type 1 receptor-induced cardiac hypertrophy. Cardiovasc Res. 2010;85(1):90–9.

    Article  CAS  PubMed  Google Scholar 

  58. Singh RK, Shi J, Zemaitaitis BW, Muma NA. Olanzapine increases RGS7 protein expression via stimulation of the Janus tyrosine kinase-signal transducer and activator of transcription signaling cascade. J Pharmacol Exp Ther. 2007;322(1):133–40.

    Article  CAS  PubMed  Google Scholar 

  59. ** M, Xu S, Li J, Yao Y, Tang C. MicroRNA-3935 promotes human trophoblast cell epithelial-mesenchymal transition through tumor necrosis factor receptor-associated factor 6/regulator of G protein signaling 2 axis. Reprod Biol Endocrinol. 2021;19(1):134.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Grant SL, Lassegue B, Griendling KK, Ushio-Fukai M, Lyons PR, Alexander RW. Specific regulation of RGS2 messenger RNA by angiotensin II in cultured vascular smooth muscle cells. Mol Pharmacol. 2000;57(3):460–7.

    Article  CAS  PubMed  Google Scholar 

  61. Ali Syeda Z, Langden SSS, Munkhzul C, Lee M, Song SJ. Regulatory mechanism of MicroRNA expression in cancer. Int J Mol Sci. 2020;21(5):1723.

    Article  PubMed  PubMed Central  Google Scholar 

  62. Wang SL, Dong XW, Zhao F, Li CX. MiR-203 inhibits cell proliferation, invasion, and migration of ovarian cancer through regulating RGS17. J Biol Regul Homeost Agents. 2021;35(3):1109–15.

    CAS  PubMed  Google Scholar 

  63. Lan T, Li Y, Fan C, Wang L, Wang W, Chen S, Yu SY. MicroRNA-204-5p reduction in rat hippocampus contributes to stress-induced pathology via targeting RGS12 signaling pathway. J Neuroinflammation. 2021;18(1):243.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. **ao D, Gao HX. Mechanism of miR-107-targeting of regulator of G-protein signaling 4 in hepatocellular carcinoma. Oncol Lett. 2019;18(5):5145–54.

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Li W, Si X, Yang J, Zhang J, Yu K, Cao Y. Regulator of G-protein signalling 3 and its regulator microRNA-133a mediate cell proliferation in gastric cancer. Arab J Gastroenterol. 2020;21(4):237–45.

    Article  PubMed  Google Scholar 

  66. **e Z, Geiger TR, Johnson EN, Nyborg JK, Druey KM. RGS13 acts as a nuclear repressor of CREB. Mol Cell. 2008;31(5):660–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Zhao Z, Shilatifard A. Epigenetic modifications of histones in cancer. Genome Biol. 2019;20(1):245.

    Article  PubMed  PubMed Central  Google Scholar 

  68. Arif M, Sadayappan S, Becker RC, Martin LJ, Urbina EM. Epigenetic modification: a regulatory mechanism in essential hypertension. Hypertens Res. 2019;42(8):1099–113.

    Article  CAS  PubMed  Google Scholar 

  69. Zhang W, Song M, Qu J, Liu GH. Epigenetic modifications in cardiovascular aging and diseases. Circ Res. 2018;123(7):773–86.

    Article  CAS  PubMed  Google Scholar 

  70. Morrison AJ. Cancer cell metabolism connects epigenetic modifications to transcriptional regulation. FEBS J. 2022;289(5):1302–14.

    Article  CAS  PubMed  Google Scholar 

  71. Wiechec E, Overgaard J, Hansen LL. A fragile site within the HPC1 region at 1q25.3 affecting RGS16, RGSL1, and RGSL2 in human breast carcinomas. Genes Chromosomes Cancer. 2008;47(9):766–80.

    Article  CAS  PubMed  Google Scholar 

  72. Wolff DW, **e Y, Deng C, Gatalica Z, Yang M, Wang B, Wang J, Lin MF, Abel PW, Tu Y. Epigenetic repression of regulator of G-protein signaling 2 promotes androgen-independent prostate cancer cell growth. Int J Cancer. 2012;130(7):1521–31.

    Article  CAS  PubMed  Google Scholar 

  73. Ying L, Lin J, Qiu F, Cao M, Chen H, Liu Z, Huang Y. Epigenetic repression of regulator of G-protein signaling 2 by ubiquitin-like with PHD and ring-finger domain 1 promotes bladder cancer progression. FEBS J. 2015;282(1):174–82.

    Article  CAS  PubMed  Google Scholar 

  74. Ali MW, Cacan E, Liu Y, Pierce JY, Creasman WT, Murph MM, Govindarajan R, Eblen ST, Greer SF, Hooks SB. Transcriptional suppression, DNA methylation, and histone deacetylation of the regulator of G-protein signaling 10 (RGS10) gene in ovarian cancer cells. PLoS ONE. 2013;8(3):e60185.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Hooks SB, Callihan P, Altman MK, Hurst JH, Ali MW, Murph MM. Regulators of G-Protein signaling RGS10 and RGS17 regulate chemoresistance in ovarian cancer cells. Mol Cancer. 2010;9:289.

    Article  PubMed  PubMed Central  Google Scholar 

  76. Cacan E, Ali MW, Boyd NH, Hooks SB, Greer SF. Inhibition of HDAC1 and DNMT1 modulate RGS10 expression and decrease ovarian cancer chemoresistance. PLoS ONE. 2014;9(1):e87455.

    Article  PubMed  PubMed Central  Google Scholar 

  77. Hurst JH, Hooks SB. Regulator of G-protein signaling (RGS) proteins in cancer biology. Biochem Pharmacol. 2009;78(10):1289–97.

    Article  CAS  PubMed  Google Scholar 

  78. Sjogren B, Neubig RR. Thinking outside of the “RGS box”: new approaches to therapeutic targeting of regulators of G protein signaling. Mol Pharmacol. 2010;78(4):550–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Chaudhary PK, Kim S. An insight into GPCR and G-proteins as cancer drivers. Cells. 2021;10(12):3288.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Latorraca NR, Masureel M, Hollingsworth SA, Heydenreich FM, Suomivuori CM, Brinton C, Townshend RJL, Bouvier M, Kobilka BK, Dror RO. How GPCR phosphorylation patterns orchestrate arrestin-mediated signaling. Cell. 2020;183(7):1813–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Evans PR, Dudek SM, Hepler JR. Regulator of G protein signaling 14: a molecular brake on synaptic plasticity linked to learning and memory. Prog Mol Biol Transl Sci. 2015;133:169–206.

    Article  PubMed  Google Scholar 

  82. Wang Y, Liu G, Ren L, Wang K, Liu A. Long non-coding RNA TUG1 recruits miR29c3p from its target gene RGS1 to promote proliferation and metastasis of melanoma cells. Int J Oncol. 2019;54(4):1317–26.

    CAS  PubMed  Google Scholar 

  83. Roh J, Shin SJ, Lee AN, Yoon DH, Suh C, Park CJ, Huh J, Park CS. RGS1 expression is associated with poor prognosis in multiple myeloma. J Clin Pathol. 2017;70(3):202–7.

    Article  CAS  PubMed  Google Scholar 

  84. Lyu JH, Park DW, Huang B, Kang SH, Lee SJ, Lee C, Bae YS, Lee JG, Baek SH. RGS2 suppresses breast cancer cell growth via a MCPIP1-dependent pathway. J Cell Biochem. 2015;116(2):260–7.

    Article  CAS  PubMed  Google Scholar 

  85. Jiang Z, Wang Z, Xu Y, Wang B, Huang W, Cai S. Analysis of RGS2 expression and prognostic significance in stage II and III colorectal cancer. Biosci Rep. 2010;30(6):383–90.

    Article  CAS  PubMed  Google Scholar 

  86. Linder A, Larsson K, Welen K, Damber JE. RGS2 is prognostic for development of castration resistance and cancer-specific survival in castration-resistant prostate cancer. Prostate. 2020;80(11):799–810.

    Article  CAS  PubMed  Google Scholar 

  87. Xu XH, Sun JM, Chen XF, Zeng XY, Zhou HZ. MicroRNA-494–3p facilitates the progression of bladder cancer by mediating the KLF9/RGS2 axis. Kaohsiung J Med Sci. 2022. https://doi.org/10.1002/kjm2.12588.

    Article  PubMed  Google Scholar 

  88. Hong Z, Hong C, Ma B, Wang Q, Zhang X, Li L, Wang C, Chen D. MicroRNA1263p inhibits the proliferation, migration, invasion, and angiogenesis of triplenegative breast cancer cells by targeting RGS3. Oncol Rep. 2019;42(4):1569–79.

    CAS  PubMed  Google Scholar 

  89. Chen Z, Wu Y, Meng Q, **a Z. Elevated microRNA-25 inhibits cell apoptosis in lung cancer by targeting RGS3. In Vitro Cell Dev Biol Anim. 2016;52(1):62–7.

    Article  CAS  PubMed  Google Scholar 

  90. Wang J, Zhou Y, Fei X, Chen X, Zhu Z. Regulator of G-protein signaling 3 targeted by miR-126 correlates with poor prognosis in gastric cancer patients. Anticancer Drugs. 2017;28(2):161–9.

    Article  CAS  PubMed  Google Scholar 

  91. Tatenhorst L, Senner V, Puttmann S, Paulus W. Regulators of G-protein signaling 3 and 4 (RGS3, RGS4) are associated with glioma cell motility. J Neuropathol Exp Neurol. 2004;63(3):210–22.

    Article  CAS  PubMed  Google Scholar 

  92. Ji L, Fan X, Zhou F, Gu J, Deng X. lncRNA RPL34-AS1 inhibits cell proliferation and invasion while promoting apoptosis by competitively binding miR-3663-3p/RGS4 in papillary thyroid cancer. J Cell Physiol. 2020;235(4):3669–78.

    Article  CAS  PubMed  Google Scholar 

  93. Mu XM, Shi W, Sun LX, Li H, Wang YR, Jiang ZZ, Zhang LY. Pristimerin inhibits breast cancer cell migration by up- regulating regulator of G protein signaling 4 expression. Asian Pac J Cancer Prev. 2012;13(4):1097–104.

    Article  PubMed  Google Scholar 

  94. He Z, Yu L, Luo S, Li Q, Huang S, An Y. RGS4 Regulates Proliferation And Apoptosis Of NSCLC Cells Via microRNA-16 And Brain-Derived Neurotrophic Factor. Onco Targets Ther. 2019;12:8701–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Guda MR, Velpula KK, Asuthkar S, Cain CP, Tsung AJ. Targeting RGS4 ablates glioblastoma proliferation. Int J Mol Sci. 2020;21(9):3300.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Xue X, Wang L, Meng X, Jiao J, Dang N. Regulator of G protein signaling 4 inhibits human melanoma cells proliferation and invasion through the PI3K/AKT signaling pathway. Oncotarget. 2017;8(45):78530–44.

    Article  PubMed  PubMed Central  Google Scholar 

  97. Liu WG, Zhuo L, Lu Y, Wang L, Ji YX, Guo Q. miR-874-3p inhibits cell migration through targeting RGS4 in osteosarcoma. J Gene Med. 2020;22(9):e3213.

    Article  CAS  PubMed  Google Scholar 

  98. Abe Y, Ogasawara S, Akiba J, Naito Y, Kondo R, Nakamura K, Kusukawa J, Yano H. Expression and role of regulator of G-protein signaling 5 in squamous cell carcinoma of the tongue. Clin Exp Dent Res. 2019;5(2):160–9.

    Article  PubMed  PubMed Central  Google Scholar 

  99. Wang D, Xu Y, Feng L, Yin P, Song SS, Wu F, Yan P, Liang Z. RGS5 decreases the proliferation of human ovarian carcinomaderived primary endothelial cells through the MAPK/ERK signaling pathway in hypoxia. Oncol Rep. 2019;41(1):165–77.

    CAS  PubMed  Google Scholar 

  100. Xu C, Li YM, Sun B, Zhong FJ, Yang LY. ATE1 inhibits liver cancer progression through RGS5-mediated suppression of Wnt/beta-catenin signaling. Mol Cancer Res. 2021;19(9):1441–53.

    Article  PubMed  PubMed Central  Google Scholar 

  101. Su S, Shahriyari L. RGS5 plays a significant role in renal cell carcinoma. R Soc Open Sci. 2020;7(4):191422.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Xu Z, Zuo Y, Wang J, Yu Z, Peng F, Chen Y, Dong Y, Hu X, Zhou Q, Ma H, Bao Y, Chen M. Overexpression of the regulator of G-protein signaling 5 reduces the survival rate and enhances the radiation response of human lung cancer cells. Oncol Rep. 2015;33(6):2899–907.

    Article  CAS  PubMed  Google Scholar 

  103. Shen J, Shrestha S, Yen YH, Scott MA, Soo C, Ting K, Peault B, Dry SM, James AW. The pericyte antigen RGS5 in perivascular soft tissue tumors. Hum Pathol. 2016;47(1):121–31.

    Article  CAS  PubMed  Google Scholar 

  104. Yang J, Platt LT, Maity B, Ahlers KE, Luo Z, Lin Z, Chakravarti B, Ibeawuchi SR, Askeland RW, Bondaruk J, Czerniak BA, Fisher RA. RGS6 is an essential tumor suppressor that prevents bladder carcinogenesis by promoting p53 activation and DNMT1 downregulation. Oncotarget. 2016;7(43):69159–72.

    Article  PubMed  PubMed Central  Google Scholar 

  105. Maity B, Stewart A, O’Malley Y, Askeland RW, Sugg SL, Fisher RA. Regulator of G protein signaling 6 is a novel suppressor of breast tumor initiation and progression. Carcinogenesis. 2013;34(8):1747–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Wang Z, Chen J, Wang S, Sun Z, Lei Z, Zhang HT, Huang J. RGS6 suppresses TGF-beta-induced epithelial-mesenchymal transition in non-small cell lung cancers via a novel mechanism dependent on its interaction with SMAD4. Cell Death Dis. 2022;13(7):656.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Jiang N, Xue R, Bu F, Tong X, Qiang J, Liu R. Decreased RGS6 expression is associated with poor prognosis in pancreatic cancer patients. Int J Clin Exp Pathol. 2014;7(7):4120–7.

    PubMed  PubMed Central  Google Scholar 

  108. Qutob N, Masuho I, Alon M, Emmanuel R, Cohen I, Di Pizio A, Madore J, Elkahloun A, Ziv T, Levy R, Gartner JJ, Hill VK, Lin JC, Hevroni Y, Greenberg P, Brodezki A, Rosenberg SA, Kosloff M, Hayward NK, Admon A, Niv MY, Scolyer RA, Martemyanov KA, Samuels Y. RGS7 is recurrently mutated in melanoma and promotes migration and invasion of human cancer cells. Sci Rep. 2018;8(1):653.

    Article  PubMed  PubMed Central  Google Scholar 

  109. Sood R, Bonner TI, Makalowska I, Stephan DA, Robbins CM, Connors TD, Morgenbesser SD, Su K, Faruque MU, Pinkett H, Graham C, Baxevanis AD, Klinger KW, Landes GM, Trent JM, Carpten JD. Cloning and characterization of 13 novel transcripts and the human RGS8 gene from the 1q25 region encompassing the hereditary prostate cancer (HPC1) locus. Genomics. 2001;73(2):211–22.

    Article  CAS  PubMed  Google Scholar 

  110. Caldiran FY, Cacan E. RGS10 suppression by DNA methylation is associated with low survival rates in colorectal carcinoma. Pathol Res Pract. 2022;236:154007.

    Article  CAS  PubMed  Google Scholar 

  111. Chang YF, Huang YQ, Wu KM, Jou AF, Shih NY, Ho JA. Diagnosing the RGS11 lung cancer biomarker: the integration of competitive immunoassay and isothermal nucleic acid exponential amplification reaction. Anal Chem. 2019;91(5):3327–35.

    Article  CAS  PubMed  Google Scholar 

  112. Fu C, Yuan G, Yang ST, Zhang D, Yang S. RGS12 represses oral cancer via the phosphorylation and SUMOylation of PTEN. J Dent Res. 2021;100(5):522–31.

    Article  CAS  PubMed  Google Scholar 

  113. Wang Y, Wang J, Zhang L, Karatas OF, Shao L, Zhang Y, Castro P, Creighton CJ, Ittmann M. RGS12 is a novel tumor-suppressor gene in African American prostate cancer that represses AKT and MNX1 expression. Cancer Res. 2017;77(16):4247–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Li Y, Liu M, Yang S, Fuller AM, Karin Eisinger-Mathason TS, Yang S. RGS12 is a novel tumor suppressor in osteosarcoma that inhibits YAP-TEAD1-Ezrin signaling. Oncogene. 2021;40(14):2553–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Liang G, Bansal G, **e Z, Druey KM. RGS16 inhibits breast cancer cell growth by mitigating phosphatidylinositol 3-kinase signaling. J Biol Chem. 2009;284(32):21719–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Miyoshi N, Ishii H, Sekimoto M, Doki Y, Mori M. RGS16 is a marker for prognosis in colorectal cancer. Ann Surg Oncol. 2009;16(12):3507–14.

    Article  PubMed  Google Scholar 

  117. Carper MB, Denvir J, Boskovic G, Primerano DA, Claudio PP. RGS16, a novel p53 and pRb cross-talk candidate inhibits migration and invasion of pancreatic cancer cells. Genes Cancer. 2014;5(11–12):420–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Sun X, Charbonneau C, Wei L, Chen Q, Terek RM. miR-181a targets RGS16 to promote chondrosarcoma growth Angiogenesis Metastasis. Mol Cancer Res. 2015;13(9):1347–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Huang R, Li G, Zhao Z, Zeng F, Zhang K, Liu Y, Wang K, Hu H. RGS16 promotes glioma progression and serves as a prognostic factor. CNS Neurosci Ther. 2020;26(8):791–803.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Zhang W, Qian S, Yang G, Zhu L, Zhou B, Wang J, Liu R, Yan Z, Qu X. MicroRNA-199 suppresses cell proliferation, migration and invasion by downregulating RGS17 in hepatocellular carcinoma. Gene. 2018;659:22–8.

    Article  CAS  PubMed  Google Scholar 

  121. Bodle CR, Mackie DI, Hayes MP, Schamp JH, Miller MR, Henry MD, Doorn JA, Houtman JCD, James MA, Roman DL. Natural products discovered in a high-throughput screen identified as inhibitors of RGS17 and as cytostatic and cytotoxic agents for lung and prostate cancer cell lines. J Nat Prod. 2017;80(7):1992–2000.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Hu Y, Zheng M, Wang S, Gao L, Gou R, Liu O, Dong H, Li X, Lin B. Identification of a five-gene signature of the RGS gene family with prognostic value in ovarian cancer. Genomics. 2021;113(4):2134–44.

    Article  CAS  PubMed  Google Scholar 

  123. Weisshaar N, Wu J, Ming Y, Madi A, Hotz Wagenblatt A, Ma S, Mieg A, Hering M, Zettl F, Mohr K, Schlimbach T, Ten Bosch N, Hertel F, Muller L, Byren H, Wang M, Borgers H, Munz M, Schmitt L, van der Hoeven F, Kloz U, Carretero R, Schleussner N, Jackstadt RF, Hofmann I, Cui G. Rgs16 promotes antitumor CD8(+) T cell exhaustion. Sci Immunol. 2022;7(71):1873.

    Article  Google Scholar 

  124. Deng M, Wei W, Duan J, Chen R, Wang N, He L, Peng Y, Ma X, Wu Z, Liu J, Li Z, Zhang Z, Jiang L, Zhou F, **e D. ZHX3 promotes the progression of urothelial carcinoma of the bladder via repressing of RGS2 and is a novel substrate of TRIM21. Cancer Sci. 2021;112(5):1758–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. O’Brien JB, Wilkinson JC, Roman DL. Regulator of G-protein signaling (RGS) proteins as drug targets: Progress and future potentials. J Biol Chem. 2019;294(49):18571–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. **e Y, Wolff DW, Wei T, Wang B, Deng C, Kirui JK, Jiang H, Qin J, Abel PW, Tu Y. Breast cancer migration and invasion depend on proteasome degradation of regulator of G-protein signaling 4. Cancer Res. 2009;69(14):5743–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Yang L, Lee MM, Leung MM, Wong YH. Regulator of G protein signaling 20 enhances cancer cell aggregation, migration, invasion and adhesion. Cell Signal. 2016;28(11):1663–72.

    Article  CAS  PubMed  Google Scholar 

  128. Li Y, Li L, Lin J, Hu X, Li B, Xue A, Shen Y, Jiang J, Zhang M, **e J, Zhao Z. Deregulation of RGS17 expression promotes breast cancer progression. J Cancer. 2015;6(8):767–75.

    Article  PubMed  PubMed Central  Google Scholar 

  129. Cacan E. Epigenetic regulation of RGS2 (Regulator of G-protein signaling 2) in chemoresistant ovarian cancer cells. J Chemother. 2017;29(3):173–8.

    Article  CAS  PubMed  Google Scholar 

  130. Altman MK, Alshamrani AA, Jia W, Nguyen HT, Fambrough JM, Tran SK, Patel MB, Hoseinzadeh P, Beedle AM, Murph MM. Suppression of the GTPase-activating protein RGS10 increases Rheb-GTP and mTOR signaling in ovarian cancer cells. Cancer Lett. 2015;369(1):175–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Tso PH, Yung LY, Wang Y, Wong YH. RGS19 stimulates cell proliferation by deregulating cell cycle control and enhancing Akt signaling. Cancer Lett. 2011;309(2):199–208.

    Article  CAS  PubMed  Google Scholar 

  132. Bodle CR, Mackie DI, Roman DL. RGS17: an emerging therapeutic target for lung and prostate cancers. Future Med Chem. 2013;5(9):995–1007.

    Article  CAS  PubMed  Google Scholar 

  133. Bodle CR, Schamp JH, O’Brien JB, Hayes MP, Wu M, Doorn JA, Roman DL. Screen targeting lung and prostate cancer oncogene identifies novel inhibitors of RGS17 and problematic chemical substructures. SLAS Discov. 2018;23(4):363–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Hurst JH, Mendpara N, Hooks SB. Regulator of G-protein signalling expression and function in ovarian cancer cell lines. Cell Mol Biol Lett. 2009;14(1):153–74.

    Article  CAS  PubMed  Google Scholar 

  135. Maity B, Yang J, Huang J, Askeland RW, Bera S, Fisher RA. Regulator of G protein signaling 6 (RGS6) induces apoptosis via a mitochondrial-dependent pathway not involving its GTPase-activating protein activity. J Biol Chem. 2011;286(2):1409–19.

    Article  CAS  PubMed  Google Scholar 

  136. Li G, Wang M, Ren L, Li H, Liu Q, Ouyang Y, He L, Li F. Regulator of G protein signaling 20 promotes proliferation and migration in bladder cancer via NF-kappaB signaling. Biomed Pharmacother. 2019;117:109112.

    Article  CAS  PubMed  Google Scholar 

  137. Cao X, Qin J, **e Y, Khan O, Dowd F, Scofield M, Lin MF, Tu Y. Regulator of G-protein signaling 2 (RGS2) inhibits androgen-independent activation of androgen receptor in prostate cancer cells. Oncogene. 2006;25(26):3719–34.

    Article  CAS  PubMed  Google Scholar 

  138. James MA, Lu Y, Liu Y, Vikis HG, You M. RGS17, an overexpressed gene in human lung and prostate cancer, induces tumor cell proliferation through the cyclic AMP-PKA-CREB pathway. Cancer Res. 2009;69(5):2108–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Li S, Yang H, Li S, Zhao Z, Wang D, Fu W. High expression of regulator of G-protein signalling 1 is associated with the poor differentiation and prognosis of gastric cancer. Oncol Lett. 2021;21(4):322.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Zhang S, Wang H, Liu J, Tao T, Zeng Z, Wang M. RGS1 and related genes as potential targets for immunotherapy in cervical cancer: computational biology and experimental validation. J Transl Med. 2022;20(1):334.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Shi D, Tong S, Han H, Hu X. RGS20 promotes tumor progression through modulating PI3K/AKT signaling activation in penile cancer. J Oncol. 2022;2022:1293622.

    Article  PubMed  PubMed Central  Google Scholar 

  142. Huang J, Yang J, Maity B, Mayuzumi D, Fisher RA. Regulator of G protein signaling 6 mediates doxorubicin-induced ATM and p53 activation by a reactive oxygen species-dependent mechanism. Cancer Res. 2011;71(20):6310–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Liu P, Zhang C, Chen J, Zhang R, Ren J, Huang Y, Zhu F, Li Z, Wu G. Combinational therapy of interferon-alpha and chemotherapy normalizes tumor vasculature by regulating pericytes including the novel marker RGS5 in melanoma. J Immunother. 2011;34(3):320–6.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This paper was funded by Starting Research Foundation of The Children’s Hospital, Zhejiang University School of Medicine (No. 481), Foundation for The Top-Notch Youth Talent Cultivation Project of Independent Design Project of National Clinical Research Center for Child Health (No. Q21A0006), and National Natural Science Foundation of China (No. 31801207) to C. T.

Author information

Authors and Affiliations

Authors

Contributions

Writing—original draft preparation, LL and CT; writing—review and editing, QX and CT; funding acquisition, CT. All authors have read and approved the manuscript.

Corresponding author

Correspondence to Chao Tang.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

The Author confirms: that the work described has not been published before; that it is not under consideration for publication elsewhere; that its publication has been approved by all co-authors.

Competing interests

The authors declare no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, L., Xu, Q. & Tang, C. RGS proteins and their roles in cancer: friend or foe?. Cancer Cell Int 23, 81 (2023). https://doi.org/10.1186/s12935-023-02932-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12935-023-02932-8

Keywords