Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Biomol Ther (Seoul) ; 32(4): 442-450, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38844801

ABSTRACT

The type-1 cannabinoid receptor (CB1R) is a potential therapeutic target in several pathological conditions, including neuropsychological disorders and neurodegenerative diseases. Owing to their structural diversity, it is not easy to derive general structure-activity relationships (SARs) for CB1R ligands. In this study, CB1R ligands were classified into six structural families, and the corresponding SAR was determined for their affinities for CB1R. In addition, we determined their functional activities for the activation of extracellular signal-regulated kinases (ERKs). Among derivatives of indol-3-yl-methanone, the highest ligand affinity was observed when a pentyl and a naphthalenyl group were attached to the N1 position of the indole ring and the carbon site of the methanone moiety, respectively. In the case of adamantane indazole-3-carboxamide derivatives, the presence of fluorine in the pentyl group, the substituent at the N1 position of the indazole ring, strongly increased the affinity for CB1R. For (naphthalen-1-yl) methanone derivatives, the presence of 4-alkoxynaphthalene in the methanone moiety was more beneficial for the affinity to CB1R than that of a heterocyclic ring. The functional activities of the tested compounds, evaluated through ERK assay, were correlated with their affinity for CB1R, suggesting their agonistic nature. In conclusion, this study provides valuable insight for designing novel ligands for CB1R, which can be used to control psychiatric disorders and drug abuse.

2.
Biochem Biophys Res Commun ; 666: 154-161, 2023 07 23.
Article in English | MEDLINE | ID: mdl-37187093

ABSTRACT

Cannabinoid receptor 2 (CB2) and lysophosphatidic acid receptor 5 (LPA5) are both classified as G-protein coupled receptors (GPCRs) activated by bioactive lipids and are highly expressed in colon cancer cells. However, crosstalk between two receptors and its potential effects on cancer cell physiology have not been fully elucidated. In the present study, the results of bioluminescence resonance energy transfer analysis showed that, among the LPA receptors, CB2 strongly and specifically interacted with LPA5. Both receptors were co-localized in the plasma membrane in the absence of agonists, and the receptors were co-internalized upon activation of either receptor alone or both receptors together. We further investigated the effects of expression of both receptors on cell proliferation and migration, and the molecular mechanisms underlying these effects in HCT116 colon cancer cells. Co-expression of receptors significantly increased cell proliferation and migration by increasing Akt phosphorylation and tumor progression-related gene expression, whereas no such effect was seen upon expression of either receptor alone. These results suggest the possibility of physical and functional crosstalk between CB2 and LPA5.


Subject(s)
Colonic Neoplasms , Receptors, Lysophosphatidic Acid , Humans , Cell Line, Tumor , Cell Proliferation , Colonic Neoplasms/pathology , Lysophospholipids/pharmacology , Receptors, Cannabinoid , Receptors, Lysophosphatidic Acid/metabolism , HCT116 Cells
3.
Biochem Biophys Res Commun ; 570: 53-59, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34271437

ABSTRACT

Lysophosphatidylinositol (LPI) and sphingosine-1-phosphate (S1P) are bioactive lipids implicated in various cellular events including proliferation, migration, and cancer progression. LPI and S1P act as ligands for G-protein coupled GPR55 and S1P receptors, respectively, and activate specific signaling pathways. Both receptors are highly expressed in various cancer tissues and associated with tumor progression. However, physical and functional crosstalk between the two receptors has not been elucidated to date. Bioluminescence resonance energy transfer (BRET) experiments in the current study showed that S1P5 strongly and specifically interacts with GPR55. We observed co-internalization of both receptors upon agonist stimulation. Notably, activation of one receptor induced co-internalization of the partner receptor. Next, we examined functional crosstalk of the two receptors. Interestingly, while activation of the individual receptors augmented cell proliferation, ERK phosphorylation and cancer-associated gene expression in HCT116 cells, co-activation of both receptors inhibited these stimulatory effects. Our collective findings indicate that GPR55 and S1P5 form a heterodimer and their co-activation attenuates the stimulatory activity of each receptor on colon cancer progression.


Subject(s)
Lysophospholipids/metabolism , Receptors, Cannabinoid/metabolism , Sphingosine-1-Phosphate Receptors/metabolism , Cell Proliferation , Cell Survival , Disease Progression , Endocytosis , Gene Expression Regulation , HCT116 Cells , Humans , Protein Binding
4.
FEBS Lett ; 595(13): 1806-1818, 2021 07.
Article in English | MEDLINE | ID: mdl-33959968

ABSTRACT

Lysophosphatidic acid (LPA) and lysophosphatidylinositol bind to the G protein-coupled receptors (GPCRs) LPA and GPR55, respectively. LPA2 , a type 2 LPA receptor, and GPR55 are highly expressed in colon cancer and involved in cancer progression. However, crosstalk between the two receptors and potential effects on cellular physiology are not fully understood. Here, using BRET analysis, we found that LPA2 and GPR55 interact in live cells. In the presence of both receptors, LPA2 and/or GPR55 activation facilitated co-internalization, and activation of GPR55, uncoupled with Gαi , induced reduction of intracellular cAMP. Notably, co-activation of receptors synergistically triggered further decline in the cAMP level, promoted cell proliferation, and increased the expression of cancer progression-related genes, suggesting that physical and functional crosstalk between LPA2 and GRR55 is involved in cancer progression.


Subject(s)
Colonic Neoplasms/metabolism , Receptors, Cannabinoid/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Up-Regulation , Bioluminescence Resonance Energy Transfer Techniques , Cell Proliferation , Cyclic AMP/metabolism , Disease Progression , Gene Expression Regulation, Neoplastic , HCT116 Cells , HEK293 Cells , Humans , Protein Binding , Signal Transduction
5.
Biochem Biophys Res Commun ; 533(4): 1233-1239, 2020 12 17.
Article in English | MEDLINE | ID: mdl-33092790

ABSTRACT

Activation of seven-transmembrane G-protein coupled receptor (GPCR) mediates extracellular signals into intracellular responses. G-protein coupled receptor 55 (GPR55) is one of GPCRs and activated by endogenous cannabinoids. A family of regulators of G-protein signaling (RGS) stimulates GTP hydrolysis of alpha subunit of G-protein (Gα) and inhibits GPCR/Gα-mediated signaling. RGS2 is member of R4 RGS family and mainly attenuates GPCR/Gαq signaling. Although RGS2 is known to modulate some GPCR signaling, the specific effects of RGS2 on GPR55-mediated signaling are not fully understood at present. Previously, we reported some RGS proteins interact with protease-activated receptors, one of GPCRs, and modulate their functions. Here, we investigated whether GPR55 interacts with RGS2, employing bioluminescence resonance energy transfer and co-immunoprecipitation analyses. Interestingly, GPR55 interacted with RGS2 alone and also formed a ternary complex with RGS2 and either Gαq or Gα12. In the presence of GPR55 alone and together with Gαq or Gα12, RGS2 translocated from the cytoplasm to plasma membrane while RGS1 remained in the cytoplasm. GPR55 activation significantly induced ERK phosphorylation and intracellular calcium mobilization, which were markedly inhibited by RGS2 in HCT116 colon cancer cell line. Furthermore, GPR55-mediated cell proliferation and migration of HCT116 cells, was significantly attenuated by RGS2. Our collective findings highlight a novel physiological function of RGS2, supporting its utility as a therapeutic target to control GPR55-induced pathophysiology.


Subject(s)
RGS Proteins/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , GTP-Binding Protein alpha Subunits/metabolism , HEK293 Cells , Humans , Neoplasms/pathology , Receptors, Cannabinoid/metabolism , Signal Transduction
6.
Cell Commun Signal ; 18(1): 86, 2020 06 09.
Article in English | MEDLINE | ID: mdl-32517689

ABSTRACT

BACKGROUND: Protease-activated receptor 4 (PAR4) is a seven transmembrane G-protein coupled receptor (GPCR) activated by endogenous proteases, such as thrombin. PAR4 is involved in various pathophysiologies including cancer, inflammation, pain, and thrombosis. Although regulators of G-protein signaling (RGS) are known to modulate GPCR/Gα-mediated pathways, their specific effects on PAR4 are not fully understood at present. We previously reported that RGS proteins attenuate PAR1- and PAR2-mediated signaling through interactions with these receptors in conjunction with distinct Gα subunits. METHODS: We employed a bioluminescence resonance energy transfer technique and confocal microscopy to examine potential interactions among PAR4, RGS, and Gα subunits. The inhibitory effects of RGS proteins on PAR4-mediated downstream signaling and cancer progression were additionally investigated by using several assays including ERK phosphorylation, calcium mobilization, RhoA activity, cancer cell proliferation, and related gene expression. RESULTS: In live cells, RGS2 interacts with PAR4 in the presence of Gαq while RGS4 binding to PAR4 occurs in the presence of Gαq and Gα12/13. Co-expression of PAR4 and Gαq induced a shift in the subcellular localization of RGS2 and RGS4 from the cytoplasm to plasma membrane. Combined PAR4 and Gα12/13 expression additionally promoted translocation of RGS4 from the cytoplasm to the membrane. Both RGS2 and RGS4 abolished PAR4-activated ERK phosphorylation, calcium mobilization and RhoA activity, as well as PAR4-mediated colon cancer cell proliferation and related gene expression. CONCLUSIONS: RGS2 and RGS4 forms ternary complex with PAR4 in Gα-dependent manner and inhibits its downstream signaling. Our findings support a novel physiological function of RGS2 and RGS4 as inhibitors of PAR4-mediated signaling through selective PAR4/RGS/Gα coupling. Video Abstract.


Subject(s)
RGS Proteins/metabolism , Receptors, Thrombin/metabolism , HEK293 Cells , HT29 Cells , Humans , Protein Binding
7.
FEBS Lett ; 592(18): 3126-3138, 2018 09.
Article in English | MEDLINE | ID: mdl-30117167

ABSTRACT

Protease-activated receptor 2 (PAR2) is a G protein-coupled receptor (GPCR) activated by endogenous proteases, in particular, trypsin. Although regulators of G protein signaling (RGS) are known to inhibit GPCR/Gα-mediated signaling, their specific effects on PAR2 are poorly understood at present. Here, we use a bioluminescence resonance energy transfer technique to investigate whether RGS16 and RGS18 bind PAR2 in live cells to regulate PAR2/Gαi/o -mediated signaling. Notably, we find that RGS16 binds to PAR2 in the presence of Gαi while RGS18 does not interact with PAR2, regardless of the presence of Gα. Both RGS16 and RGS18 inhibit PAR2/Gαi/o -mediated signaling. To our knowledge, the current study is the first to highlight the effects of RGS proteins on PAR2-mediated signaling.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits/metabolism , RGS Proteins/metabolism , Receptor, PAR-2/metabolism , Animals , Bioluminescence Resonance Energy Transfer Techniques/methods , Cell Line, Tumor , Cell Membrane/metabolism , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , HEK293 Cells , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Microscopy, Confocal , Protein Binding , RGS Proteins/genetics , Receptor, PAR-2/genetics , Signal Transduction
8.
Biochem Biophys Res Commun ; 489(2): 109-115, 2017 07 22.
Article in English | MEDLINE | ID: mdl-28528976

ABSTRACT

Osteoblasts originate from mesenchymal stem cells that also differentiate into adipocytes, myoblasts, chondrocytes and fibroblasts. Osteogenic differentiation involves diverse regulatory proteins, including transcription and growth factors. Neurally differentiated embryonal carcinoma-derived protein (Necdin) has been identified as a key regulator of cell differentiation in various tissues, including neuronal, adipose, and muscular tissues; although its role in bone tissue remains to be established. Here, we investigated the potential involvement of Necdin in osteogenic differentiation. Our experiments revealed high expression of Necdin during osteoblast differentiation. Moreover, both transient and stable expression of Necdin induced osteoblast-specific markers in an osteogenic cell line through formation of a complex with melanoma-associated antigen D1 (MAGE-D1) and distal-less Homeobox 5 (Dlx5) and Runx2 promoter activation. Necdin expression was further associated with suppression of both cell proliferation and death in osteoblasts. Our results suggest that Necdin plays roles in cellular differentiation, proliferation and death in bone tissue.


Subject(s)
Cell Differentiation , Homeodomain Proteins/metabolism , Neoplasm Proteins/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Osteoblasts/metabolism , Osteogenesis , Animals , Cell Survival , Cells, Cultured , HeLa Cells , Humans , Mice , Osteoblasts/cytology
9.
J Exp Zool A Ecol Integr Physiol ; 327(10): 611-619, 2017 12.
Article in English | MEDLINE | ID: mdl-29542267

ABSTRACT

Warm temperature acclimation-associated 65-kDa protein (Wap65) is known to respond to elevated water temperatures and the corresponding gene from several fish species has been cloned. Expression of Wap65 gene is induced by various physiological stresses, such as increase in water temperature, immune response and heavy metal exposure. Two isolated Wap65 genes, Wap65-1 and Wap65-2, display distinct tissue distribution and physiological functions despite high sequence homology. In a previous study, we identified the Wap65-1 gene (kmWap65-1) from Kumgang fat minnow, Rhynchocypris kumgangensis, a small freshwater fish endemic to Korea. The kmWap65-1 gene showed sequence homology with teleost Wap65-1 and mammalian hemopexin, and was highly expressed in response to increased water temperature and bacterial lipopolysaccharide (LPS) exposure. Here, we isolated kmWap65-2 from liver tissue of Kumgang fat minnow and compared the expression profiles of both kmWap65 genes following exposure to various physiological stresses, including thermal changes, bacterial challenge, and environmental toxins. Notably, while kmWap65-1 expression was significantly increased in response to high water temperature, LPS, cadmium, and iron, kmWap65-2 displayed no alterations in expression at high water temperature. However, kmWap65-2 expression was upregulated slightly in response to LPS and highly in presence of copper, bisphenol A, and estradiol. Based on the collective findings, we propose that kmWap65-1 and kmWap65-2 are multifunctional proteins with distinct functions that could serve as useful biomarkers for assessing physiological stress and associated responses in Kumgang fat minnow.


Subject(s)
Acclimatization/physiology , Cyprinidae/physiology , Fish Proteins/metabolism , Hot Temperature , Amino Acid Sequence , Animals , Fish Proteins/genetics , Liver/metabolism , Models, Molecular , Protein Conformation
10.
Proc Natl Acad Sci U S A ; 113(39): 10974-9, 2016 09 27.
Article in English | MEDLINE | ID: mdl-27625425

ABSTRACT

In mammals, initial detection of olfactory stimuli is mediated by sensory neurons in the main olfactory epithelium (MOE) and the vomeronasal organ (VNO). The heterotrimeric GTP-binding protein Go is widely expressed in the MOE and VNO of mice. Early studies indicated that Go expression in VNO sensory neurons is critical for directing social and sexual behaviors in female mice [Oboti L, et al. (2014) BMC Biol 12:31]. However, the physiological functions of Go in the MOE have remained poorly defined. Here, we examined the role of Go in the MOE using mice lacking the α subunit of Go Development of the olfactory bulb (OB) was perturbed in mutant mice as a result of reduced neurogenesis and increased cell death. The balance between cell types of OB interneurons was altered in mutant mice, with an increase in the number of tyrosine hydroxylase-positive interneurons at the expense of calbindin-positive interneurons. Sexual behavior toward female mice and preference for female urine odors by olfactory sensory neurons in the MOE were abolished in mutant male mice. Our data suggest that Go signaling is essential for the structural and functional integrity of the MOE and for specification of OB interneurons, which in turn are required for the transmission of pheromone signals and the initiation of mating behavior with the opposite sex.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Olfactory Mucosa/metabolism , Sexual Behavior, Animal , Animals , Apoptosis/genetics , Cell Count , Cells, Cultured , GTP-Binding Protein alpha Subunits, Gi-Go/deficiency , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Gene Deletion , Interneurons/metabolism , Male , Mice , Models, Biological , Neurogenesis/genetics , Olfactory Bulb/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Tyrosine 3-Monooxygenase/metabolism , Vomeronasal Organ/metabolism
11.
Cell Signal ; 28(5): 391-400, 2016 May.
Article in English | MEDLINE | ID: mdl-26829215

ABSTRACT

Activation of seven-transmembrane-domain-possessing G protein-coupled receptors (GPCRs) by extracellular stimuli elicits intracellular responses. One class of GPCRs-protease-activated receptors (PARs)-is activated by endogenous proteases, such as thrombin and trypsin. Members of the regulator of G protein signaling (RGS) family stimulate GTP hydrolysis of G protein alpha (Gα) subunits, thereby inhibiting GPCR/Gα-mediated signaling. We previously reported that RGS2 and RGS4 inhibit PAR1/Gα-mediated signaling by interacting with PAR1 in a Gα-dependent manner. Here, employing the bioluminescence resonance energy transfer (BRET) technique, we identified RGS8 as a novel PAR1-interacting protein. Very little BRET activity was observed between PAR1-Venus (PAR1-Ven) and RGS8-Luciferase (RGS8-Luc) in the absence of Gα. However, in the presence of Gαo, BRET activity was specifically and significantly increased. This interaction was confirmed by biochemical and immunofluorescence assays. Notably, RGS8 inhibited PAR1/Gαi/o-mediated adenylyl cyclase and ERK activation, and prevented Gαo-induced neurite outgrowth and activation of Necdin protein, a downstream target of Gαo. Our findings suggest a novel function of RGS8 and reveal cellular mechanisms by which RGS8 mediates PAR1 inhibition.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , RGS Proteins/metabolism , Receptor, PAR-1/metabolism , Signal Transduction , Cell Line , Extracellular Signal-Regulated MAP Kinases/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/antagonists & inhibitors , HEK293 Cells , Humans , Receptor, PAR-1/antagonists & inhibitors
12.
J Exp Zool A Ecol Genet Physiol ; 325(1): 65-74, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26612495

ABSTRACT

Water temperature is one of the most important factors in fish physiology; thus, it is important to identify genes that respond to changes in water temperature. In this study, we identified a warm- temperature acclimation-associated 65-kDa protein (Wap65) in the Kumgang fat minnow Rhynchocypris kumgangensis, a small, cold-freshwater fish species endemic to Korea. Kumgang fat minnow Wap65-1 (kmWap65-1) was cloned using polymerase chain reaction (PCR)-based strategies, and was found to be highly homologous with teleost Wap65-1 and mammalian hemopexin, a heme-binding protein that transfers plasma heme into hepatocytes. kmWap65-1 mRNA was expressed mainly in the liver and its expression levels were significantly increased by both short- and long-term exposure to high temperature, which was evaluated by real-time quantitative PCR. Furthermore, the expression levels of kmWap65-1 were highly elevated by exposure to bacterial lipopolysaccharide. These results indicate that kmWap65-1 expression is associated with environmental stresses such as increases in water temperature and bacterial infection. J. Exp. Zool. 325A:65-74, 2016. © 2015 Wiley Periodicals, Inc.


Subject(s)
Acclimatization/genetics , Cyprinidae/physiology , Phylogeny , Proteins/genetics , Animals , Cyprinidae/genetics , Cyprinidae/metabolism , Fish Proteins , Hemopexin/genetics , Hot Temperature , Proteins/isolation & purification , Proteins/metabolism , RNA, Messenger/genetics , Sequence Analysis, DNA
13.
In Vitro Cell Dev Biol Anim ; 51(9): 890-3, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26138241

ABSTRACT

Honeybees are among the most important pollinators in nature, and honeybee-associated products are useful in various areas, including the food industry. However, honeybees may be infected by various types of pathogens. The study of honeybee-associated diseases would greatly benefit from a successful cell culture system, but although some honeybee cell culture techniques have been introduced, these methods have not yet been fully established. Here, we describe a primary cell culture method for the honeybee, Apis mellifera. We isolated, sterilized, and seeded egg cells into non-coated cell culture dishes to generate cell aggregates. After approximately 10 d, aggregates were dissociated and seeded to cell culture dishes. Cell passages were continuously performed, with sub-culturing every 3-4 d. The cells expressed non-adherent phenotypes. Their growth increased with the passage number when they were cultured in growth medium based on L-15 insect medium but not Schneider's insect medium. Finally, polymerase chain reaction confirmed that the cells originated from A. mellifera. Our results suggest that the culturing methods described herein are appropriate for isolating primary cells from honeybee eggs. These methods could thus facilitate the study of honeybee-associated pathogenesis, development, and toxicology.


Subject(s)
Bees/cytology , Primary Cell Culture/methods , Animals , Phenotype
14.
Cell Commun Signal ; 12: 39, 2014 Jul 10.
Article in English | MEDLINE | ID: mdl-25012566

ABSTRACT

BACKGROUND: Heterotrimeric GTP-binding proteins (G-proteins) play an important role in mediating signal transduction generated by neurotransmitters or hormones. Go, a member of the Gi/Go subfamily, is the most abundant G-protein found in the brain. Recently, the alpha subunit of Go (Gαo) was characterized as an inducer of neuronal differentiation. However, its underlying molecular mechanisms have remained unclear to date, since the downstream effectors of Gαo are ambiguous. RESULTS: A neurally differentiated embryonal carcinoma-derived protein (Necdin) was isolated as an interacting partner for Gαo from a mouse brain cDNA library using yeast two-hybrid screening. Interactions between the proteins were confirmed with several affinity binding assays, both in vitro and in vivo. Necdin interacted directly and preferentially with activated Gαo, compared to wild-type protein. Interestingly, Gαo did not interact with Gαi, despite high sequence homology between the two proteins. We subsequently analyzed whether Gαo modulates the cellular activities of Necdin. Notably, expression of Gαo significantly augmented Necdin-mediated cellular responses, such as proliferation and differentiation. Moreover, activation of type 1 cannabinoid receptor (CB1R), a Gi/oα-coupled receptor, augmented cell growth suppression, which was mediated by Gαo and Necdin in U87MG cells containing CB1R, Gαo, and Necdin as normal components. CONCLUSIONS: These results collectively suggest that Necdin is a candidate downstream effector for Gαo. Our findings provide novel insights into the cellular roles of Gαo and its coupled receptor.


Subject(s)
Cell Differentiation/physiology , Cell Proliferation/physiology , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Animals , Brain/metabolism , Cell Line , DNA, Complementary/genetics , DNA, Complementary/metabolism , Humans , Mice , Receptor, Cannabinoid, CB1/metabolism , Two-Hybrid System Techniques
15.
Cell Mol Biol Lett ; 19(3): 347-60, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25002257

ABSTRACT

Cannabinoids display various pharmacological activities, including tumor regression, anti-inflammatory and neuroprotective effects. To investigate the molecular mechanisms underlying the pharmacological effects of cannabinoids, we used a yeast two-hybrid system to screen a mouse brain cDNA library for proteins interacting with type 1 cannabinoid receptor (CB1R). Using the intracellular loop 3 of CB1R as bait, we identified 14-3-3ß as an interacting partner of CB1R and confirmed their interaction using affinity-binding assays. 14-3-3ß has been reported to induce a cell cycle delay at the G2/M phase. We tested the effects of cannabinoids on cell cycle progression in HeLa cells synchronized using a double-thymidine block-and-release protocol and found an increase in the population of G2/M phase cells. We further found that CB1R activation augmented the interaction of 14-3-3ß with Wee1 and Cdc25B, and promoted phosphorylation of Cdc2 at Tyr-15. These results suggest that cannabinoids induce cell cycle delay at the G2/M phase by activating 14-3-3ß.


Subject(s)
14-3-3 Proteins/metabolism , Cell Cycle/physiology , Receptor, Cannabinoid, CB1/metabolism , Signal Transduction/physiology , 14-3-3 Proteins/genetics , Animals , Cannabinoids/pharmacology , Cell Cycle/genetics , Cell Division/drug effects , Cell Division/genetics , Cell Division/physiology , Flow Cytometry , G2 Phase/drug effects , G2 Phase/genetics , G2 Phase/physiology , G2 Phase Cell Cycle Checkpoints/drug effects , G2 Phase Cell Cycle Checkpoints/genetics , G2 Phase Cell Cycle Checkpoints/physiology , HEK293 Cells , HeLa Cells , Humans , Immunoblotting , Mice , Protein Binding , Receptor, Cannabinoid, CB1/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Two-Hybrid System Techniques
16.
PLoS One ; 9(4): e95355, 2014.
Article in English | MEDLINE | ID: mdl-24743392

ABSTRACT

Protease-activated receptor 1 (PAR1) is a G-protein coupled receptor (GPCR) that is activated by natural proteases to regulate many physiological actions. We previously reported that PAR1 couples to Gi, Gq and G12 to activate linked signaling pathways. Regulators of G protein signaling (RGS) proteins serve as GTPase activating proteins to inhibit GPCR/G protein signaling. Some RGS proteins interact directly with certain GPCRs to modulate their signals, though cellular mechanisms dictating selective RGS/GPCR coupling are poorly understood. Here, using bioluminescence resonance energy transfer (BRET), we tested whether RGS2 and RGS4 bind to PAR1 in live COS-7 cells to regulate PAR1/Gα-mediated signaling. We report that PAR1 selectively interacts with either RGS2 or RGS4 in a G protein-dependent manner. Very little BRET activity is observed between PAR1-Venus (PAR1-Ven) and either RGS2-Luciferase (RGS2-Luc) or RGS4-Luc in the absence of Gα. However, in the presence of specific Gα subunits, BRET activity was markedly enhanced between PAR1-RGS2 by Gαq/11, and PAR1-RGS4 by Gαo, but not by other Gα subunits. Gαq/11-YFP/RGS2-Luc BRET activity is promoted by PAR1 and is markedly enhanced by agonist (TFLLR) stimulation. However, PAR1-Ven/RGS-Luc BRET activity was blocked by a PAR1 mutant (R205A) that eliminates PAR1-Gq/11 coupling. The purified intracellular third loop of PAR1 binds directly to purified His-RGS2 or His-RGS4. In cells, RGS2 and RGS4 inhibited PAR1/Gα-mediated calcium and MAPK/ERK signaling, respectively, but not RhoA signaling. Our findings indicate that RGS2 and RGS4 interact directly with PAR1 in Gα-dependent manner to modulate PAR1/Gα-mediated signaling, and highlight a cellular mechanism for selective GPCR/G protein/RGS coupling.


Subject(s)
RGS Proteins/metabolism , Receptor, PAR-1/metabolism , Signal Transduction , Animals , COS Cells , Chlorocebus aethiops , Humans , Protein Binding , Xenopus laevis
17.
Mol Med Rep ; 7(4): 1288-92, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23404367

ABSTRACT

Alpinia officinarum (A. officinarum), a member of the ginger family, is used in traditional medicine to treat stomach ache, cold and swelling. Previous studies have demonstrated an anticancer effect of the A. officinarum extract and its major components in several cancer cell lines. However, the molecular mechanisms underlying the activity of this extract in breast cancer cells have not been fully elucidated to date. The aim of the present study was to investigate the molecular mechanisms underlying the activity of a methanolic extract of A. officinarum, by examining its effects on the proliferation of the breast cancer cell line MCF-7. Notably, the extract inhibited MCF-7 cell proliferation in a dose- and time­dependent manner. To further elucidate the molecular mechanism, we examined whether the A. officinarum extract affected cell cycle progression in MCF-7 cells. The extract inhibited S-phase cell cycle progression by suppressing the expression levels of S-phase cell cycle regulatory proteins, including E2F1, cyclin­dependent protein kinase 2 and cyclin A. Additionally, nuclear morphology and flow cytometry with Annexin V/propidium iodide dual staining demonstrated that apoptosis was induced. Western blot analysis using antibodies against apoptosis­related proteins showed that cell death induced by the extract is mediated via caspase­ and mitochondrial­dependent pathways. These findings collectively indicate that the A. officinarum extract exerts an antiproliferative activity in MCF-7 breast cancer cells by inducing S-phase cell cycle arrest and apoptosis.


Subject(s)
Alpinia/chemistry , Breast Neoplasms/drug therapy , Cell Cycle Proteins/biosynthesis , Plant Extracts/pharmacology , Apoptosis/drug effects , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , Plant Extracts/chemistry
18.
Mol Med Rep ; 3(3): 427-32, 2010.
Article in English | MEDLINE | ID: mdl-21472257

ABSTRACT

Cirsium japonicum is a wild perennial herb that has been used as an anti-hemorrhagic, anti-hypertensive and uretic agent in traditional Chinese medicine. Recently, it was reported that C. japonicum inhibits the growth of implanted cancer cells. However, the molecular mechanisms underlying the anti-cancer properties of C. japonicum are not fully understood. In this study, we investigated the effect of a methanol extract of C. japonicum on cell growth in the human breast cancer cell line MCF-7. C. japonicum extract inhibited the cell proliferation of MCF-7 cells in a time- and dose-dependent manner, as evaluated by the MTT assay. Furthermore, C. japonicum extract induced an anti-proliferative effect by causing G1 phase cell cycle arrest and also induced apoptosis by affecting mitochondrial apoptotic events, as determined by nuclear derangement, flow cytometry and Western blot analysis. Taken together, our findings indicate that C. japonicum extract induces the inhibition of MCF-7 cell growth at both the proliferation and apoptosis levels.

19.
Mol Med Rep ; 3(6): 1041-5, 2010.
Article in English | MEDLINE | ID: mdl-21472352

ABSTRACT

Torilis japonica is a wild biennial herb and has been used as a traditional medicine for the treatment of inflammation, skin disease and impotence. Here, we studied the effects of a T. japonica extract on the proliferation of the U87MG human glioblastoma cell line. The extract inhibited cell proliferation in a dose- and time-dependent manner, as determined using the MTT assay. We next investigated the molecular mechanisms underlying its anti-proliferation properties by examining cell cycle progression and cell death. T. japonica extract induced S-phase cell cycle arrest and inhibited the expression of cell cycle-regulatory proteins, including cyclin A, cyclin-dependent protein kinase 2 and E2F1. The extract also induced apoptotic cell death as evaluated by nuclear morphology and flow cytometry using Annexin-V/PI dual staining. Furthermore, Western blot analysis showed that apoptotic cell death was mediated by both mitochondria-independent and caspase-dependent pathways. Together, our findings indicate that the T. japonica extract contains bioactive compounds with anti-cancer effects. These materials may be useful in the chemotherapy of human glioblastoma.

20.
Proc Natl Acad Sci U S A ; 103(50): 19158-63, 2006 Dec 12.
Article in English | MEDLINE | ID: mdl-17148597

ABSTRACT

G(o), a member of the G(o/i) family, is the most abundant heterotrimeric G protein in brain. Most functions of G(o) are mediated by the G(betagamma) dimer; effector(s) for its alpha-subunit have not been clearly defined. Here we report that G(oalpha) interacts directly with cAMP-dependent protein kinase (PKA) through its GTPase domain. This interaction did not inhibit the kinase function of PKA but interfered with nuclear translocation of PKA while sparing its cytosolic function. This regulatory mechanism by which G(o) bifurcates PKA signaling may provide insights into how G(o) regulates complex processes such as neuritogenesis, synaptic plasticity, and cell transformation.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Signal Transduction , Animals , Cell Line , Cell Nucleus/metabolism , Chlorocebus aethiops , Cytosol/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Humans , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL