Abstract
Regulators of G-protein signaling (RGS) proteins are GTPase-activating proteins (GAP) for various Gα subunits of heterotrimeric G proteins. Through this mechanism, RGS proteins regulate the magnitude and duration of G-protein-coupled receptor signaling and are often referred to as fine tuners of G-protein signaling. Increasing evidence suggests that RGS proteins themselves are regulated through multiple mechanisms, which may provide an even finer tuning of G-protein signaling and crosstalk between G-protein-coupled receptors and other signaling pathways. This review summarizes the current data on the control of RGS function through regulated expression, intracellular localization, and covalent modification of RGS proteins, as related to cell function and the pathogenesis of diseases.
Keywords: G protein signaling expression, G protein signaling localization, G protein signaling phosphorylation
this article is part of a collection on G Protein Kinase A Signaling in Cardiovascular Physiology and Disease. Other articles appearing in this collection, as well as a full archive of all collections, can be found online at http://ajpheart.physiology.org/.
Introduction
In the mid-1990's, several investigators (34, 38, 39, 43, 90) identified a new class of proteins that regulated the function of heterotrimeric G proteins in a variety of model systems. Further studies (80, 185) demonstrated that these proteins accelerate the intrinsic GTPase activity of Gα subunits, thereby functioning as GTPase-activating proteins. This family is now collectively known as the regulator of G-protein signaling (RGS) proteins and has been divided into eight subfamilies based on sequence homology (188). All of the RGS family members express the evolutionary conserved RGS domain, which interacts with the GTP-bound Gα subunits and accelerates their GTPase activity, with variable degrees of selectivity among Gαi, Gαq/11, and Gα12/13 subunits. In addition, many RGS proteins possess extensions of various lengths, often containing one or more functional domains other than the RGS box (188). These domains and the regions outside of the RGS box may facilitate interactions with noncanonical partners and mediate G-protein-independent functions, (2, 149) or may serve regulatory purposes as discussed in this review.
Although the biochemical functions of RGS proteins have been extensively studied, our understanding of the physiological functions of RGS proteins is relatively modest. In this emerging field, investigators have positioned RGS proteins as key modulators in the cardiovascular (57, 187, 199) and neurologic (168, 174, 175) systems, lymphocyte trafficking and inflammatory disorders (29, 42), and cancer biology (81). The potential importance of RGS proteins in disease has driven efforts to pharmacologically target RGS protein function (154, 155); however, this approach would be most effective when applied with a greater understanding of RGS regulation. Therefore, identifying the mechanisms that control RGS activity has become an increasing area of investigation.
This review provides a comprehensive summary of the existing data exploring the mechanisms by which RGS proteins are regulated. Common modes of control of RGS activity include regulation of expression, intracellular localization, and posttranslational modifications. Therefore, we have organized this review into these specified modules and provide detailed information on how these mechanisms regulate specific RGS proteins in Tables 1–4. Importantly, a given RGS protein is often regulated by several mechanisms and, vise versa, several RGS proteins can be regulated by one mechanism within the same cell, which is described throughout the text. Furthermore, the regulation of a given RGS protein may differ dependent on the cell type, indicating that the knowledge obtained on a particular RGS protein obtained in one cell type may not be applicable for the other cell type. While the quality of data in the cited (peer-reviewed) publications is quite variable, we invite the readers to ascertain the stringency of the studies with their own criteria. By highlighting our current knowledge of the regulation RGS proteins, we propose potential areas in which research is needed for understanding how the fine tuning of G-protein signaling occurs through regulation of RGS proteins in cells, animal models, and humans.
Table 1.
Regulated expression of RGS proteins
RGS | Stimuli/Disease Models | Expression | mRNA/Protein | Mechanism | Cells/Tissues | Ref. No. |
---|---|---|---|---|---|---|
SST2 | Pheromone | ↑1–6 h | Protein | (GPA1) | Yeast | (37, 39) |
RGS1 | Concanavalin A | ↑1–4 h | mRNA | (Gi), (PKC) | Mononuclear cells | (67) |
RGS1 | SAC/phorbol | ↑2–96 | mRNA | (PKC) | B lymphocytes | (71) |
RGS1 | Anti-CD40 + anti-μ | ↑4–48 h | mRNA | B lymphocytes | (151) | |
RGS1 | LPS | ↑4–48 h | mRNA, protein | Dendritic cells | (150) | |
↑0.25–1 h | mRNA | TLR1/2/4/6/9 | Macrophages | (132) | ||
↓12–24 h | mRNA | TLR1/2/4/6/9 | Macrophages | (132) | ||
RGS1 | IFN-β | ↑4–20 h | mRNA, protein | IFNAR2c receptor | Mononuclear cells | (173) |
RGS1 in vivo | LPS/sepsis | ↑2 h | mRNA | Porcine heart | (122) | |
RGS2 | Concanavalin A | ↑0.5–1.5 h | mRNA | (Gi), (Ca2+) | Mononuclear cells | (67) |
RGS2 | Carbachol | ↑0.5–4 h | mRNA | (Gi/q), PKC | SH-SY5Y cells | (157, 158) |
RGS2 | Angiotensin II | ↑0.5–4 h | mRNA, protein | (Gi/q), PKC, iPLA2, PKA | VSMC | (56, 101, 192, 193) |
↑1–12 h | mRNA | (Gi/q), Ca2+/calmodulin | H295R adrenal cells | (137) | ||
↑1–12 h | mRNA | (Gi/q) | Cardiac fibroblasts | (200) | ||
RGS2 | S1P | ↑1–2 h | mRNA | (Gq) | VSMC | (64) |
RGS2 | Phenylephrine | ↑0.5–3 h | mRNA, protein | (Gi/q) | Cardiomyocytes | (62, 204) |
RGS2 | ATP | ↑2 h | mRNA | (Gi/q) | Osteoblasts | (142) |
RGS2 | Phorbol | ↑1–2 h | mRNA | PKC | VSMC | (56) |
↑0.5–6 h | mRNA | (PKC) | Cardiomyocytes | (62, 204) | ||
↑2 h | mRNA | (PKC) | Osteoblasts | (142) | ||
↑1–3 h | mRNA | PKC | SH-SY5Y cells | (157) | ||
RGS2 | PGE1/2 | ↑2 h | mRNA | (Gs) | T cells | (6) |
RGS2 | Parathyroid hormones | ↑0.5–6 h | mRNA | (Gs), cAMP, PKA | Osteoblasts | (108, 142, 169, 176) |
(PTH, PTHrP) | ↑1–3 h | mRNA | (Gs), cAMP, PKA | UMR106 cells | (89, 176) | |
RGS2 | Isoproterenol | ↑1–3 h | mRNA, protein | (Gs, cAMP) | Cardiomyocytes | (62, 116) |
RGS2 | Thyrotropin | ↑1–24 h | mRNA | (Gs) | Thyroid epithelial cells | (48) |
RGS2 | Cholera toxin | ↑1.5 h | mRNA | Gs, cAMP, PKA | UMR106 cells | (89) |
RGS2 | Forskolin | ↑3 h | mRNA, protein | cAMP, PKA | Cardiomyocytes | (62) |
↑4–24 h | mRNA | (cAMP) | UMR106 cells | (89) | ||
↑4–24 h | mRNA | (cAMP) | VSMC | (28) | ||
↑1–3 h | mRNA, protein | cAMP, PKA | Osteoblasts | (142, 176) | ||
RGS2 | PDGF, FGF | ↑4–24 h | mRNA | VSMC | (28, 60) | |
RGS2 | LPS | ↓1–24 h | mRNA | TLR1/2/4/6/9 | Macrophages | (132) |
↓1–1.5 h | mRNA | TLR4, PKC-η, PLD | Raw264.7 cells | (95) | ||
RGS2 | IL-2 | ↓2 h | mRNA | Reversed by cAMP | T cells | (6) |
RGS2 | IFNβ-1a | ↑72 h | mRNA | Mononuclear leukocytes | (55) | |
RGS2 | H2O2, peroxynitrite | ↑1–4 h | mRNA, protein | 1321N1 cells | (203) | |
RGS2 | Heat shock | ↑0.5–2 h | mRNA | SH-SY5Y cells | (159) | |
↑1–3 h | protein | 1321N1 cells | (203) | |||
RGS2 | Camptothecin, geldanamycin | ↑1–4 h | mRNA | Cdk2 | SH-SY5Y cells | (158) |
RGS2 | Ischemia/recovery | ↑12–24 h | mRNA, protein | PKC | Astrocytes | (47) |
RGS2 | Mechanical stress | ↑6–24 h | mRNA, protein | cAMP | Periodontal ligament cells | (147) |
RGS2 | Differentiation | ↑0.5 h–9 days | mRNA | cAMP, SP1 | 3T3-L1 cells | (26, 82) |
RGS2 | Differentiation | ↓1–5 days | mRNA | SH-SY5Y cells | (159) | |
RGS2 in vivo | PTH, PGE2 | ↑1 h | mRNA | (Gs), inhibited by vitamin D | Bone | (108, 178) |
RGS2 in vivo | Angiotensin II | ↑1 day | mRNA | (Gq) | Isolated cardiac fibroblasts | (200) |
↓3–14 days | ||||||
RGS2 in vivo | Ovulation (hCG) | ↑4–12 h | mRNA | Ovary | (179) | |
RGS2 in vivo | Cardiac hypertrophy: | |||||
Gαq* transgenic mice | ↓0.5–14 mo | mRNA, protein | Gαq | Heart, cardiac myocytes | (202) | |
Aortic constriction | ↓2–7 days | mRNA | Heart | (202) | ||
RGS2 in vivo | Vascular hypertrophy | ↓2 days | mRNA | Angiotensin II dependent | Aorta (abdominal, thoracic) | (182) |
↑14–28 days | ||||||
RGS2 in vivo | Cardiac resynchronization | ↑3 wk | Protein | Heart | (20, 21) | |
RGS3 | Forskolin | ↓4–24 h | mRNA | (PKA) | VSMC | (28) |
RGS3 | FGF | ↑24 h | mRNA | Cardiomyocytes | (201) | |
RGS3 in vivo | Congestive heart failure | ↓16 mo | mRNA, protein | Heart (SHHF rats) | (201) | |
RGS3 in vivo | Cardiac resynchronization | ↑3 wk | Protein | Heart | (20, 21) | |
RGS4 | Angiotensin II | ↑2–12 h | mRNA | (Gq), CaM, CaMK, PKC | H295R cells | (138) |
RGS4 | Opioids | ↑1–8 h | mRNA | Gi | PC12-μOR/κOR transfected cells | (112) |
↓18 h | Protein | Gi, proteasomal degradation | SH-SY5Y cells | (181) | ||
RGS4 | Adenosine | ↓3 h | mRNA | cAMP | PC12 | |
RGS4 | Forskolin | ↓3 h | mRNA | cAMP | PC12 | (127) |
↓24 h | mRNA | (cAMP) | VSMC | (28) | ||
RGS4 | IL-1β | ↑3 h-3 days | mRNA, protein | ERK, p38, NF-kβ | Colonic muscle strips and SMC | (76–78) |
RGS4 | Camptothecin, geldanamycin | ↓1–4 h | mRNA | Cdk2 | SH-SY5Y cells | (158) |
RGS4 | Proteasome inhibitors | ↑4 h | Protein | RGS stabilization | Breast cancer cells | (190) |
RGS4 | FGF | ↑24 h | mRNA | Cardiomyocytes | (201) | |
RGS4 | PDGF-BB, PDGF-DD | ↓6–24 h | mRNA | VSMC | (60) | |
RGS4 | Dedifferentiation | ↓Passages 2–6 | mRNA, protein | VSMC | (66) | |
RGS4 | Differentiation (NGF) | ↓24 h | mRNA, protein | PC12 cells | (93) | |
RGS4 | Differentiation | ↑Day 15 | mRNA | Chondrocytes | (3) | |
RGS4 in vivo | Cardiac hypertrophy | ↑7 days | mRNA | Heart | (201) | |
RGS4 in vivo | Congestive heart failure | ↓16 mo | mRNA, protein | Heart (SHHF rats) | (201) | |
RGS4 in vivo | Low-salt diet | ↑15 days | mRNA | rat adrenal gland | (138) | |
Angiotensin II infusion | ↑2–6 h | mRNA | ||||
RGS4 in vivo | Vascular hypertrophy | ↓2 days | mRNA | Angiotensin II dependent | Aorta (abdominal, thoracic) | (182) |
↑14–28 days | ||||||
RGS4 in vivo | PPAR-δ (apoE−/− mice) | ↑8 wk | mRNA | Atherosclerotic plaque | (5) | |
RGS4 in vivo | LPS treatment | ↑24–72 h | mRNA, Protein | Heart | (124) | |
RGS5 | β2AR-agonsists | ↓4–24 h | mRNA | (Gαs) | Airway SMC | (198) |
RGS5 | Hypoxia | ↑1–12 h | mRNA | HIF-1α | HUVEC | (88) |
RGS5 | PDGF-BB, PDGF-DD | ↓6–24 h | mRNA | VSMC | (60) | |
RGS5 | Dedifferentiation | ↑Passages 2–5 | mRNA | VSMC | (66) | |
RGS5 | Differentiation | ↓Days 6–15 | mRNA | Chondrocytes | (3) | |
RGS5 in vivo | Atherogenic diet | ↓6 mo | mRNA | Atherosclerotic plaque | (99) | |
RGS5 in vivo | PPAR-δ (apoE−/− mice) | ↑8 wk | mRNA | Atherosclerotic plaque | (5) | |
RGS5 in vivo | Isoproterenol | ↑14 days | mRNA, protein | β2AR mediated | Atria | (86) |
RGS5 in vivo | Vascular hypertrophy | ↓2 days | mRNA | Angiotensin II dependent | Aorta (abdominal, thoracic) | (182) |
↑14–28 days | ||||||
RGS7 | Differentiation | ↑Days 6–15 | mRNA | Chondrocytes | (3) | |
RGS7 | TNF-α | ↑4 h | Protein | p38MAPK, RGS7 stabilization | ECV-304 cells | (7) |
TNF-α, LPS | ↑48 h | mRNA, protein | RAW cells, BV2 cells | (63) | ||
RGS7 in vivo | TNF-α, LPS | ↑4–6 h | Protein | TNFR1 | Brain | (7) |
Spinal cord injury | ↑48 h | Protein | Microglia/macrophages | (63) | ||
RGS10 | Cisplatin | ↑24–48 h | mRNA | SKOV-3 cells | (72) | |
RGS10 | Differentiation | ↑Days 9–15 | mRNA | Chondrocytes | (3) | |
RGS10 | Tea polyphenols | ↑4–24 h | mRNA | Colon cancer cells | (104) | |
RGS13 | cAMP | ↓1.5–6 h | mRNA | PKA | Mast cells | (194) |
⇆ | Protein | RGS13 protein stabilization | ||||
RGS13 | Anti-CD40 + anti-μ | ↑4–48 h | mRNA | B lymphocytes | (151) | |
RGS13 | IL-17 | ↑2–4 h | mRNA | B cells | (189) | |
RGS14 | LPS | ↓4–48 h | mRNA, protein | Dendritic cells | (150) | |
RGS14 | Doxorubicin | ↑16 h | mRNA | p53 | RKO, MCF7, U-87, EB1 | (15) |
RGS14 | Tea polyphenols | ↑8–16 h | mRNA | Colon cancer cells | (104) | |
RGS16 | PGE1/2 | ↓4–48 h | mRNA | (Gs) | T cells | (6) |
RGS16 | Endothelin-1 | ↑24 h | Protein | (Gq), Rho | Cardiac myocytes | (161) |
RGS16 | S1P | ↑24 h | Protein | Gi, Rac | Cardiac myocytes | (161) |
↑1–2 h | mRNA | Gi | VSMC | (64) | ||
RGS16 | LPS | ↑2 h | mRNA | NF-κB | Pre-B cells | (100) |
↑24 h | mRNA, protein | IL-1 secretion | Cardiac myocytes | (124, 125) | ||
↑2–6 h | mRNA | VSMC, aorta | (65) | |||
↑4–24 h | mRNA | Dendritic cells | (150) | |||
RGS16 | TNF-α | ↑24 h | Protein | IL-1 secretion | Cardiac myocytes | (125) |
↑1–4 h | mRNA | Inhibited by Ca2+ | T lymphocytes (CEM) | (50) | ||
RGS16 | IL-1 | ↑24 h | Protein | Cardiac myocytes | (125) | |
RGS16 | IL-2 | ↑2 h | mRNA | Inhibited by cAMP | T lymphocytes | (6) |
RGS16 | IL-17 | ↑2–8 h | mRNA, protein | Traf3/6, NF-κB | B cells | (189) |
RGS16 | Phorbol | ↑2–8 h | mRNA | PKC, TNFα secretion | T lymphocytes (CEM) | (50) |
RGS16 | Phitoemagglutinin | ↑72 h | mRNA | Reversed by IFNβ-1a | Mononuclear leukocytes | (55) |
RGS16 | Differentiation | ↑(N/D) | mRNA | Megakaryocytes | (12) | |
RGS16 | Dedifferentiation | ↓Passages 3–6 | mRNA | VSMC | (66) | |
RGS16 in vivo | LPS/sepsis | ↑2 h | mRNA | Heart (porcine) | (122, 124) | |
↑24–72 h | protein | Heart (rat) | ||||
RGS17 | Cisplatin | ↑24–48 h | mRNA | SKOV-3 cells | (72) | |
RGS18 | RANKL | ↓4 days | mRNA, protein | (OGR1/GPR68, Gq) | RAW264.7, monocytes | (84) |
RGS18 | LPS | ↓4–24 h | mRNA | Dendritic cells | (150) | |
RGS18 | Differentiation | ↑(N/D) | mRNA | Megakaryocytes | (12) | |
RGS19/GAIP | Differentiation | ↓20–30 days | mRNA | Gi | HT-29, Caco-2 cells | (118) |
RGS20/RGSZ | LPS | ↑4 h | mRNA | Dendritic cells | (150) |
Data are grouped for each regulator of G-protein signaling (RGS) protein based on stimuli as follows: G-protein-coupled receptor (GPCR) stimuli/signaling (Gq/i followed by Gs), cytokines, stress, growth/differentiation conditions, and in vivo models. GPCR agonists or drugs acting through G-protein signaling are in italics. Data are grouped for each regulators of RGS protein in chronological order of publications. The predicted, but not confirmed, signaling is in parentheses. Functional significance of regulated RGS expression is discussed in the review.
AR, adrenergic receptor; S1P, sphingosine-1-phosphate; HIF-1, hypoxia inducible factor; VSMC, vascular smooth muscle cells; HUVEC, human umbilical vein endothelial cells; PGE2, prostaglandin E2; PTH, parathyroid hormone; TLR, Toll-like receptors; PPAR, peroxisome proliferator-activated receptor; hCG, human chorionic gonadotropin; RANKL, receptor activator of NF-kB ligand; N/D, not described; ↑, up; ↓, down.
Table 2.
Regulation of RGS intracellular localization
RGS | Cell Type | Ectopic/Endogenous | Stimulus/Modification | Localization (From/To) | Ref. No. |
---|---|---|---|---|---|
RGS2 | HEK293 | Ectopic | Expression of active Gαq | Nucleus/plasma membrane | (68) |
RGS2 | HEK293 | Ectopic | Expression of Gαs, Gαq, β2AR, AT1aR | Nucleus/plasma membrane | (141) |
RGS2 | COS-7 | Ectopic | Heat shock, proteotoxic stress | Nucleus/nucleoli | (22) |
RGS2 | VSMC | Endogenous | Phosphorylation by PKGI | Cytoplasm/plasma membrane | (119, 167) |
RGS2 | PC3-AR | Ectopic | Melatonin, 8-bromo-cGMP | Nucleus/cytoplasm | (133) |
RGS2 | HEK293 | Ectopic | Expression of active Gαq, M3R | Cytoplasm and nucleus/plasma membrane | (31) |
RGS2 | HEK | Ectopic | NH2-terminal 71 amino acids | Targets to plasma membrane | (58) |
RGS2 | M1-HEK | Ectopic | R44H (naturally occurring mutation) | Decreased affinity for plasma membrane | (59) |
RGS3 | HMC | Ectopic | Expression of active Gα11, AIF4−, NaF, endothelin-1 | Cytoplasm/plasma membrane | (46) |
RGS3 | Rat astrocyte | Endogenous | Atrial natriuretic peptide | Cytoplasm/plasma membrane | (126) |
RGS3 | COS-7 | Ectopic | Heat shock, proteotoxic stress | Nucleus/nucleoli | (22) |
RGS4 | S. cerevisiae | Ectopic | NH2-terminal 33 residues | Targets to plasma membrane | (10, 160) |
RGS4 | HEK293 | Ectopic | Expression of active Gαi | Cytoplasm/plasma membrane | (44) |
RGS4 | Rat astrocyte | Endogenous | Atrial natriuretic peptide | Cytoplasm/plasma membrane | (126) |
RGS4 | HEK293 | Ectopic | Expression of Gαi2, M2R | Cytoplasm/plasma membrane | (141) |
RGS4 | PC3-AR | Ectopic | Melatonin, 8-bromo-cGMP | Cytoplasm/nucleus | (133) |
RGS4 | HEK293 | Ectopic | Expression of 14-3-3ε | Cytoplasmic retention | (1) |
RGS4 | SMC | Endogenous | Phosphorylation by PKA or PKG | Cytoplasm/plasma membrane | (79) |
RGS6 | COS-7 | Ectopic | Heat shock, proteotoxic stress | Nucleus/nucleoli | (22) |
RGS6 | HEK293 | Ectopic | Expression of R7BP | Cytoplasm/plasma membrane | (41) |
RGS7 | HEK-293 | Ectopic | Gαo expression, palmitoylation | Cytoplasm/plasma membrane | (165) |
RGS7 | HEK293 | Ectopic | Expression of R7BP | Cytoplasm/plasma membrane | (41) |
RGS8 | DDT1MF2 | Ectopic | Expression of active Gαo, Gαi | Nucleus/plasma membrane | (106, 144) |
RGS9-1 | Sf9 cells | Ectopic | Expression of R9AP | Cytoplasm/plasma membrane | (74) |
RGS9-1 | HEK293 | Ectopic | Expression of R7BP | Cytoplasm/plasma membrane | (41) |
RGS9-2 | HEK293 | Ectopic | Expression of R7BP | Nucleus/plasma membrane | (41) |
RGS10 | HEK293 | Ectopic | Phosphorylation by PKA (Ser168) | Plasma membrane/nucleus and cytoplasm | (17) |
RGS10 | PC3-AR | Ectopic | Melatnonin, 8-bromo-cGMP | Nucleus/cytoplasm | (133) |
RGS11 | HEK293 | Ectopic | Expression of R7BP | Cytoplasm/plasma membrane | (41) |
RGS13 | HeLa | Ectopic | Expression of active Gαi | Cytoplasm/plasma membrane | (151) |
RGS13 | HeLa | Ectopic | Active Gαs expression | Cytoplasm/nucleus | (151) |
NIH3T3 | Ectopic | cAMP, PKA expression | Cytoplasm/nucleus | (191) | |
RGS13 | COS-7 | Ectopic | Heat shock, proteotoxic stress | Nucleus/nucleoli | (22) |
RGS14 | HeLa | Ectopic | Mild heat stress | Cytoplasm/promyelocity leukemia bodies | (30) |
RGS14 | HeLa | Ectopic | Expression of Gαi1/3 | Nucleus and cytoplasm/plasma membrane | (152) |
RGS16 | Yeast | Ectopic | NH2-terminal alpha-helix | Targets to plasma membrane | (23) |
COS-7 | Ectopic | Palmitoylation | Targets to lipid rafts | (69) | |
RGS20 | COS-7 | Ectopic | Heat shock, proteotoxic stress | Nucleus/nucleoli | (22) |
GAIP | COS-7 | Ectopic | Palmitoylation | Cytoplasm/plasma membrane | (33) |
p115RhoGEF | HEK293 | Endogenous | U46619 | Cytoplasm/plasma membrane | (14) |
p115RhoGEF | MDCK, HeLa, U2-OS | Ectopic | Activation of G12/13 by S1P2 | Cytoplasm /plasma membrane | (107) |
p115RhoGEF | PC-12 | Ectopic | Rho inhibition (C3) | Plasma membrane/cytoplasm | (13) |
Data are grouped for each RGS protein in chronological order of publications.
Table 3.
Interaction of RGS proteins with GPCRs
RGS | In vitro/In Cells | Ectopic/Endogenous | Receptor/Site | Ref. No. |
---|---|---|---|---|
RGS2 (NH2 terminus) | In vitro, CHO | Ectopic | M1/3/5 muscarinic receptors (i3 loop) | (11) |
RGS2 | In vitro, HEK293 | Ectopic | α-Adrenergic receptor (i3 loop, spinophilin) | (183) |
RGS2 | In vitro, CHO | Ectopic | α1Aadrenergic receptor (i3 loop: K219, S220, R238) | (61) |
RGS2 | In vitro, CHO | Ectopic | Sphingosine 1-phosphate receptor 1 | (91) |
RGS2 (NH2 terminus: K62, K63, Q67) | In vitro | Ectopic | Cholecystokinin receptor-2 (COOH terminus: P-S434, P-T439) | (94) |
RGS4 | In vitro, CHO | Ectopic | M1/5 muscarinic receptors (i3 loop) | (11) |
RGS4 | In vitro | Ectopic | μ/δ-Opioid receptors (i3 loop, COOH terminus) | (54) |
RGS4 | Rat striatum | Endogenous | Metabotropic glutamate receptor subtype 5 | (148) |
RGS4 (NH2 terminus) | In vitro, HEK293 | Ectopic | μ/δ-Opioid receptors (i4 loop) | (97) |
RGS7/Gβ5 (DEP domain) | In vitro | Ectopic | M3 muscarinic acetylcholine receptor (i3 loop, COOH terminus) | (146) |
RGS8 (NH2 terminus, K8, K9) | In vitro, HEK293T | Ectopic | M1/M3 muscarinic acetylcholine receptors (i3 loop) | (83) |
RGS8 (NH2 terminus) | In vitro, HEK293T | Ectopic | Melanin-concentrating hormone receptor 1 | (109) |
RGS9-2/ Gβ5 | PC12 | Ectopic, endogenous | μ-Opioid receptor | (130) |
RGS9-2 (DEP domain) | PC12 | Ectopic | D2 dopamine receptors | (92) |
LARG (PDZ domain) | HEK293 | Ectopic | LPA receptors 1 and 2 (COOH teminus) | (195) |
PDZ-RhoGEF (PDZ domain) | HEK293 | Ectopic | LPA receptor 1 and 2 | (195) |
Data are grouped for each RGS protein in chronological order of publications. The regions/residues and intermediate molecules providing the interaction are in parentheses.
Table 4.
Regulation of RGS proteins by covalent modification
RGS | Cell/Tissue | Endogenous/Ectopic | Covalent Modification | Site | Effect of Modification | Ref. No. |
---|---|---|---|---|---|---|
SstII | Yeast | Endogenous, ectopic | Phosphorylation | Ser539 | Slows degradation of SstII | (52) |
RGS2 | HEK293T | Ectopic | Phosphorylation | P-tyrosine | Undetermined | (35) |
RGS2 | COS-7 | Ectopic | Phosphorylation | Inhibits GAP activity | (32) | |
RGS2 | VSMC, Rat1 fibroblasts | Endogenous | Phosphorylation | Ser46 | Promotes membrane localization and GAP activity | (119, 167) |
Ser64 | ||||||
RGS2 | In vitro | Ectopic | Palmitoylation | Cys116- | Inhibits GAP activity | (114) |
Cys106- | Promotes GAP activity | |||||
Cys199- | Promotes GAP activity | |||||
RGS3 | Astrocytes | Endogenous | Phosphorylation | Membrane localization | (126) | |
Enhances RGS3 binding to Gαq and Gαi | ||||||
RGS3 | GGH(3) | Ectopic | Palmitoylation | Undetermined | (19) | |
RGS3 | CHO | Ectopic | Phosphorylation | Ser-264 | Provides interaction with 14-3-3 | (115) |
RGS4 | Astrocytes | Endogenous, ectopic | Phosphorylation | Membrane localization | (126) | |
Enhances RGS4 binding to Gαq and Gαi | ||||||
RGS4 | HEK293T | Ectopic | Phosphorylation | P-tyrosine | Undetermined | (35) |
RGS4 | Smooth muscle cells | Endogenous, ectopic | Phosphorylation | Ser52 | Membrane localization | (79) |
Increases Gαq binding | ||||||
RGS4 | Cardiac myocytes | Endogenous | Phosphorylation | Increases interaction with Gαq | (171) | |
RGS4 | S. cerevisiae | Ectopic | Palmitoylation | Cys2, Cys12 | No effect on localization or activity | (160) |
RGS4 | Sf9 | Ectopic | Palmitoylation | Cys2/12- | Promotes Cys95 palmitoylation | (177) |
inhibits or stimulates GAP activity (dependent on assay and Cys95 palmitoylation) | ||||||
Cys95- | Inhibits GAP activity | |||||
RGS4 | HEK293 | Ectopic | Palmitoylation | Cys95 | Promotes GAP activity | (120) |
RGS4 | MDA-MB-231 | Ectopic | Palmitoylation | Cys2 | Stabilizes RGS5 protein | (180) |
HEK293 | Blocks α-adrenergic Ca2+ signaling | |||||
RGS4 | Embryonic fibroblasts | Ectopic | Arginylation | Cys2 | Promotes degradation | (96) |
RGS4 | ATE1−/− mice, 3T3 cells | Endogenous, ectopic | Arginylation | NH2 terminus | Promotes degradation | (75) |
RGS4 | SH-SY5Y | Ectopic | Ubiquitination | Promotes degradation | (181) | |
RGS5 | HEK293T | Ectopic | Phosphorylation | Ser166 | Inhibits GAP activity | (110) |
Decreased Gα binding | ||||||
RGS5 | Embryonic fibroblasts | Ectopic | Arginylation | Cys2 | Promotes degradation | (96) |
RGS5 | ATE1−/− mice, 3T3 cells | Endogenous, ectopic | Arginylation | NH2 terminus | Promotes degradation | (75) |
RGS7 | Mouse brain, HEK293T cells | Endogenous, ectopic | Phosphorylation | Ser241, Thr245, Thr247 | Promotes protein stability | (7) |
RGS7 | Mouse brain, HEK293T cells | Endogenous, ectopic | Phosphorylation | Ser434 | Decreases GAP activity by binding to 14-3-3 | (9) |
RGS7 | Sf9 | Ectopic | Palmitoylation | Membrane-bound RGS7 is palmitoylated | (139) | |
RGS7 | HEK293 | Ectopic | Palmitoylation | Cys133 | Required for Gαo-mediated translocation to plasma membrane | (165) |
RGS9-1 | Outer rod, Sf9 cells, mouse retina | Endogenous, ectopic | Phosphorylation | Ser475 | Phosphorylation is regulated by light | (73) |
RGS9-1 | Photoreceptor outer segments | Endogenous, ectopic | Phosphorylation | Ser427Ser428 | Inhibits GAP activity | (4) |
RGS9-1 | Mouse retina, Sf9 | Endogenous, ectopic | Phosphorylation | Ser475 | Decreased affinity for R9AP | (156, 186) |
RGS9-2 | Periaqueductal gray matter membranes | Endogenous | Phosphorylation | Ser | Promotes 14-3-3 interaction | (53) |
RGS10 | HEK293 | Ectopic | Phosphorylation | Ser168 | Nuclear localization, sequestration from plasma membrane | (17) |
RGS10 | Platelets | Endogenous | Phosphorylation | (51) | ||
RGS10 | Sf9 | Ectopic | Palmitoylation | Cys66 | Inhibits or stimulates GAP activity (assay dependent) | (177) |
RGS10 | GGH(3) | Ectopic | Palmitoylation | Cys60 | Required for inhibition of GnRH signaling | (19) |
RGS13 | HEK293T | Ectopic | Phosphorylation | Thr41 | Inhibits degradation | (194) |
RGS14 | B35 | Ectopic | Phosphorylation | Ser258, Thr494 | Enhances GDI activity | (70) |
RGS16 | HEK293T | Ectopic | Phosphorylation | Ser53 | Inhibits GAP activity | (24) |
Ser194 | ||||||
RGS16 | HEK293T | Ectopic | Phosphorylation (EGFR) | Tyr168>Tyr177 | Enhances GAP activity | (35) |
RGS16 | MCF-7, CHO-K1 | Endogenous, ectopic | Phosphorylation (Src) | Tyr177>Tyr168 | Enhances protein stability | (36) |
RGS16 | HEK293T | Ectopic | Palmitoylation | Cys2Cys12 | Required for inhibition of Gi and Gq signaling | (45) |
RGS16 | HEK293 | Ectopic | Palmitoylation | Cys98 | Required for GAP activity | (120) |
RGS16 | HEK293/COS-7 | Ectopic | Palmitoylation | Cys2Cys12Cys98 | Required for inhibition of Gi/Gq signaling; lipid raft localization | (69) |
RGS16 | ATE1−/− mice, 3T3 cells | Endogenous, ectopic | Arginylation | NH2 terminus | Promotes degradation | (75) |
RGS18 | Platelets | Endogenous | Phosphorylation | Ser49 | (51) | |
RGS19/GAIP | HT-29 | Endogenous, ectopic | Phosphorylation | Ser151 | Stimulates GAP activity | (117) |
Mediates autophagic pathway | ||||||
RGS19/GAIP | AtT-20, HEK293T | Ectopic | Phosphorylation | Ser24 | (49) | |
Rat liver | Endogenous | |||||
RGS19/GAIP | COS7 | Ectopic | Palmitoylation | NH2 terminus | Membrane-anchored GAIP is palmitoylated | (33) |
RGS20/RGSZ | COS-7 | Ectopic | Ubiquitination | Promotes degradation | (121) | |
RGS20/RGSZ | Periaqueductal gray matter | Endogenous | Sumoylation | Promotes Gαi and Gαz interaction | (134) | |
PDZ-RhoGEF | HEK293T | Ectopic | Phosphorylation | COOH terminus | Activation of Rho | (27) |
LARG | HEK293T | Ectopic | Phosphorylation | Tyr | Promotes Rho activation by Gα12 | (163) |
Data are grouped for each RGS protein in the following order: phosphorylation, palmitoylation, and other modification.
GAP, GTPase-activating proteins; GnRH, gonadotropin-releasing hormone; GDI, guanine nucleotide dissociation inhibitor; GAIP, G alpha-interacting protein.
Regulated Expression of RGS Proteins
Table 1 summarizes the current data on the regulated expression of RGS proteins in response to numerous stimuli in various cell culture and animal models. Altogether, a few trends can be noted as described below.
Regulation of RGS expression by G-protein-coupled receptor signaling.
Early studies (37, 39) have demonstrated that the expression of yeast SST2, later recognized as a member of the RGS family, is upregulated in response to G-protein-mediated pheromone signaling. Subsequently, the expression of several RGS proteins has been shown to be increased upon stimulation of various cells with Gq/Gi-coupled agonists, including carbachol (RGS2), angiotensin II (RGS2 and RGS4), ATP (RGS2), phenylephrine (RGS2), sphingosine-1-phosphate (RGS2, RGS16), opioids (RGS4), and endothelin-1 (RGS16; see Table 1). The importance of Gq/Gi signaling in mediating agonist-induced expression of RGS proteins was demonstrated in many of these studies, potentially suggesting that stimulation of RGS expression by a given agonist may serve as a feedback mechanism for the regulation of its own signaling, as well as cross-desensitization of other Gq/Gi-coupled receptor signaling. For example, knockdown of RGS2, the expression of which is transiently (1–12 h) induced by angiotensin II in cultured cardiac fibroblasts, augmented angiotensin II-induced IP3 production, collagen synthesis, and proliferation (200). Interestingly, angiotensin II infusion in vivo resulted in a biphasic effect, with the initial induction of RGS2 expression in cardiac fibroblasts at day 1, followed by a decline below the control levels at days 3–14 (200). This may suggest that the initial RGS2 upregulation may serve to regulate angiotensin II signaling, whereas a subsequent RGS2 downregulation may contribute to the pathogenesis of cardiac hypertrophy and fibrosis; the latter is supported by other models (discussed in Regulation of RGS expression in animal models). Likewise, opioid receptor agonists induced a rapid but transient (1–8 h) induction of RGS4 mRNA in PC12-μOR/κOR transfected cells (112), whereas prolonged stimulation (24 h) of SH-SY5Y cells with μ- or δ- opioid receptor (MOR and DOR, respectively) agonists resulted in Gi-mediated ubiquitination and degradation of the RGS4 protein (181). Importantly, downregulation of RGS4 expression by MOR agonist (DAMGO) resulted in cross-sensitization of Gi/o-coupled DOR and the Gq-coupled M3 muscarinic receptor (M3R) signaling without affecting α2-adrenergic receptor or bradykinin BK2 receptor signaling (indicative of receptor-specific effects of RGS4; Ref. 181).
RGS2 expression is also stimulated in various cells by Gs-coupled agonists and cAMP-elevating drugs, including prostaglandin E2 (6), parathyroid hormone (89, 108, 142, 169, 176), isoproterenol (62, 116), cholera toxin (89, 164), and forskolin (28, 62, 89, 123, 127, 142) (see Table 1). Together, these results may suggest that stimulation of RGS2 expression by a Gs-coupled agonist may provide cross-desensitization of Gq/Gi-coupled receptor signaling. However, only a few studies have directly assessed the possibility of cross-regulation between Gs and Gq/Gi by using knockdown/knockout approaches. In this context, studies by Roy et al. (142) demonstrated that forskolin pretreatment leads to desensitization of endothelin-1 and ATP signaling in wild-type (WT) but not in RGS2−/− osteoblasts. RGS2 was shown to interact with various isoforms of adenylyl cyclases and to inhibit their activity (140, 141, 145, 153). Ectopic RGS2 overexpression has led to inhibition of cAMP production induced by odorant in olfactory neurons (153) or by isoproterenol in HEK293/β2AR cells (140, 145) and in rat neonatal ventricular cardiomyocytes (116). This may suggest that stimulation of RGS2 expression by a Gs-coupled agonist may also lead to cross-desensitization of other Gs-coupled receptors through inhibition of adenylyl cyclase, which has yet to be demonstrated. Of note, other investigators (62) showed that RGS2 overexpression does not affect isoproterenol-induced cAMP production in rat adult ventricular myocytes. Given that RGS2 may have variable effects on different adenylyl cyclase isoforms, this effect of RGS2 may be cell specific, depending on which adenylyl cyclase isoforms are expressed in a given cell, and may differ even between adult (62) and neonatal (116) cardiomyocytes.
In contrast to RGS2, the expression of RGS3 (28), RGS4 (28, 112), RGS5 (198), and RGS16 (6) is downregulated by Gs/cAMP-coupled agonists in various cell types (see Table 1). Of interest is the regulation of RGS13 expression in mast cells, in which RGS13 mRNA is decreased by cAMP, whereas RGS13 protein is stabilized by PKA-dependent phosphorylation (194), suggesting that the balance between these two processes may determine RGS13 expression levels. cAMP is known to regulate the acute Gq/Gi signaling (i.e., Ca2+ release from intracellular stores) at least in part through 1) PKA-dependent phosphorylation and inhibition of phospholipase-Cβ2 that produces IP3 (103) or of IP3 receptors (16); and 2) through activation of Ca2+ reuptake by sarcoplasmic reticulum Ca2+-ATPase (SERCA2) via PKA-dependent phosphorylation of the SERCA2 inhibitor phospholamban and subsequent release of phospholamban from SERCA2 (105). However, downregulation of RGS3/4/5/16 by Gs/cAMP-coupled agonists may suggest that Gq/Gi signaling could be also enhanced by cAMP over time. One study (198) addressed this functionally in airway smooth muscle cells and showed that downregulation of Gq/Gi-specific RGS5 mRNA and protein by long-term treatment with isoproterenol paralleled increased Ca2+ responses to bradykinin, thrombin, and histamine. Furthermore, RGS5 knockdown produced a similar effect in this study. However, as a given cell may express both RGS2 (that is induced by cAMP) and RGS3/4/5/16 (that are downregulated by cAMP), the overall effect of long-term treatment with Gs/cAMP-coupled agonists on Gq/Gi signaling may be cell specific, depending on the expression profile of RGS proteins.
Regulation of RGS expression by LPS and inflammatory cytokines.
Bacterial LPS and proinflammatory cytokines are important mediators of the immune response and inflammation. Not surprisingly, most of these studies were performed on immune cells, although some explored smooth muscle cells and cardiac myocytes, which are also targets of inflammation. In immune cells, LPS variably affected RGS expression, depending on the RGS protein and the cell type (see Table 1). LPS was shown to upregulate RGS1 in dendritic cells and macrophages (132, 150), RGS16 in pre-B and dendritic cells (100, 150), and RGS20/RGSZ1 in dendritic cells (150). Interestingly, in macrophages, the initial rapid upregulation of RGS1 mRNA by LPS was followed by a decrease in its expression below control levels (95, 132) . On the other hand, LPS decreased RGS2 mRNA in macrophages (95, 132), as well as RGS14 and RGS18 mRNAs in dendritic cells (150). Functionally, one study (95) using RGS2 small interfering siRNA and RGS2 overexpression approaches has demonstrated that downregulation of RGS2 mRNA by LPS in macrophages may mediate LPS-induced foam cell formation, although it remains to be determined whether this function of RGS2 relates to the regulation of G-protein signaling. The expression of some RGS proteins in various immune cells is induced by inflammatory cytokines, including interferon-β [RGS1 (173), RGS2 (55)], interleukin-1 [RGS16 (100)], interleukin-2 [RGS16 (6)], interleukin-17 [RGS13, RGS16 (189)], and TNFα [RGS16 (50)]. Given the established role of Gi signaling in migration of immune cells towards various chemokines, one would anticipate that the regulated expression of RGS proteins by LPS and inflammatory cytokines would affect this process, although this remains to be investigated.
In nonimmune cells, IL-1β has been shown to stimulate a rapid and sustained expression of RGS4 in colonic smooth muscle cells and in ex vivo-treated colonic muscle strips, which coincided with inhibition of acetylcholine-stimulated phospholipase C (PLC) activation and smooth muscle contraction by IL-1β (76–78); these effects were reversed by RGS4 knockdown (78). LPS, IL-1, and TNF-α increased the expression of RGS4 and RGS16 in cardiac myocytes, which corresponded with the inhibition of endothelin-1-induced PLC activation (124, 125). Finally, LPS stimulated expression of RGS16 in vascular smooth muscle cells and in ex vivo-treated aortic rings, which paralleled the inhibition of phenylephrine- and angiotensin II-induced contraction (65). Overall, these data suggest that RGS4/16 upregulation may diminish Gq/Gi signaling and contraction in LPS/TNFα-pretreated vascular smooth muscle cells and cardiac myocytes. Although these RGS proteins may contribute to the vasodilation and acute heart failure observed during sepsis, the causative role of RGS4 and RGS16 in the effects of LPS described in the latter studies (65, 124, 125) remains to be determined. Interestingly, while LPS-induced RGS16 expression was associated with inhibition of phenylephrine- or angiotensin II-induced contraction of ex vivo aortic rings, LPS pretreatment did not affect contraction in response to endothelin-1 and even enhanced serotonin-induced contraction (65). The former observation (related to endothelin-1) may represent a receptor-selective function of RGS proteins as reviewed previously (2, 113), although the interaction of RGS16 with G-protein-coupled receptors (GPCRs) has not been investigated (see Table 3). The latter observation (related to serotonin) may represent the regulation of smooth muscle contraction through a transporter-mediated serotonin uptake independent of G-protein signaling (102).
RGS expression under cell stress conditions.
Mammalian cells experience and variably respond to stress under various injurious conditions (hyperoxia, hypoxia/ischemia, mechanical stress and drug treatment). Several studies have examined how inducers of cell stress regulate RGS expression. RGS2 was shown to be upregulated in astrocytoma cells upon oxidative stress and heat shock or ischemia (47, 203) and in neuroblastoma SH-SY5Y cells upon heat shock and cell cycle arrest (157, 158). In the study of Endale et al. (47), overexpression of RGS2 mimicked ischemia-induced apoptosis. Similarly, hypoxia increased RGS5 expression in endothelial cells in a HIF-1α-dependent manner, which mediated hypoxia-induced apoptosis, as shown by utilizing RGS5 siRNA (88). Mechanical stress (compressive force) of periodontal ligament cells resulted in a cAMP-dependent expression of RGS2, which limited further cAMP production as determined using antisense S-oligonucleotides to RGS2 (147). Finally, treatment of ovarian cancer cells with therapeutic cytotoxic drugs (cisplatin, vincristine, and docetaxel) led to upregulation of RGS10 and RGS17, which mediated cell toxicity as determined by siRNA approach (72). These data suggest that increased expression of RGS proteins can mediate the detrimental effects of cell stress under certain conditions. Future studies should examine if and how such regulations of RGS expression are related to the control of G-protein signaling and function in the context of cell stress.
Regulation of RGS expression in animal models.
A large body of data on the regulation of RGS expression in the brain as related to neurological diseases and drug abuse was reviewed elsewhere (174, 175). This chapter focuses on the regulation of RGS expression in animal models as related to cardiovascular diseases, including hypertension, atherosclerosis, heart failure, and sepsis. Some of these topics are also reviewed elsewhere (155, 199).
It was initially reported that levels of RGS4 are increased in the pulmonary artery-banded mouse model of cardiac hypertrophy (201). In the follow-up study, this group showed that unlike WT mice, transgenic mice with cardiac-specific overexpression of RGS4 did not respond to phenylephrine by MAP kinase activation in the heart and exhibited reduced cardiac hypertrophy, with no induction of the “fetal” gene program, 7 days post-transverse aortic constriction (135). Interestingly, RGS4-transgenic mice had a significantly reduced survival 1–2 days post-transverse aortic constriction, suggesting that RGS4 also controls cardiac responses to acute pressure overload (135). Furthermore, when RGS4-transgenic mice were crossed with mice overexpressing Gαq in the heart (a model for cardiac hypertrophy), the Gαq/RGS4 transgenic mice had reduced cardiac hypertrophy and normalized left ventricular function, which was accompanied with reduced activity of PKCε and decreased expression of some hypertrophic genes (atrial natriuretic factor, skeletal muscle α-actin), compared with Gαq-transgenic mice (136). In contrast to the above studies, the expression of RGS3 and RGS4 mRNA and protein was dramatically decreased in the heart during congestive heart failure in 16- to 20-mo-old SHHF rats (201). However, it would be interesting to examine the expression of these proteins at the earlier age (3–5 mo), when elevated blood pressure and cardiac hypertrophy are already developed before heart failure.
In contrast to RGS4, the expression of RGS2 was markedly decreased in three models of cardiac hypertrophy: transgenic cardiac-specific expression of an activated Gαq mutant (202), aortic banding-induced pressure overload (202), and prolonged infusion of angiotensin II (200). Importantly, knockdown of RGS2 increased Gq signaling and hypertrophy of cultured cardiac myocytes and fibroblasts in these studies. The functional significance of RGS2 downregulation in mediating cardiac hypertrophy was further demonstrated using RGS2-knockout mice, which exhibit increased myocardial Gq signaling and cardiac hypertrophy, compared with WT mice (166). Furthermore, in the dyssynchronous heart failure canine model, cardiac resynchronization therapy resulted in increased cardiac expression of RGS2 and RGS3, which was associated with improved heart function and with suppressed Gαi signaling, yielding increased Gαs-biased cAMP responses to β2-adrenergic stimulation in isolated cardiac resynchronization therapy myocytes. (20, 21). On the other hand, RGS4 and RGS16 mRNA and protein are increased in the heart in the LPS-induced model of sepsis known to be associated with acute heart failure, which coincided with decreased PLC activity in the heart (124).
In the abdominal or thoracic aorta banding models of hypertension and vascular hypertrophy, biphasic regulation of RGS2, RGS4, and RGS5 was observed in the thoracic and abdominal aortas, respectively. Banding resulted in a dramatic decline of their mRNAs on day 2, followed by a several fold increase over control aortas on day 28 postbanding (182), which is in contrast to a sustained downregulation of cardiac RGS2 in the thoracic aorta-banding model (202). This dynamic expression of RGS2/4/5 in the aorta correlated with increased Gq/Gi agonist-induced contraction of aortic rings on day 2 and decreased contraction on day 28 ex vivo (182). However, this study has not investigated how increased RGS2/4/5 may have contributed to the development of hypertension and vascular hypertrophy occurring during days 14–28. Given that RGS2−/− mice develop enhanced vascular contraction and hypertension (167), it is possible that at least RGS2 upregulation plays a compensatory role in the former study. Additionally, RGS5 mRNA was sharply downregulated in atherosclerotic plaques in the atherogenic diet-induced model of atherosclerosis (99). In agreement with this study, the reduction of atherosclerosis by peroxisome proliferator-activated receptor-δ agonists in apoE−/− mice was accompanied by an increase of RGS4 and RGS5 mRNAs in atherosclerotic plaques (5). These data are consistent with the proliferative phenotype of VSMC in plaques, which could be regulated by RGS4 and RGS5, although this notion is yet to be investigated.
Together, these data demonstrate that the expression of a given RGS protein is variably regulated in different models of cardiovascular disease, and the expression of more than one RGS protein may be altered in a given model. However, the specific contribution of each RGS protein to the development of disease in these models remains largely uncertain and requires further investigation.
Regulation of Intracellular Localization of RGS Proteins
To perform the canonical GAP function, RGS proteins must localize to the plasma membrane (where GPCRs and G proteins are localized). Several mechanisms for the regulation of RGS intracellular localization are summarized in Table 2 and are described below.
RGS2, RGS4, and RGS16 are targeted to the membrane by specific regions NH2-terminal to their RGS domains (10, 23, 58, 160). The R44H mutation of RGS2 within the amino terminal amphipathic α-helix (responsible for plasma membrane-targeting) leads to less efficient membrane binding and inhibition of Gq signaling (59). Interestingly, this R44H mutation of RGS2 is associated with hypertension in the Japanese population (197). Palmitoylation of RGS proteins is another mechanism for membrane targeting, which is reviewed below (see Regulation of RGS Proteins by Covalent Modification).
RGS proteins can be also recruited to the membrane by activated (GTP-bound) Gα subunits that have a high binding affinity for RGS proteins. Many studies used overexpressed constitutively active (GTPase-deficient) Gα subunits to show translocation of RGS proteins from the cytosol to the membrane. Expression of constitutively active Gαq/11 and/or Gαi/o has led to membrane localization of RGS2 (31, 68), RGS3 (46), RGS4 (44), and RGS8 (106, 144). Given the transient nature of the RGS/Gα-GTP interaction, it is challenging to demonstrate GPCR agonist-induced RGS translocation to the membrane in intact cells. Nevertheless, membrane translocation of RGS3 in response to endothelin-1 (46), of RGS4 and RGS9–2 in response to μ/δ-opioid receptor agonists (97, 130), of p115RhoGEF in response to sphingosine-1-phosphate (S1P) or the thromboxane A(2) receptor agonist (13, 107), or of RGS3 and RGS4 in response to a non-G-protein coupled agonist, atrial natriuretic peptide (126), have been reported. The latter data suggest that the Gα-GTP/RGS interaction is not the only mechanism of RGS recruitment to the membrane and downstream signaling could be also involved. Indeed, roles for Ca2+ mobilization [RGS3 (46, 172)] and RhoA and PI3K activation [p115RhoGEF (13, 14)], or cGMP production [RGS2 (119, 133, 167), RGS3, RGS4 (126), and RGS10 (133)] have been suggested in driving membrane translocation of RGS proteins. Interestingly, some studies have shown that the expression of WT Gα subunits also promote membrane localization of RGS proteins, which includes the recruitment of RGS2 and RGS4 to the membrane by expression of WT Gαq and Gαs (141) and of RGS14 by expression of WT Gαi1 and Gαi3 (152). However, it is possible that overexpression of WT Gα subunits and RGS proteins may enable a low affinity interaction, which could be sufficient for the recruitment of RGS proteins to the membrane (our unpublished observations). Finally, membrane targeting of R7 family members (RGS6/7/9/11) is mediated through their interaction with membrane anchors, RGS7-binding protein (R7BP) and/or RGS9-anchoring protein (R9AP), which facilitate the regulatory function of R7 proteins, which is reviewed elsewhere (85). Even though the expression (and function) of R7 members of RGS family is largely restricted to the brain and retina, it was also shown that RGS6 is expressed in cardiac myocytes (40) and modulates parasympathetic regulation of heart rate through the regulation of muscarinic receptor-activated potassium channels (129, 196). Hence, modulation of RGS6 intracellular localization in cardiac myocytes may be of importance for heart function and disease, and should be evaluated in the future.
Several RGS proteins were shown to interact with various GPCRs (through regions outside of the RGS domain) by binding the third intracellular (i3) loop or the COOH terminus of GPCRs, either directly or through intermediate proteins such as spinophilin, which has been previously reviewed (2, 113). Table 3 provides updated information on RGS-GPCR interactions and the regions involved. While this interaction could provide receptor-specific selectivity of RGS proteins in regulation of signaling, it may also serve to recruit RGS proteins to the membrane. Indeed, RGS2 plasma membrane localization has been shown to be induced by expression of the β2-adrenergic receptor (141), AT1A angiotensin II receptor (141), and the M3 muscarinic receptor (31), whereas RGS4 membrane localization was promoted by expression of the M2 muscarinic receptor (141) (Table 2). The COOH terminus and the i3 loop of GPCRs are known to be phosphorylated by GRK and PKA/PKC respectively, which mediates GPCR desensitization and internalization (170). Therefore, it would be interesting to examine if and how these processes are affected by RGS interaction and how the latter is affected by GPCR phosphorylation.
Finally, many RGS proteins were shown to undergo regulated localization to intracellular compartments other than the plasma membrane, including the nucleus [reviewed by Sethakorn et al. (149)]. While nuclear localization of RGS proteins may suggest functions unrelated to regulation of G-protein signaling, it may also serve to sequester RGS proteins from the cytoplasm, thus regulating their canonical function. Interestingly, some studies suggest that nuclear/cytosolic localization is a regulated process. For example, melatonin receptor activation led to nuclear accumulation of RGS4 but induced translocation of RGS2 and RGS10 from the nucleus to the cytoplasm (133), whereas PKA-dependent phosphorylation of RGS10 induced its nuclear translocation and thus nullified RGS10 activity at the plasma membrane (17).
Regulation of RGS Proteins Through Covalent Modifications
Table 4 summarizes the current data on the effect of posttranslational modifications on the activity of RGS proteins. Prominent covalent modifications include phosphorylation and palmitoylation; however, ubiquitination, sumoylation, and arginylation have also been reported. These modifications have a prominent role in modulating RGS protein function and also represent mechanisms of G-protein signaling regulation by both G-protein and non-G-protein pathways.
Regulation of RGS proteins by phosphorylation.
Phosphorylation of many RGS proteins has been reported, spanning across all RGS subfamilies. In several cases, phosphorylation of RGS proteins [RGS2 (32), RGS5 (110), RGS9–1 (4), and RGS16 (24)] resulted in inhibition of GAP activity, as demonstrated by in vitro GTPase activity assays and measurements of downstream signaling effectors. Alternatively, phosphorylation can indirectly inhibit the GAP activity of RGS proteins. For example, phosphorylation of RGS3 outside of the RGS domain (115, 184) and of RGS7 within the RGS domain (9) leads to their interaction with the phospho-serine-binding protein 14–3-3, resulting in inhibition of RGS activity. Structural studies revealed that 14–3-3 binding induces conformational changes in both the NH2-terminal region and the COOH-terminal RGS domain of phosphorylated RGS3 (131), while functional studies demonstrated that 14–3-3 binding to RGS3 inhibits its interaction with G proteins (115). Similarly, RGS4, RGS5, and RGS16 were reported to interact with 14–3-3, which led to inhibition of RGS4 and RGS16 GAP activity through competition between 14–3-3 and the RGS/G-protein interaction (1). However, RGS5 GAP function was not significantly affected, which may indicate that the 14–3-3 interaction as a mechanism of regulation may be specific to certain RGS proteins (1).
Conversely, phosphorylation of RGS4 in astrocytes (126), smooth muscle cells (79), and cardiac myocytes (171) appears to stimulate their GAP activity through increased interaction with Gαq and/or membrane localization. In smooth muscle cells, inhibition of PLC activity by cAMP was mediated by PKA-dependent phosphorylation of GRK2 [containing a Gq-specific RGS-like domain(18)] and of RGS4 (79). Overall, these results suggest that the effects of phosphorylation on RGS function may be cell-type specific, may differ depending on whether proteins tested were endogenous or ectopic, or may be dependent on the site of phosphorylation and/or kinase. In support of the latter notion, phosphorylation of RGS2 by PKG (at S46/S64) increased RGS2 GAP activity (167), whereas phosphorylation by PKC (site unknown) inhibited RGS2 GAP activity (32). Likewise, phosphorylation of RGS16 at S53/S194 inhibits GAP activity (24), whereas phosphorylation at Y168/Y177 by the EGF receptor or by Src kinases augmented RGS16 inhibition of Gi signaling, either through enhanced GAP activity (35) or through RGS16 protein stabilization (36). Of note, RGS2 and RGS4 can be also phosphorylated on tyrosine residues (35), although the specific sites and the significance of phosphorylation remain to be determined.
As described above for RGS16 (36), phosphorylation can promote the stability of RGS proteins. It was first demonstrated that pheromone-induced phosphorylation of SST2 at S539 by MAP kinase stabilized the SST2 protein in yeast (52). Furthermore, Xie et al. (194) reported that while cAMP significantly reduced mRNA levels of RGS13, the protein levels remained largely constant due to PKA-mediated phosphorylation and subsequent stabilization of the RGS13 protein, likely through protection from proteasomal degradation (194). Functionally, this promoted RGS13-mediated inhibition of carbachol-induced ERK phosphorylation (194). RGS7 provides another example of phosphorylation-regulated stability. Benzing et al. (7) reported that TNF-α treatment promoted the stability of RGS7, which was mediated by p38MAP kinase-dependent phosphorylation of RGS7 at S241, T245, and T247 sites. These authors also demonstrated that in vivo administration of TNF-α or LPS resulted in increased RGS7 protein expression in mouse brain (7). In addition to promoting long-term protein stability, TNF-α treatment also resulted in short-term dephosphorylation of RGS7 at S434 and concomitant loss of interaction with 14–3-3, which negatively regulates the activity of RGS7 (8). Thus RGS7 exemplifies a protein that can be regulated by the same stimulus through dual, phospho-site-specific, mechanisms. However, it is noteworthy that the major mechanism for stabilization of RGS7 as well of other members of the R7 family (RSG6/9/11) is likely through their interaction with Gβ5, as knockout of Gβ5 results in a drastic reduction or elimination of R7 protein expression without having an effect on mRNA levels (25).
Protein phosphorylation has been also demonstrated to affect localization of RGS proteins. Pedram et al. (126) reported that atrial natriuretic peptide induced PKG-mediated phosphorylation of RGS3 and RGS4, enhancing membrane localization and association with Gαq and Gαi and presumably augmenting Gq/Gi regulation. On the other hand, 14–3-3 interaction with phosphorylated RGS3/4/5/7/16 proteins (1, 9, 115) may also affect their localization through cytoplasmic retention by 14–3-3, as was demonstrated for 14–3-3-bound RGS4 (1). Phosphorylation of RGS10 by PKA induced its translocation to the nucleus, thus preventing its GAP function at the plasma membrane (17). Cyclic AMP/PKA signaling also induced nuclear translocation of RGS13, although indirectly through association of RGS13 with PKA-phosphorylated CREB (191). Interestingly, in the nucleus, RGS13 acts also noncanonically as a repressor of CREB-dependent gene transcription (191).
Regulation of RGS proteins by palmitoylation.
Palmitoylation of cysteines has been repeatedly reported to promote membrane localization, as is the case for several RGS proteins including RGS4 (120, 128, 177, 180), RGS7 (139, 165), RGS16 (45, 69, 120), and GAIP (33). Logically, these palmitoylated RGS proteins would demonstrate increased GAP activity secondary to increased colocalization with their G-protein partners at the membrane. However, data from several groups suggest that the effects of palmitoylation on RGS proteins may be more complex. For example, Ni et al. (114) demonstrated that the specific palmitoylated residues on RGS2 dictate whether the modification inhibits (Cys-116) or promotes (Cys-106, Cys-199) GAP activity, at least in vitro. Palmitoylation of RGS4 within the RGS box at Cys-95 inhibited GAP activity in Sf9 cells (177) but promoted it in HEK293 cells (120). Palmitoylation of RGS4 outside of the RGS box at Cys-2 and Cys-12 promoted palmitoylation at Cys-95 and might also modulate RGS4 GAP activity, although the effect varied dependent on the GAP assay (177). Palmitoylation of RGS4 at Cys-2 also increased RGS4 stability and potentiated the inhibition of α-adrenergic-induced calcium signaling by RGS4 (180). Other studies suggest that specific membrane compartmentalization, compared with general membrane binding, may have a greater effect of RGS function, at least for some RGS proteins. Mutation of cysteine residues crucial for palmitoylation in RGS16 demonstrated that while membrane binding was not affected, localization into lipid rafts was severely abrogated and resulted in decreased GAP activity of RGS16 (45, 69, 120). Palmitoylation of RGS10 could either attenuate or potentiate GAP activity, depending on whether the assay was performed as an in-solution single-turnover assay or in reconstituted phospholipid vesicles measuring steady-state GTP hydrolysis (177). However, in GGH3 cells, expression of a palmitoylation-deficient mutant of RGS10 attenuated its ability to regulate GnRHR-mediated PLC activation (19), which is more consistent with a positive role for palmitoylation on RGS10 GAP activity. In the same study, the authors (19) also reported that palmitoylation of RGS3 was dependent on GnRHR stimulation, although a functional significance was not investigated.
Regulation of RGS proteins by arginylation, ubiquitination, or sumoylation.
Arginylation, the process of conjugating arginine residues to NH2-terminal aspartate, glutamate, and cysteine residues, is a key step in the ubiquitin-dependent proteasomal degradation N-end rule pathway (143). Several RGS proteins were demonstrated to be arginylated, resulting in the degradation of these proteins. Hu et al. (75) reported that the arginyl-transferase ATE1 could modify RGS4/5/6, and expression of these proteins was dramatically increased in ATE1−/− mice. Of note, increased expression of these RGS proteins was not observed in all tissues of the knockout animals, but rather RGS4 and RGS16 appeared to be strongly upregulated in the brain and lung, whereas RGS5 showed robust stabilization in the heart (75). These authors also demonstrated that RGS4 was stabilized under low oxygen conditions, and a concurrent study (96) showed that proteolysis of RGS5 was decreased during hypoxia. The increased levels of RGS proteins in ATE1 knockout mice are postulated to contribute to the massive cardiovascular defects observed in these animals, and indeed ERK activation and induction of GPCR-regulated genes are impaired in the ATE1-deficient embryos (96). Overall these data suggest an intricate interplay of oxygen-sensing mechanisms and regulation of RGS protein levels to finely tune G-protein activity in the heart and possibly in other tissues as well.
Other groups have examined receptor-mediated downregulation of RGS proteins. For example, in neuroblastoma SH-SY5Y cells, μ- and δ-opioid receptor (MOR and DOR) activation led to ubiquitination and proteolysis of RGS4. This degradation was sensitive to pertussis toxin and proteasomal and lysosomal inhibitors. Furthermore, downregulation of RGS4 by a MOR agonist resulted in augmentation of DOR-induced MAP kinase and M3-muscarinic receptor-induced cAMP accumulation but had no effect on α2-adrenergic receptor or bradykinin receptor signaling (181). These results once again exemplify how a GPCR can modulate RGS protein levels to cross-regulate other GPCRs. As another example, constitutively active Gαo induced RGS20 ubiquitination and downregulated RGS20 expression through proteasomal degradation (121). In the same study, activation of the Gαo/i-coupled serotonin receptor led to RGS20 degradation, which resulted in loss of inhibition of Gi signaling and was reversed upon addition of the proteasome inhibitor lactacystin.
Lastly, there is one report (134) on sumoylation of RGS proteins, RGSZ1 and RGSZ2, which are conjugated to SUMO1, 2, and 3 in synaptosomal membranes of the periaqueductal gray matter. The sumoylated Rz proteins can coprecipitate with Gαz and μ-opioid receptors, thus serving as a scaffold for this complex. Administration of morphine before membrane extraction increased the Gαz/i and RGSZ2 interactions, which peaked thirty min posttreatment, corresponding to the maximal point of morphine-induced analgesia (134).
Overall, the literature demonstrates that posttranslational modifications can modulate RGS activity via multiple modes of regulation such as intracellular localization, protein-protein interactions, and protein stability. The data also indicate complexity within these mechanisms; for example, the effect of phosphorylation or palmitoylation often depends on the specific residue that is modified.
Conclusions
This review has summarized the current information on the regulation of RGS proteins through their expression, intracellular localization, and covalent modifications. Overall, the literature has demonstrated that regulation of RGS proteins occurs on multiple levels and offers a complexity that allows for plastic control of G-protein function. However, it is anticipated that more studies will be performed in the following directions, as discussed below.
While multiple studies report the regulated expression of RGS mRNA, it is important to assess RGS expression at the protein level as well (which a few studies have convincingly demonstrated, see Table 1). One study exemplifies the importance of this notion by showing that cAMP signaling has opposite effects on RGS13 mRNA and protein expression (194). However, the availability of specific antibodies against some RGS proteins remains a significant obstacle. Furthermore, while many studies demonstrate stimuli-regulated RGS expression and concomitant changes in G-protein signaling, the specific requirement for individual RGS proteins was largely not examined. Additionally, microRNAs (miR) represent another mechanism of regulated expression that was not discussed in this review, as at this time, only RGS2 (87, 111) and RGS17 (162) genes have been reported to be regulated by miRs.
Many investigations have explored functions through overexpression of RGS genes; however, it is essential to examine if the conclusions translate to the endogenous RGS proteins. While the overexpression of several RGS proteins have proven valuable in characterizing their biochemical properties, this approach cannot ascertain whether RGS proteins function redundantly. Additionally, ectopic expression may prevent accurate interpretation of protein function, for instance, by potentially negating regulatory mechanisms. The function of endogenous RGS proteins can be addressed through studies of RGS knockout mice, which provide the additional advantage of examining in vivo function. Importantly, heterozygotes should be also examined to evaluate the effect of partial downregulation of the RGS protein, which would support or refute the functional significance of partial RGS downregulation observed in a number of cell culture studies. However, given that several RGS proteins may be regulated in a similar manner in the same cell type, and given the functional redundancy of some RGS proteins, the double/triple knockout mice would have to be generated. RNA interference-mediated knockdown can serve as an alternative method to examine the endogenous RGS proteins, and it offers the advantage of circumventing the possibility of compensation mechanisms (i.e., possible upregulation of other RGS family members) in knockout mice. However, the potential off-target effects of siRNA were not considered in many of the discussed studies and should be controlled for by using separate siRNAs against different regions of the gene and by rescue experiments using siRNA-resistant RGS mutants. When possible, highly stringent studies should utilize complementary techniques for understanding the RGS regulation and function. In addition, recent development of pharmacological inhibitors of RGS proteins may provide an alternative useful method to study the role of endogenous RGS proteins (154, 155). However, while the in vitro efficacy and specificity of some molecules are promising, their in situ and in vivo efficiency requires validation.
Finally, we anticipate more studies on RGS regulation and function in animal models and humans as they relate to the pathogenesis of disease. Emerging data indicate that RGS proteins are subjected to regulated expression in various diseases, including cardiovascular pathologies and neurological disorders, and are becoming increasingly appreciated in several types of cancers (155, 199). We expect that RGS dysregulation will be demonstrated in other diseases, and the specific contribution of RGS proteins to the pathogenesis of disease will be investigated, which may point to a given RGS as a potential therapeutic target for a disease treatment (154, 155).
GRANTS
This study was supported by National Institutes of Health Awards R01-GM-85058 (to N. O. Dulin), T32-HL-007237 (to J. Kach), and T32-HD-009007 (to N. Sethakorn); and American Heart Association Fellowships 10PRE4190120 (to J. Kach) and 10PRE2630163 (to N. Sethakorn).
DISCLOSURES
No conflicts of interest, financial or otherwise, are declared by the author(s).
AUTHOR CONTRIBUTIONS
Author contributions: J.K., N.S., and N.O.D. drafted manuscript; J.K., N.S., and N.O.D. edited and revised manuscript; J.K., N.S., and N.O.D. approved final version of manuscript.
REFERENCES
- 1.Abramow-Newerly M, Ming H, Chidiac P. Modulation of subfamily B/R4 RGS protein function by 14–3-3 proteins. Cell Signal 18: 2209–2222, 2006 [DOI] [PubMed] [Google Scholar]
- 2.Abramow-Newerly M, Roy AA, Nunn C, Chidiac P. RGS proteins have a signalling complex: interactions between RGS proteins and GPCRs, effectors, and auxiliary proteins. Cell Signal 18: 579–591, 2006 [DOI] [PubMed] [Google Scholar]
- 3.Appleton CT, James CG, Beier F. Regulator of G-protein signaling (RGS) proteins differentially control chondrocyte differentiation. J Cell Physiol 207: 735–745, 2006 [DOI] [PubMed] [Google Scholar]
- 4.Balasubramanian N, Levay K, Keren-Raifman T, Faurobert E, Slepak VZ. Phosphorylation of the regulator of G protein signaling RGS9–1 by protein kinase A is a potential mechanism of light- and Ca2+-mediated regulation of G protein function in photoreceptors. Biochemistry 40: 12619–12627, 2001 [DOI] [PubMed] [Google Scholar]
- 5.Barish GD, Atkins AR, Downes M, Olson P, Chong LW, Nelson M, Zou Y, Hwang H, Kang H, Curtiss L, Evans RM, Lee CH. PPARdelta regulates multiple proinflammatory pathways to suppress atherosclerosis. Proc Natl Acad Sci USA 105: 4271–4276, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Beadling C, Druey KM, Richter G, Kehrl JH, Smith KA. Regulators of G protein signaling exhibit distinct patterns of gene expression and target G protein specificity in human lymphocytes. J Immunol 162: 2677–2682, 1999 [PubMed] [Google Scholar]
- 7.Benzing T, Brandes R, Sellin L, Schermer B, Lecker S, Walz G, Kim E. Upregulation of RGS7 may contribute to tumor necrosis factor-induced changes in central nervous function. Nat Med 5: 913–918, 1999 [DOI] [PubMed] [Google Scholar]
- 8.Benzing T, Kottgen M, Johnson M, Schermer B, Zentgraf H, Walz G, Kim E. Interaction of 14–3-3 protein with regulator of G protein signaling 7 is dynamically regulated by tumor necrosis factor-alpha. J Biol Chem 277: 32954–32962, 2002 [DOI] [PubMed] [Google Scholar]
- 9.Benzing T, Yaffe MB, Arnould T, Sellin L, Schermer B, Schilling B, Schreiber R, Kunzelmann K, Leparc GG, Kim E, Walz G. 14–3-3 Interacts with regulator of G protein signaling proteins and modulates their activity. J Biol Chem 275: 28167–28172, 2000 [DOI] [PubMed] [Google Scholar]
- 10.Bernstein LS, Grillo AA, Loranger SS, Linder ME. RGS4 binds to membranes through an amphipathic alpha-helix. J Biol Chem 275: 18520–18526, 2000 [DOI] [PubMed] [Google Scholar]
- 11.Bernstein LS, Ramineni S, Hague C, Cladman W, Chidiac P, Levey AI, Hepler JR. RGS2 binds directly and selectively to the M1 muscarinic acetylcholine receptor third intracellular loop to modulate Gq/11alpha signaling. J Biol Chem 279: 21248–21256, 2004 [DOI] [PubMed] [Google Scholar]
- 12.Berthebaud M, Riviere C, Jarrier P, Foudi A, Zhang Y, Compagno D, Galy A, Vainchenker W, Louache F. RGS16 is a negative regulator of SDF-1-CXCR4 signaling in megakaryocytes. Blood 106: 2962–2968, 2005 [DOI] [PubMed] [Google Scholar]
- 13.Bhattacharyya R, Banerjee J, Khalili K, Wedegaertner PB. Differences in Galpha12- and Galpha13-mediated plasma membrane recruitment of p115-RhoGEF. Cell Signal 21: 996–1006, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Bhattacharyya R, Wedegaertner PB. Characterization of G alpha 13-dependent plasma membrane recruitment of p115RhoGEF. Biochem J 371: 709–720, 2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.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 94: 7868–7872, 1997 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Bugrim AE. Regulation of Ca2+ release by cAMP-dependent protein kinase. A mechanism for agonist-specific calcium signaling? Cell Calcium 25: 219–226, 1999 [DOI] [PubMed] [Google Scholar]
- 17.Burgon PG, Lee WL, Nixon AB, Peralta EG, Casey PJ. Phosphorylation and nuclear translocation of a regulator of G protein signaling (RGS10). J Biol Chem 276: 32828–32834, 2001 [DOI] [PubMed] [Google Scholar]
- 18.Carman CV, Parent JL, Day PW, Pronin AN, Sternweis PM, Wedegaertner PB, Gilman AG, Benovic JL, Kozasa T. Selective regulation of Galpha(q/11) by an RGS domain in the G protein-coupled receptor kinase, GRK2. J Biol Chem 274: 34483–34492, 1999 [DOI] [PubMed] [Google Scholar]
- 19.Castro-Fernandez C, Janovick JA, Brothers SP, Fisher RA, Ji TH, Conn PM. Regulation of RGS3 and RGS10 palmitoylation by GnRH. Endocrinology 143: 1310–1317, 2002 [DOI] [PubMed] [Google Scholar]
- 20.Chakir K, Daya SK, Aiba T, Tunin RS, Dimaano VL, Abraham TP, Jaques-Robinson KM, Lai EW, Pacak K, Zhu WZ, Xiao RP, Tomaselli GF, Kass DA. Mechanisms of enhanced beta-adrenergic reserve from cardiac resynchronization therapy. Circulation 119: 1231–1240, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Chakir K, Depry C, Dimaano VL, Zhu WZ, Vanderheyden M, Bartunek J, Abraham TP, Tomaselli GF, Liu SB, Xiang YK, Zhang M, Takimoto E, Dulin N, Xiao RP, Zhang J, Kass DA. Galphas-biased beta2-adrenergic receptor signaling from restoring synchronous contraction in the failing heart. Sci Transl Med 3: 100ra188, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Chatterjee TK, Fisher RA. Mild heat and proteotoxic stress promote unique subcellular trafficking and nucleolar accumulation of RGS6 and other RGS proteins. Role of the RGS domain in stress-induced trafficking of RGS proteins. J Biol Chem 278: 30272–30282, 2003 [DOI] [PubMed] [Google Scholar]
- 23.Chen C, Seow KT, Guo K, Yaw LP, Lin SC. The membrane association domain of RGS16 contains unique amphipathic features that are conserved in RGS4 and RGS5. J Biol Chem 274: 19799–19806, 1999 [DOI] [PubMed] [Google Scholar]
- 24.Chen C, Wang H, Fong CW, Lin SC. Multiple phosphorylation sites in RGS16 differentially modulate its GAP activity. FEBS Lett 504: 16–22, 2001 [DOI] [PubMed] [Google Scholar]
- 25.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 100: 6604–6609, 2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Cheng YS, Lee TS, Hsu HC, Kou YR, Wu YL. Characterization of the transcriptional regulation of the regulator of G protein signaling 2 (RGS2) gene during 3T3–L1 preadipocyte differentiation. J Cell Biochem 105: 922–930, 2008 [DOI] [PubMed] [Google Scholar]
- 27.Chikumi H, Fukuhara S, Gutkind JS. Regulation of G protein-linked guanine nucleotide exchange factors for Rho, PDZ-RhoGEF, and LARG by tyrosine phosphorylation: evidence of a role for focal adhesion kinase. J Biol Chem 277: 12463–12473, 2002 [DOI] [PubMed] [Google Scholar]
- 28.Cho H, Harrison K, Schwartz O, Kehrl JH. The aorta and heart differentially express RGS (regulators of G-protein signalling) proteins that selectively regulate sphingosine 1-phosphate, angiotensin II and endothelin-1 signalling. Biochem J 371: 973–980, 2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Cho H, Kehrl JH. Regulation of immune function by G protein-coupled receptors, trimeric G proteins, and RGS proteins. Progr Mol Biol Transl Sci 86: 249–298, 2009 [DOI] [PubMed] [Google Scholar]
- 30.Cho H, Kim DU, Kehrl JH. RGS14 is a centrosomal and nuclear cytoplasmic shuttling protein that traffics to promyelocytic leukemia nuclear bodies following heat shock. J Biol Chem 280: 805–814, 2005 [DOI] [PubMed] [Google Scholar]
- 31.Clark MA, Sethi PR, Lambert NA. Active Galpha(q) subunits and M3 acetylcholine receptors promote distinct modes of association of RGS2 with the plasma membrane. FEBS Lett 581: 764–770, 2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Cunningham ML, Waldo GL, Hollinger S, Hepler JR, Harden TK. Protein kinase C phosphorylates RGS2 and modulates its capacity for negative regulation of Galpha 11 signaling. J Biol Chem 276: 5438–5444, 2001 [DOI] [PubMed] [Google Scholar]
- 33.De Vries L, Elenko E, Hubler L, Jones TL, Farquhar MG. GAIP is membrane-anchored by palmitoylation and interacts with the activated (GTP-bound) form of G alpha i subunits. Proc Natl Acad Sci USA 93: 15203–15208, 1996 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.De Vries L, Mousli M, Wurmser A, Farquhar MG. GAIP, a protein that specifically interacts with the trimeric G protein G alpha i3, is a member of a protein family with a highly conserved core domain. Proc Natl Acad Sci USA 92: 11916–11920, 1995 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Derrien A, Druey KM. RGS16 function is regulated by epidermal growth factor receptor-mediated tyrosine phosphorylation. J Biol Chem 276: 48532–48538, 2001 [DOI] [PubMed] [Google Scholar]
- 36.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 278: 16107–16116, 2003 [DOI] [PubMed] [Google Scholar]
- 37.Dietzel C, Kurjan J. Pheromonal regulation and sequence of the Saccharomyces cerevisiae SST2 gene: a model for desensitization to pheromone. Mol Cell Biol 7: 4169–4177, 1987 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.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 15: 3635–3643, 1995 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.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 16: 5194–5209, 1996 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Doupnik CA, Xu T, Shinaman JM. Profile of RGS expression in single rat atrial myocytes. Biochim Biophys Acta 1522: 97–107, 2001 [DOI] [PubMed] [Google Scholar]
- 41.Drenan RM, Doupnik CA, Boyle MP, Muglia LJ, Huettner JE, Linder ME, Blumer KJ. Palmitoylation regulates plasma membrane-nuclear shuttling of R7BP, a novel membrane anchor for the RGS7 family. J Cell Biol 169: 623–633, 2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Druey KM. Regulation of G-protein-coupled signaling pathways in allergic inflammation. Immunol Res 43: 62–76, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Druey KM, Blumer KJ, Kang VH, Kehrl JH. Inhibition of G-protein-mediated MAP kinase activation by a new mammalian gene family. Nature 379: 742–746, 1996 [DOI] [PubMed] [Google Scholar]
- 44.Druey KM, Sullivan BM, Brown D, Fischer ER, Watson N, Blumer KJ, Gerfen CR, Scheschonka A, Kehrl JH. Expression of GTPase-deficient Gialpha2 results in translocation of cytoplasmic RGS4 to the plasma membrane. J Biol Chem 273: 18405–18410, 1998 [DOI] [PubMed] [Google Scholar]
- 45.Druey KM, Ugur O, Caron JM, Chen CK, Backlund PS, Jones TL. Amino-terminal cysteine residues of RGS16 are required for palmitoylation and modulation of Gi- and Gq-mediated signaling. J Biol Chem 274: 18836–18842, 1999 [DOI] [PubMed] [Google Scholar]
- 46.Dulin NO, Sorokin A, Reed E, Elliott S, Kehrl JH, Dunn MJ. RGS3 inhibits G protein-mediated signaling via translocation to the membrane and binding to Galpha11. Mol Cell Biol 19: 714–723, 1999 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Endale M, Kim SD, Lee WM, Kim S, Suk K, Cho JY, Park HJ, Wagley Y, Kim S, Oh JW, Rhee MH. Ischemia induces regulator of G protein signaling 2 (RGS2) protein upregulation and enhances apoptosis in astrocytes. Am J Physiol Cell Physiol 298: C611–C623, 2010 [DOI] [PubMed] [Google Scholar]
- 48.Eszlinger M, Holzapfel HP, Voigt C, Arkenau C, Paschke R. RGS 2 expression is regulated by TSH and inhibits TSH receptor signaling. Eur J Endocrinol 151: 383–390, 2004 [DOI] [PubMed] [Google Scholar]
- 49.Fischer T, Elenko E, Wan L, Thomas G, Farquhar MG. Membrane-associated GAIP is a phosphoprotein and can be phosphorylated by clathrin-coated vesicles. Proc Natl Acad Sci USA 97: 4040–4045, 2000 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Fong CW, Zhang Y, Neo SY, Lin SC. Specific induction of RGS16 (regulator of G-protein signalling 16) mRNA by protein kinase C in CEM leukaemia cells is mediated via tumour necrosis factor alpha in a calcium-sensitive manner. Biochem J 352: 747–753, 2000 [PMC free article] [PubMed] [Google Scholar]
- 51.Garcia A, Prabhakar S, Hughan S, Anderson TW, Brock CJ, Pearce AC, Dwek RA, Watson SP, Hebestreit HF, Zitzmann N. Differential proteome analysis of TRAP-activated platelets: involvement of DOK-2 and phosphorylation of RGS proteins. Blood 103: 2088–2095, 2004 [DOI] [PubMed] [Google Scholar]
- 52.Garrison TR, Zhang Y, Pausch M, Apanovitch D, Aebersold R, Dohlman HG. Feedback phosphorylation of an RGS protein by MAP kinase in yeast. J Biol Chem 274: 36387–36391, 1999 [DOI] [PubMed] [Google Scholar]
- 53.Garzon J, Rodriguez-Munoz M, Lopez-Fando A, Sanchez-Blazquez P. Activation of mu-opioid receptors transfers control of Galpha subunits to the regulator of G-protein signaling. RGS9–2: role in receptor desensitization. J Biol Chem 280: 8951–8960, 2005 [DOI] [PubMed] [Google Scholar]
- 54.Georgoussi Z, Leontiadis L, Mazarakou G, Merkouris M, Hyde K, Hamm H. Selective interactions between G protein subunits and RGS4 with the C-terminal domains of the mu- and delta-opioid receptors regulate opioid receptor signaling. Cell Signal 18: 771–782, 2006 [DOI] [PubMed] [Google Scholar]
- 55.Giorelli M, Livrea P, Defazio G, Iacovelli L, Capobianco L, Picascia A, Sallese M, Martino D, Aniello MS, Trojano M, De Blasi A. Interferon beta-1a counteracts effects of activation on the expression of G-protein-coupled receptor kinases 2 and 3, beta-arrestin-1, and regulators of G-protein signalling 2 and 16 in human mononuclear leukocytes. Cell Signal 14: 673–678, 2002 [DOI] [PubMed] [Google Scholar]
- 56.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 57: 460–467, 2000 [DOI] [PubMed] [Google Scholar]
- 57.Gu S, Cifelli C, Wang S, Heximer SP. RGS proteins: identifying new GAPs in the understanding of blood pressure regulation and cardiovascular function. Clin Sci (Lond) 116: 391–399, 2009 [DOI] [PubMed] [Google Scholar]
- 58.Gu S, He J, Ho WT, Ramineni S, Thal DM, Natesh R, Tesmer JJ, Hepler JR, Heximer SP. Unique hydrophobic extension of the RGS2 amphipathic helix domain imparts increased plasma membrane binding and function relative to other RGS R4/B subfamily members. J Biol Chem 282: 33064–33075, 2007 [DOI] [PubMed] [Google Scholar]
- 59.Gu S, Tirgari S, Heximer SP. The RGS2 gene product from a candidate hypertension allele shows decreased plasma membrane association and inhibition of Gq. Mol Pharmacol 73: 1037–1043, 2008 [DOI] [PubMed] [Google Scholar]
- 60.Gunaje JJ, Bahrami AJ, Schwartz SM, Daum G, Mahoney WM., Jr PDGF-dependent regulation of regulator of G-protein signaling-5 expression and vascular smooth muscle cell functionality. Am J Physiol Cell Physiol 301: C478–C489, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Hague C, Bernstein LS, Ramineni S, Chen Z, Minneman KP, Hepler JR. Selective inhibition of alpha1A-adrenergic receptor signaling by RGS2 association with the receptor third intracellular loop. J Biol Chem 280: 27289–27295, 2005 [DOI] [PubMed] [Google Scholar]
- 62.Hao J, Michalek C, Zhang W, Zhu M, Xu X, Mende U. Regulation of cardiomyocyte signaling by RGS proteins: differential selectivity towards G proteins and susceptibility to regulation. J Mol Cell Cardiol 41: 51–61, 2006 [DOI] [PubMed] [Google Scholar]
- 63.Hausmann ON, Hu WH, Keren-Raifman T, Witherow DS, Wang Q, Levay K, Frydel B, Slepak VZ, Bethea JR. Spinal cord injury induces expression of RGS7 in microglia/macrophages in rats. Eur J Neurosci 15: 602–612, 2002 [DOI] [PubMed] [Google Scholar]
- 64.Hendriks-Balk MC, Hajji N, van Loenen PB, Michel MC, Peters SL, Alewijnse AE. Sphingosine-1-phosphate regulates RGS2 and RGS16 mRNA expression in vascular smooth muscle cells. Eur J Pharmacol 606: 25–31, 2009 [DOI] [PubMed] [Google Scholar]
- 65.Hendriks-Balk MC, Tjon-Atsoi M, Hajji N, Alewijnse AE, Peters SL. LPS differentially affects vasoconstrictor responses: a potential role for RGS16? J Physiol Biochem 65: 71–83, 2009 [DOI] [PubMed] [Google Scholar]
- 66.Hendriks-Balk MC, van Loenen PB, Hajji N, Michel MC, Peters SL, Alewijnse AE. S1P receptor signalling and RGS proteins; expression and function in vascular smooth muscle cells and transfected CHO cells. Eur J Pharmacol 600: 1–9, 2008 [DOI] [PubMed] [Google Scholar]
- 67.Heximer SP, Cristillo AD, Forsdyke DR. Comparison of mRNA expression of two regulators of G-protein signaling, RGS1/BL34/1R20 and RGS2/G0S8, in cultured human blood mononuclear cells. DNA Cell Biol 16: 589–598, 1997 [DOI] [PubMed] [Google Scholar]
- 68.Heximer SP, Lim H, Bernard JL, Blumer KJ. Mechanisms governing subcellular localization and function of human RGS2. J Biol Chem 276: 14195–14203, 2001 [DOI] [PubMed] [Google Scholar]
- 69.Hiol A, Davey PC, Osterhout JL, Waheed AA, Fischer ER, Chen CK, Milligan G, Druey KM, Jones TL. Palmitoylation regulates regulators of G-protein signaling (RGS) 16 function. I. Mutation of amino-terminal cysteine residues on RGS16 prevents its targeting to lipid rafts and palmitoylation of an internal cysteine residue. J Biol Chem 278: 19301–19308, 2003 [DOI] [PubMed] [Google Scholar]
- 70.Hollinger S, Ramineni S, Hepler JR. Phosphorylation of RGS14 by protein kinase A potentiates its activity toward Galphai. Biochemistry 42: 811–819, 2003 [DOI] [PubMed] [Google Scholar]
- 71.Hong JX, Wilson GL, Fox CH, Kehrl JH. Isolation and characterization of a novel B cell activation gene. J Immunol 150: 3895–3904, 1993 [PubMed] [Google Scholar]
- 72.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 9: 289, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Hu G, Jang GF, Cowan CW, Wensel TG, Palczewski K. Phosphorylation of RGS9–1 by an endogenous protein kinase in rod outer segments. J Biol Chem 276: 22287–22295, 2001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Hu G, Wensel TG. R9AP, a membrane anchor for the photoreceptor GTPase accelerating protein, RGS9-1. Proc Natl Acad Sci USA 99: 9755–9760, 2002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.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 437: 981–986, 2005 [DOI] [PubMed] [Google Scholar]
- 76.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 412: 35–43, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Hu W, Li F, Mahavadi S, Murthy KS. Upregulation of RGS4 expression by IL-1beta in colonic smooth muscle is enhanced by ERK1/2 and p38 MAPK and inhibited by the PI3K/Akt/GSK3beta pathway. Am J Physiol Cell Physiol 296: C1310–C1320, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Hu W, Mahavadi S, Li F, Murthy KS. Upregulation of RGS4 and downregulation of CPI-17 mediate inhibition of colonic muscle contraction by interleukin-1β. Am J Physiol Cell Physiol 293: C1991–C2000, 2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Huang J, Zhou H, Mahavadi S, Sriwai W, Murthy KS. Inhibition of Gαq-dependent PLC-β1 activity by PKG and PKA is mediated by phosphorylation of RGS4 and GRK2. Am J Physiol Cell Physiol 292: C200–C208, 2007 [DOI] [PubMed] [Google Scholar]
- 80.Hunt TW, Fields TA, Casey PJ, Peralta EG. RGS10 is a selective activator of G alpha i GTPase activity. Nature 383: 175–177, 1996 [DOI] [PubMed] [Google Scholar]
- 81.Hurst JH, Hooks SB. Regulator of G-protein signaling (RGS) proteins in cancer biology. Biochem Pharmacol 78: 1289–1297, 2009 [DOI] [PubMed] [Google Scholar]
- 82.Imagawa M, Tsuchiya T, Nishihara T. Identification of inducible genes at the early stage of adipocyte differentiation of 3T3–L1 cells. Biochem Biophys Res Commun 254: 299–305, 1999 [DOI] [PubMed] [Google Scholar]
- 83.Itoh M, Nagatomo K, Kubo Y, Saitoh O. Alternative splicing of RGS8 gene changes the binding property to the M1 muscarinic receptor to confer receptor type-specific Gq regulation. J Neurochem 99: 1505–1516, 2006 [DOI] [PubMed] [Google Scholar]
- 84.Iwai K, Koike M, Ohshima S, Miyatake K, Uchiyama Y, Saeki Y, Ishii M. RGS18 acts as a negative regulator of osteoclastogenesis by modulating the acid-sensing OGR1/NFAT signaling pathway. J Bone Miner Res 22: 1612–1620, 2007 [DOI] [PubMed] [Google Scholar]
- 85.Jayaraman M, Zhou H, Jia L, Cain MD, Blumer KJ. R9AP and R7BP: traffic cops for the RGS7 family in phototransduction and neuronal GPCR signaling. Trends Pharmacol Sci 30: 17–24, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Jean-Baptiste G, Li X, Yang Z, Heubach J, Gaudio S, Khoury C, Ravens U, Greenwood MT. Beta adrenergic receptor-mediated atrial specific up-regulation of RGS5. Life Sci 76: 1533–1545, 2005 [DOI] [PubMed] [Google Scholar]
- 87.Jensen KP, Covault J. Human miR-1271 is a miR-96 paralog with distinct non-conserved brain expression pattern. Nucleic Acids Res 39: 701–711, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Jin Y, An X, Ye Z, Cully B, Wu J, Li J. RGS5, a hypoxia-inducible apoptotic stimulator in endothelial cells. J Biol Chem 284: 23436–23443, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Ko JK, Choi KH, Kim IS, Jung EK, Park DH. Inducible RGS2 is a cross-talk regulator for parathyroid hormone signaling in rat osteoblast-like UMR106 cells. Biochem Biophys Res Commun 287: 1025–1033, 2001 [DOI] [PubMed] [Google Scholar]
- 90.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 84: 115–125, 1996 [DOI] [PubMed] [Google Scholar]
- 91.Kohno T, Igarashi Y. Attenuation of cell motility observed with high doses of sphingosine 1-phosphate or phosphorylated FTY720 involves RGS2 through its interactions with the receptor S1P. Genes Cells 13: 747–757, 2008 [DOI] [PubMed] [Google Scholar]
- 92.Kovoor A, Seyffarth P, Ebert J, Barghshoon S, Chen CK, Schwarz S, Axelrod JD, Cheyette BN, Simon MI, Lester HA, Schwarz J. D2 dopamine receptors colocalize regulator of G-protein signaling 9–2 (RGS9–2) via the RGS9 DEP domain, and RGS9 knock-out mice develop dyskinesias associated with dopamine pathways. J Neurosci 25: 2157–2165, 2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Krumins AM, Barker SA, Huang C, Sunahara RK, Yu K, Wilkie TM, Gold SJ, Mumby SM. Differentially regulated expression of endogenous RGS4 and RGS7. J Biol Chem 279: 2593–2599, 2004 [DOI] [PubMed] [Google Scholar]
- 94.Langer I, Tikhonova IG, Boulegue C, Esteve JP, Vatinel S, Ferrand A, Moroder L, Robberecht P, Fourmy D. Evidence for a direct and functional interaction between the regulators of G protein signaling-2 and phosphorylated C terminus of cholecystokinin-2 receptor. Mol Pharmacol 75: 502–513, 2009 [DOI] [PubMed] [Google Scholar]
- 95.Lee HK, Yeo S, Kim JS, Lee JG, Bae YS, Lee C, Baek SH. Protein kinase C-eta and phospholipase D2 pathway regulates foam cell formation via regulator of G protein signaling 2. Mol Pharmacol 78: 478–485, 2010 [DOI] [PubMed] [Google Scholar]
- 96.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 102: 15030–15035, 2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Leontiadis LJ, Papakonstantinou MP, Georgoussi Z. Regulator of G protein signaling 4 confers selectivity to specific G proteins to modulate mu- and delta-opioid receptor signaling. Cell Signal 21: 1218–1228, 2009 [DOI] [PubMed] [Google Scholar]
- 98.Li F, Murthy KS, Khalili K, Hu W. Cloning and characterization of rabbit Rgs4 promoter in gut smooth muscle. Gene 451: 45–53, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Li J, Adams LD, Wang X, Pabon L, Schwartz SM, Sane DC, Geary RL. Regulator of G protein signaling 5 marks peripheral arterial smooth muscle cells and is downregulated in atherosclerotic plaque. J Vasc Surg 40: 519–528, 2004 [DOI] [PubMed] [Google Scholar]
- 100.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 276: 18579–18590, 2001 [DOI] [PubMed] [Google Scholar]
- 101.Li Y, Hashim S., Anand-Srivastava M. B Angiotensin II-evoked enhanced expression of RGS2 attenuates Gi-mediated adenylyl cyclase signaling in A10 cells. Cardiovasc Res 66: 503–511, 2005 [DOI] [PubMed] [Google Scholar]
- 102.Linder AE, Diaz J, Ni W, Szasz T, Burnett R, Watts SW. Vascular reactivity, 5-HT uptake, and blood pressure in the serotonin transporter knockout rat. Am J Physiol Heart Circ Physiol 294: H1745–H1752, 2008 [DOI] [PubMed] [Google Scholar]
- 103.Liu M, Simon MI. Regulation by cAMP-dependent protein kinease of a G-protein-mediated phospholipase C. Nature 382: 83–87, 1996 [DOI] [PubMed] [Google Scholar]
- 104.Lu J, Gosslau A, Liu AY, Chen KY. PCR differential display-based identification of regulator of G protein signaling 10 as the target gene in human colon cancer cells induced by black tea polyphenol theaflavin monogallate. Eur J Pharmacol 601: 66–72, 2008 [DOI] [PubMed] [Google Scholar]
- 105.Lygren B, Carlson CR, Santamaria K, Lissandron V, McSorley T, Litzenberg J, Lorenz D, Wiesner B, Rosenthal W, Zaccolo M, Tasken K, Klussmann E. AKAP complex regulates Ca2+ re-uptake into heart sarcoplasmic reticulum. EMBO Rep 8: 1061–1067, 2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Masuho I, Itoh M, Itoh H, Saitoh O. The mechanism of membrane-translocation of regulator of G-protein signaling (RGS) 8 induced by Galpha expression. J Neurochem 88: 161–168, 2004 [DOI] [PubMed] [Google Scholar]
- 107.Meyer BH, Freuler F, Guerini D, Siehler S. Reversible translocation of p115-RhoGEF by G(12/13)-coupled receptors. J Cell Biochem 104: 1660–1670, 2008 [DOI] [PubMed] [Google Scholar]
- 108.Miles RR, Sluka JP, Santerre RF, Hale LV, Bloem L, Boguslawski G, Thirunavukkarasu K, Hock JM, Onyia JE. Dynamic regulation of RGS2 in bone: potential new insights into parathyroid hormone signaling mechanisms. Endocrinology 141: 28–36, 2000 [DOI] [PubMed] [Google Scholar]
- 109.Miyamoto-Matsubara M, Saitoh O, Maruyama K, Aizaki Y, Saito Y. Regulation of melanin-concentrating hormone receptor 1 signaling by RGS8 with the receptor third intracellular loop. Cell Signal 20: 2084–2094, 2008 [DOI] [PubMed] [Google Scholar]
- 110.Moroi K, Nishiyama M, Kawabata S, Ichiba H, Yajima T, Kimura S. Phosphorylation of Ser166 in RGS5 by protein kinase C causes loss of RGS function. Life Sci 81: 40–50, 2007 [DOI] [PubMed] [Google Scholar]
- 111.Muinos-Gimeno M, Espinosa-Parrilla Y, Guidi M, Kagerbauer B, Sipila T, Maron E, Pettai K, Kananen L, Navines R, Martin-Santos R, Gratacos M, Metspalu A, Hovatta I, Estivill X. Human microRNAs miR-22, miR-138–2, miR-148a, and miR-488 are associated with panic disorder and regulate several anxiety candidate genes and related pathways. Biol Psychiatry 69: 526–533, 2011 [DOI] [PubMed] [Google Scholar]
- 112.Nakagawa T, Minami M, Satoh M. Up-regulation of RGS4 mRNA by opioid receptor agonists in PC12 cells expressing cloned mu- or kappa-opioid receptors. Eur J Pharmacol 433: 29–36, 2001 [DOI] [PubMed] [Google Scholar]
- 113.Neitzel KL, Hepler JR. Cellular mechanisms that determine selective RGS protein regulation of G protein-coupled receptor signaling. Semin Cell Dev Biol 17: 383–389, 2006 [DOI] [PubMed] [Google Scholar]
- 114.Ni J, Qu L, Yang H, Wang M, Huang Y. Palmitoylation and its effect on the GTPase-activating activity and conformation of RGS2. Int J Biochem Cell Biol 38: 2209–2218, 2006 [DOI] [PubMed] [Google Scholar]
- 115.Niu J, Scheschonka A, Druey KM, Davis A, Reed E, Kolenko V, Bodnar R, Voyno-Yasenetskaya T, Du X, Kehrl J, Dulin NO. RGS3 interacts with 14–3-3 via the N-terminal region distinct from the RGS (regulator of G-protein signalling) domain. Biochem J 365: 677–684, 2002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Nunn C, Zou MX, Sobiesiak AJ, Roy AA, Kirshenbaum LA, Chidiac P. RGS2 inhibits beta-adrenergic receptor-induced cardiomyocyte hypertrophy. Cell Signal 22: 1231–1239, 2010 [DOI] [PubMed] [Google Scholar]
- 117.Ogier-Denis E, Pattingre S, El Benna J, Codogno P. Erk1/2-dependent phosphorylation of Galpha-interacting protein stimulates its GTPase accelerating activity and autophagy in human colon cancer cells. J Biol Chem 275: 39090–39095, 2000 [DOI] [PubMed] [Google Scholar]
- 118.Ogier-Denis E, Petiot A, Bauvy C, Codogno P. Control of the expression and activity of the Galpha-interacting protein (GAIP) in human intestinal cells. J Biol Chem 272: 24599–24603, 1997 [DOI] [PubMed] [Google Scholar]
- 119.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 282: 31656–31665, 2007 [DOI] [PubMed] [Google Scholar]
- 120.Osterhout JL, Waheed AA, Hiol A, Ward RJ, Davey PC, Nini L, Wang J, Milligan G, Jones TL, Druey KM. Palmitoylation regulates regulator of G-protein signaling (RGS) 16 function. II. Palmitoylation of a cysteine residue in the RGS box is critical for RGS16 GTPase accelerating activity and regulation of Gi-coupled signalling. J Biol Chem 278: 19309–19316, 2003 [DOI] [PubMed] [Google Scholar]
- 121.Pagano M, Jordan JD, Neves SR, Nguyen T, Iyengar R. Galphao/i-stimulated proteosomal degradation of RGS20: a mechanism for temporal integration of Gs and Gi pathways. Cell Signal 20: 1190–1197, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Panetta R, Guo Y, Magder S, Greenwood MT. Regulators of G-protein signaling (RGS) 1 and 16 are induced in response to bacterial lipopolysaccharide and stimulate c-fos promoter expression. Biochem Biophys Res Commun 259: 550–556, 1999 [DOI] [PubMed] [Google Scholar]
- 123.Park ES, Echetebu CO, Soloff S, Soloff MS. Oxytocin stimulation of RGS2 mRNA expression in cultured human myometrial cells. Am J Physiol Endocrinol Metab 282: E580–E584, 2002 [DOI] [PubMed] [Google Scholar]
- 124.Patten M, Bunemann J, Thoma B, Kramer E, Thoenes M, Stube S, Mittmann C, Wieland T. Endotoxin induces desensitization of cardiac endothelin-1 receptor signaling by increased expression of RGS4 and RGS16. Cardiovasc Res 53: 156–164, 2002 [DOI] [PubMed] [Google Scholar]
- 125.Patten M, Stube S, Thoma B, Wieland T. Interleukin-1beta mediates endotoxin- and tumor necrosis factor alpha-induced RGS16 protein expression in cultured cardiac myocytes. Naunyn Schmiedeberg's Arch Pharmacol 368: 360–365, 2003 [DOI] [PubMed] [Google Scholar]
- 126.Pedram A, Razandi M, Kehrl J, Levin ER. Natriuretic peptides inhibit G protein activation. Mediation through cross-talk between cyclic GMP-dependent protein kinase and regulators of G protein-signaling proteins. J Biol Chem 275: 7365–7372, 2000 [DOI] [PubMed] [Google Scholar]
- 127.Pepperl DJ, Shah-Basu S, VanLeeuwen D, Granneman JG, MacKenzie RG. Regulation of RGS mRNAs by cAMP in PC12 cells. Biochem Biophys Res Commun 243: 52–55, 1998 [DOI] [PubMed] [Google Scholar]
- 128.Popov SG, Krishna UM, Falck JR, Wilkie TM. Ca2+/Calmodulin reverses phosphatidylinositol 3,4, 5-trisphosphate-dependent inhibition of regulators of G protein-signaling GTPase-activating protein activity. J Biol Chem 275: 18962–18968, 2000 [DOI] [PubMed] [Google Scholar]
- 129.Posokhova E, Wydeven N, Allen KL, Wickman K, Martemyanov KA. RGS6/Gss5 complex accelerates IKACh gating kinetics in atrial myocytes and modulates parasympathetic regulation of heart rate. Circ Res 107: 1350–1354, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Psifogeorgou K, Papakosta P, Russo SJ, Neve RL, Kardassis D, Gold SJ, Zachariou V. RGS9-2 is a negative modulator of mu-opioid receptor function. J Neurochem 103: 617–625, 2007 [DOI] [PubMed] [Google Scholar]
- 131.Rezabkova L, Boura E, Herman P, Vecer J, Bourova L, Sulc M, Svoboda P, Obsilova V, Obsil T. 14-3-3 protein interacts with and affects the structure of RGS domain of regulator of G protein signaling 3 (RGS3). J Struct Biol 170: 451–461, 2010 [DOI] [PubMed] [Google Scholar]
- 132.Riekenberg S, Farhat K, Debarry J, Heine H, Jung G, Wiesmuller KH, Ulmer AJ. Regulators of G-protein signalling are modulated by bacterial lipopeptides and lipopolysaccharide. FEBS J 276: 649–659, 2009 [DOI] [PubMed] [Google Scholar]
- 133.Rimler A, Jockers R, Lupowitz Z, Sampson SR, Zisapel N. Differential effects of melatonin and its downstream effector PKCalpha on subcellular localization of RGS proteins. J Pineal Res 40: 144–152, 2006 [DOI] [PubMed] [Google Scholar]
- 134.Rodriguez-Munoz M, Bermudez D, Sanchez-Blazquez P, Garzon J. Sumoylated RGS-Rz proteins act as scaffolds for Mu-opioid receptors and G-protein complexes in mouse brain. Neuropsychopharmacology 32: 842–850, 2007 [DOI] [PubMed] [Google Scholar]
- 135.Rogers JH, Tamirisa P, Kovacs A, Weinheimer C, Courtois M, Blumer KJ, Kelly DP, Muslin AJ. RGS4 causes increased mortality and reduced cardiac hypertrophy in response to pressure overload. J Clin Invest 104: 567–576, 1999 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Rogers JH, Tsirka A, Kovacs A, Blumer KJ, Dorn GW, II, Muslin AJ. RGS4 reduces contractile dysfunction and hypertrophic gene induction in Galpha q overexpressing mice. J Mol Cell Cardiol 33: 209–218, 2001 [DOI] [PubMed] [Google Scholar]
- 137.Romero DG, Plonczynski MW, Gomez-Sanchez EP, Yanes LL., Gomez-Sanchez C. E RGS2 is regulated by angiotensin II and functions as a negative feedback of aldosterone production in H295R human adrenocortical cells. Endocrinology 147: 3889–3897, 2006 [DOI] [PubMed] [Google Scholar]
- 138.Romero DG, Zhou MY, Yanes LL, Plonczynski MW, Washington TR, Gomez-Sanchez CE., Gomez-Sanchez E. P Regulators of G-protein signaling 4 in adrenal gland: localization, regulation, and role in aldosterone secretion. J Endocrinol 194: 429–440, 2007 [DOI] [PubMed] [Google Scholar]
- 139.Rose JJ, Taylor JB, Shi J, Cockett MI, Jones PG, Hepler JR. RGS7 is palmitoylated and exists as biochemically distinct forms. J Neurochem 75: 2103–2112, 2000 [DOI] [PubMed] [Google Scholar]
- 140.Roy AA, Baragli A, Bernstein LS, Hepler JR, Hebert TE, Chidiac P. RGS2 interacts with Gs and adenylyl cyclase in living cells. Cell Signal 18: 336–348, 2006 [DOI] [PubMed] [Google Scholar]
- 141.Roy AA, Lemberg KE, Chidiac P. Recruitment of RGS2 and RGS4 to the plasma membrane by G proteins and receptors reflects functional interactions. Mol Pharmacol 64: 587–593, 2003 [DOI] [PubMed] [Google Scholar]
- 142.Roy AA, Nunn C, Ming H, Zou MX, Penninger J, Kirshenbaum LA, Dixon SJ, Chidiac P. Up-regulation of endogenous RGS2 mediates cross-desensitization between Gs and Gq signaling in osteoblasts. J Biol Chem 281: 32684–32693, 2006 [DOI] [PubMed] [Google Scholar]
- 143.Saha S, Kashina A. Posttranslational arginylation as a global biological regulator. Dev Biol 358: 1–8, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Saitoh O, Masuho I, Terakawa I, Nomoto S, Asano T, Kubo Y. Regulator of G protein signaling 8 (RGS8) requires its NH2 terminus for subcellular localization and acute desensitization of G protein-gated K+ channels. J Biol Chem 276: 5052–5058, 2001 [DOI] [PubMed] [Google Scholar]
- 145.Salim S, Dessauer CW. Analysis of the interaction between RGS2 and adenylyl cyclase. Meth Enzymol 390: 83–99, 2004 [DOI] [PubMed] [Google Scholar]
- 146.Sandiford SL, Wang Q, Levay K, Buchwald P, Slepak VZ. Molecular organization of the complex between the muscarinic M3 receptor and the regulator of G protein signaling, Gbeta(5)-RGS7. Biochemistry 49: 4998–5006, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Santos de Araujo RM, Oba Y, Moriyama K. Role of regulator of G-protein signaling 2 (RGS2) in periodontal ligament cells under mechanical stress. Cell Biochem Funct 25: 753–758, 2007 [DOI] [PubMed] [Google Scholar]
- 148.Schwendt M, McGinty JF. Regulator of G-protein signaling 4 interacts with metabotropic glutamate receptor subtype 5 in rat striatum: relevance to amphetamine behavioral sensitization. J Pharmacol Exp Ther 323: 650–657, 2007 [DOI] [PubMed] [Google Scholar]
- 149.Sethakorn N, Yau DM, Dulin NO. Non-canonical functions of RGS proteins. Cell Signal 22: 1274–1281, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Shi GX, Harrison K, Han SB, Moratz C, Kehrl JH. Toll-like receptor signaling alters the expression of regulator of G protein signaling proteins in dendritic cells: implications for G protein-coupled receptor signaling. J Immunol 172: 5175–5184, 2004 [DOI] [PubMed] [Google Scholar]
- 151.Shi GX, Harrison K, Wilson GL, Moratz C, Kehrl JH. RGS13 regulates germinal center B lymphocytes responsiveness to CXC chemokine ligand (CXCL)12 and CXCL13. J Immunol 169: 2507–2515, 2002 [DOI] [PubMed] [Google Scholar]
- 152.Shu FJ, Ramineni S, Amyot W, Hepler JR. Selective interactions between Gi alpha1 and Gi alpha3 and the GoLoco/GPR domain of RGS14 influence its dynamic subcellular localization. Cell Signal 19: 163–176, 2007 [DOI] [PubMed] [Google Scholar]
- 153.Sinnarajah S, Dessauer CW, Srikumar D, Chen J, Yuen J, Yilma S, Dennis JC, Morrison EE, Vodyanoy V, Kehrl JH. RGS2 regulates signal transduction in olfactory neurons by attenuating activation of adenylyl cyclase III. Nature 409: 1051–1055, 2001 [DOI] [PubMed] [Google Scholar]
- 154.Sjogren B. Regulator of G protein signaling proteins as drug targets: current state and future possibilities. Adv Pharmacol 62: 315–347, 2011 [DOI] [PubMed] [Google Scholar]
- 155.Sjogren B, Blazer LL, Neubig RR. Regulators of G protein signaling proteins as targets for drug discovery. Progr Mol Biol Translational Sci 91: 81–119, 2010 [DOI] [PubMed] [Google Scholar]
- 156.Sokal I, Hu G, Liang Y, Mao M, Wensel TG, Palczewski K. Identification of protein kinase C isozymes responsible for the phosphorylation of photoreceptor-specific RGS9-1 at Ser475. J Biol Chem 278: 8316–8325, 2003 [DOI] [PubMed] [Google Scholar]
- 157.Song L, De Sarno P, Jope RS. Muscarinic receptor stimulation increases regulators of G-protein signaling 2 mRNA levels through a protein kinase C-dependent mechanism. J Biol Chem 274: 29689–29693, 1999 [DOI] [PubMed] [Google Scholar]
- 158.Song L, Jope RS. Cellular stress increases RGS2 mRNA and decreases RGS4 mRNA levels in SH-SY5Y cells. Neurosci Lett 402: 205–209, 2006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Song L, Zmijewski JW, Jope RS. RGS2: regulation of expression and nuclear localization. Biochem Biophys Res Commun 283: 102–106, 2001 [DOI] [PubMed] [Google Scholar]
- 160.Srinivasa SP, Bernstein LS, Blumer KJ, Linder ME. Plasma membrane localization is required for RGS4 function in Saccharomyces cerevisiae. Proc Natl Acad Sci USA 95: 5584–5589, 1998 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Stuebe S, Wieland T, Kraemer E, Stritzky A, Schroeder D, Seekamp S, Vogt A, Chen CK, Patten M. Sphingosine-1-phosphate and endothelin-1 induce the expression of rgs16 protein in cardiac myocytes by transcriptional activation of the rgs16 gene. Naunyn Schmiedeberg's Arch Pharmacol 376: 363–373, 2008 [DOI] [PubMed] [Google Scholar]
- 162.Sun Y, Fang R, Li C, Li L, Li F, Ye X, Chen H. Hsa-mir-182 suppresses lung tumorigenesis through down regulation of RGS17 expression in vitro. Biochem Biophys Res Commun 396: 501–507, 2010 [DOI] [PubMed] [Google Scholar]
- 163.Suzuki N, Nakamura S, Mano H, Kozasa T. Galpha 12 activates Rho GTPase through tyrosine-phosphorylated leukemia-associated RhoGEF. Proc Natl Acad Sci USA 100: 733–738, 2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Takesono A, Zahner J, Blumer KJ, Nagao T, Kurose H. Negative regulation of alpha2-adrenergic receptor-mediated Gi signalling by a novel pathway. Biochem J 343: 77–85, 1999 [PMC free article] [PubMed] [Google Scholar]
- 165.Takida S, Fischer CC, Wedegaertner PB. Palmitoylation and plasma membrane targeting of RGS7 are promoted by alpha o. Mol Pharmacol 67: 132–139, 2005 [DOI] [PubMed] [Google Scholar]
- 166.Takimoto E, Koitabashi N, Hsu S, Ketner EA, Zhang M, Nagayama T, Bedja D, Gabrielson KL, Blanton R, Siderovski DP, Mendelsohn ME, Kass DA. Regulator of G protein signaling 2 mediates cardiac compensation to pressure overload and antihypertrophic effects of PDE5 inhibition in mice. J Clin Invest 119: 408–420, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Tang KM, Wang GR, Lu P, Karas RH, Aronovitz M, Heximer SP, Kaltenbronn KM, Blumer KJ, Siderovski DP, Zhu Y, Mendelsohn ME. Regulator of G-protein signaling-2 mediates vascular smooth muscle relaxation and blood pressure. Nat Med 9: 1506–1512, 2003 [DOI] [PubMed] [Google Scholar]
- 168.Terzi D, Stergiou E, King SL, Zachariou V. Regulators of G protein signaling in neuropsychiatric disorders. Progr Mol Biol Transl Sci 86: 299–333, 2009 [DOI] [PubMed] [Google Scholar]
- 169.Thirunavukkarasu K, Halladay DL, Miles RR, Geringer CD, Onyia JE. Analysis of regulator of G-protein signaling-2 (RGS-2) expression and function in osteoblastic cells. J Cell Biochem 85: 837–850, 2002 [DOI] [PubMed] [Google Scholar]
- 170.Tobin AB, Butcher AJ, Kong Location KC, location, location… site-specific GPCR phosphorylation offers a mechanism for cell-type-specific signalling. Trends Pharmacol Sci 29: 413–420, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Tokudome T, Kishimoto I, Horio T, Arai Y, Schwenke DO, Hino J, Okano I, Kawano Y, Kohno M, Miyazato M, Nakao K, Kangawa K. Regulator of G-protein signaling subtype 4 mediates antihypertrophic effect of locally secreted natriuretic peptides in the heart. Circulation 117: 2329–2339, 2008 [DOI] [PubMed] [Google Scholar]
- 172.Tosetti P, Pathak N, Jacob MH, Dunlap K. RGS3 mediates a calcium-dependent termination of G protein signaling in sensory neurons. Proc Natl Acad Sci USA 100: 7337–7342, 2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Tran T, Paz P, Velichko S, Cifrese J, Belur P, Yamaguchi KD, Ku K, Mirshahpanah P, Reder AT, Croze E. Interferonbeta-1b induces the expression of RGS1 a negative regulator of G-protein signaling. Int J Cell Biol 2010: 529376, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Traynor J. Regulator of G protein-signaling proteins and addictive drugs. Ann NY Acad Sci 1187: 341–352, 2010 [DOI] [PubMed] [Google Scholar]
- 175.Traynor JR, Neubig RR. Regulators of G protein signaling & drugs of abuse. Mol Interv 5: 30–41, 2005 [DOI] [PubMed] [Google Scholar]
- 176.Tsingotjidou A, Nervina JM, Pham L, Bezouglaia O, Tetradis S. Parathyroid hormone induces RGS-2 expression by a cyclic adenosine 3′,5′-monophosphate-mediated pathway in primary neonatal murine osteoblasts. Bone 30: 677–684, 2002 [DOI] [PubMed] [Google Scholar]
- 177.Tu Y, Popov S, Slaughter C, Ross EM. Palmitoylation of a conserved cysteine in the regulator of G protein signaling (RGS) domain modulates the GTPase-activating activity of RGS4 and RGS10. J Biol Chem 274: 38260–38267, 1999 [DOI] [PubMed] [Google Scholar]
- 178.Ueno Y, Shinki T, Nagai Y, Murayama H, Fujii K, Suda T. In vivo administration of 1,25-dihydroxyvitamin D3 suppresses the expression of RANKL mRNA in bone of thyroparathyroidectomized rats constantly infused with PTH. J Cell Biochem 90: 267–277, 2003 [DOI] [PubMed] [Google Scholar]
- 179.Ujioka T, Russell DL, Okamura H, Richards JS, Espey LL. Expression of regulator of G-protein signaling protein-2 gene in the rat ovary at the time of ovulation. Biol Reprod 63: 1513–1517, 2000 [DOI] [PubMed] [Google Scholar]
- 180.Wang J, Xie Y, Wolff DW, Abel PW, Tu Y. DHHC protein-dependent palmitoylation protects regulator of G-protein signaling 4 from proteasome degradation. FEBS Lett 584: 4570–4574, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Wang Q, Traynor JR. Opioid-induced down-regulation of RGS4: role of ubiquitination and implications for receptor cross-talk. J Biol Chem 286: 7854–7864, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Wang X, Adams LD, Pabon LM, Mahoney WM, Jr, Beaudry D, Gunaje J, Geary RL, Deblois D, Schwartz SM. RGS5, RGS4, and RGS2 expression and aortic contractibility are dynamically co-regulated during aortic banding-induced hypertrophy. J Mol Cell Cardiol 44: 539–550, 2008 [DOI] [PubMed] [Google Scholar]
- 183.Wang X, Zeng W, Soyombo AA, Tang W, Ross EM, Barnes AP, Milgram SL, Penninger JM, Allen PB, Greengard P, Muallem S. Spinophilin regulates Ca2+ signalling by binding the N-terminal domain of RGS2 and the third intracellular loop of G-protein-coupled receptors. Nat Cell Biol 7: 405–411, 2005 [DOI] [PubMed] [Google Scholar]
- 184.Ward RJ, Milligan G. A key serine for the GTPase-activating protein function of regulator of G protein signaling proteins is not a general target for 14–3-3 interactions. Mol Pharmacol 68: 1821–1830, 2005 [DOI] [PubMed] [Google Scholar]
- 185.Watson N, Linder ME, Druey KM, Kehrl JH, Blumer KJ. RGS family members: GTPase-activating proteins for heterotrimeric G-protein alpha-subunits. Nature 383: 172–175, 1996 [DOI] [PubMed] [Google Scholar]
- 186.Wensel TG. RGS9–1 phosphorylation and Ca2+. Adv Exp Med Biol 514: 125–129, 2002 [DOI] [PubMed] [Google Scholar]
- 187.Wieland T, Lutz S, Chidiac P. Regulators of G protein signalling: a spotlight on emerging functions in the cardiovascular system. Curr Opin Pharmacol 7: 201–207, 2007 [DOI] [PubMed] [Google Scholar]
- 188.Willars GB. Mammalian RGS proteins: multifunctional regulators of cellular signalling. Semin Cell Dev Biol 17: 363–376, 2006 [DOI] [PubMed] [Google Scholar]
- 189.Xie 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 184: 2289–2296, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Xie 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 69: 5743–5751, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Xie Z, Geiger TR, Johnson EN, Nyborg JK, Druey KM. RGS13 acts as a nuclear repressor of CREB. Mol Cell 31: 660–670, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Xie Z, Gong MC, Su W, Turk J, Guo Z. Group VIA phospholipase A2 (iPLA2beta) participates in angiotensin II-induced transcriptional up-regulation of regulator of g-protein signaling-2 in vascular smooth muscle cells. J Biol Chem 282: 25278–25289, 2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Xie Z, Liu D, Liu S, Calderon L, Zhao G, Turk J, Guo Z. Identification of a cAMP-response element in the RGS2 promoter as a Key cis-regulatory element for RGS2 transcriptional regulation by angiotensin II in cultured VSMC. J Biol Chem 286: 44646–44658, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Xie Z, Yang Z, Druey KM. Phosphorylation of RGS13 by the cyclic AMP-dependent protein kinase inhibits RGS13 degradation. J Mol Cell Biol 2: 357–365, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Yamada T, Ohoka Y, Kogo M, Inagaki S. Physical and functional interactions of the lysophosphatidic acid receptors with PDZ domain-containing Rho guanine nucleotide exchange factors (RhoGEFs). J Biol Chem 280: 19358–19363, 2005 [DOI] [PubMed] [Google Scholar]
- 196.Yang J, Huang J, Maity B, Gao Z, Lorca RA, Gudmundsson H, Li J, Stewart A, Swaminathan PD, Ibeawuchi SR, Shepherd A, Chen CK, Kutschke W, Mohler PJ, Mohapatra DP, Anderson ME, Fisher RA. RGS6, a modulator of parasympathetic activation in heart. Circ Res 107: 1345–1349, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Yang J, Kamide K, Kokubo Y, Takiuchi S, Tanaka C, Banno M, Miwa Y, Yoshii M, Horio T, Okayama A, Tomoike H, Kawano Y, Miyata T. Genetic variations of regulator of G-protein signaling 2 in hypertensive patients and in the general population. J Hypertens 23: 1497–1505, 2005 [DOI] [PubMed] [Google Scholar]
- 198.Yang Z, Cooper PR, Damera G, Mukhopadhyay I, Cho H, Kehrl JH, Panettieri RA, Jr, Druey KM. Beta-agonist-associated reduction in RGS5 expression promotes airway smooth muscle hyper-responsiveness. J Biol Chem 286: 11444–11455, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Zhang P, Mende U. Regulators of G-protein signaling in the heart and their potential as therapeutic targets. Circ Res 109: 320–333, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Zhang P, Su J, King ME, Maldonado AE, Park C, Mende U. Regulator of G protein signaling 2 is a functionally important negative regulator of angiotensin II-induced cardiac fibroblast responses. Am J Physiol Heart Circ Physiol 301: H147–H156, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Zhang S, Watson N, Zahner J, Rottman JN, Blumer KJ, Muslin AJ. RGS3 and RGS4 are GTPase activating proteins in the heart. J Mol Cell Cardiol 30: 269–276, 1998 [DOI] [PubMed] [Google Scholar]
- 202.Zhang W, Anger T, Su J, Hao J, Xu X, Zhu M, Gach A, Cui L, Liao R, Mende U. Selective loss of fine tuning of Gq/11 signaling by RGS2 protein exacerbates cardiomyocyte hypertrophy. J Biol Chem 281: 5811–5820, 2006 [DOI] [PubMed] [Google Scholar]
- 203.Zmijewski JW, Song L, Harkins L, Cobbs CS, Jope RS. Oxidative stress and heat shock stimulate RGS2 expression in 1321N1 astrocytoma cells. Arch Biochem Biophys 392: 192–196, 2001 [DOI] [PubMed] [Google Scholar]
- 204.Zou MX, Roy AA, Zhao Q, Kirshenbaum LA, Karmazyn M, Chidiac P. RGS2 is upregulated by and attenuates the hypertrophic effect of alpha1-adrenergic activation in cultured ventricular myocytes. Cell Signal 18: 1655–1663, 2006 [DOI] [PubMed] [Google Scholar]