Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 287(40): 33377-88, 2012 Sep 28.
Article in English | MEDLINE | ID: mdl-22859298

ABSTRACT

Excessive alcohol consumption is associated with most cases of chronic pancreatitis, a progressive necrotizing inflammatory disease that can result in pancreatic insufficiency due to acinar atrophy and fibrosis and an increased risk of pancreatic cancer. At a cellular level acute alcohol exposure can sensitize pancreatic acinar cells to secretagogue stimulation, resulting in dysregulation of intracellular Ca(2+) homeostasis and premature digestive enzyme activation; however, the molecular mechanisms by which ethanol exerts these toxic effects have remained undefined. In this study we identify Raf-1 kinase inhibitory protein as an essential mediator of ethanol-induced sensitization of cholecystokinin- and carbachol-regulated Ca(2+) signaling in pancreatic acinar cells. We show that exposure of rodent acinar cells to ethanol induces protein kinase C-dependent Raf-1 kinase inhibitory protein phosphorylation, sensitization of cholecystokinin-stimulated Ca(2+) signaling, and potentiation of both basal and cholecystokinin-stimulated extracellular signal-regulated kinase activation. Furthermore, we show that either suppression of Raf-1 kinase inhibitory protein expression using short hairpin RNA or gene ablation prevented the sensitizing effects of ethanol on cholecystokinin- and carbachol-stimulated Ca(2+) signaling and intracellular chymotrypsin activation in pancreatic acinar cells, suggesting that the modulation of Raf-1 inhibitory protein expression may have future therapeutic utility in the prevention or treatment of alcohol-associated pancreatitis.


Subject(s)
Phosphatidylethanolamine Binding Protein/physiology , Acinar Cells/cytology , Adenosine Triphosphate/metabolism , Animals , Binding Sites , Calcium/metabolism , Cell Line , Central Nervous System Depressants/pharmacology , Chymotrypsin/chemistry , Ethanol/pharmacology , Extracellular Matrix/metabolism , Mice , Pancreas/metabolism , Pancreatitis/chemically induced , Pancreatitis/prevention & control , Phosphatidylethanolamine Binding Protein/genetics , Phosphorylation , Protein Kinase C/metabolism , Rats , Receptors, G-Protein-Coupled/metabolism , Risk , Signal Transduction
2.
Lab Invest ; 92(3): 420-36, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22157717

ABSTRACT

Experimental data indicate that colorectal cancer cells with CD133 expression exhibit enhanced tumorigenicity over CD133-negative (CD133-) cells. We hypothesized that CD133-positive (CD133+) cells, compared with CD133-, are more tumorigenic because they are more interactive with and responsive to their stromal microenvironment. Freshly dissected and dissociated cells from a primary colon cancer were separated into carcinoma-associated fibroblasts (CAF) and the epithelial cells; the latter were further separated into CD133+ and CD133- cells using fluorescence-activated cell sorter. The CD133+ cells formed large tumors in non-obese diabetic-severe combined immunodeficient (NOD-SCID) mice, demonstrating the phenotypic cellular diversity of the original tumor, whereas CD133- cells were unable to sustain significant growth. Affymetrix gene array analyses using t-test, fold-change and multiple test correction identified candidate genes that were differentially expressed between the CD133+ vs CD133- cells. RT-PCR verified differences in expression for 30 of the 46 genes selected. Genes upregulated (+ vs - cells) included CD133 (9.3-fold) and CXCR4 (4-fold), integrin ß8 and fibroblast growth factor receptor 2. The CAF highly express the respective ligands: stromal-derived factor-1 (SDF-1), vitronectin and FGF family members, suggesting a reciprocal relationship between the CD133+ and CAF cells. SDF-1 caused an increase in intracellular calcium in cells expressing both CD133 and CXCR4, confirming functional CXCR4. The CD133+/CXCR4+ phenotype is increased to 32% when the cells are grown in suspension compared with only 9% when the cells were allowed to attach. In Matrigel 3-D culture, the CD133+/CXCR4+ group treated with SDF-1 grew more colonies compared with vehicle, as well as significantly larger colony sizes of tumor spheres. These data demonstrate proof of principle that the enhanced tumorigenic potential of CD133+, compared with CD133-, cells is due to their increased ability to interact with their neighboring CAF.


Subject(s)
Antigens, CD/metabolism , Carcinoma/metabolism , Colonic Neoplasms/metabolism , Glycoproteins/metabolism , Peptides/metabolism , AC133 Antigen , Aged , Animals , Cell Culture Techniques , Cell Transformation, Neoplastic , Cells, Cultured , Chemokine CXCL12/metabolism , Female , Fibroblasts/metabolism , Gene Expression Profiling , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Paracrine Communication , Phenotype , Receptors, CXCR4/metabolism
3.
BMC Mol Biol ; 12: 29, 2011 Jul 11.
Article in English | MEDLINE | ID: mdl-21745389

ABSTRACT

BACKGROUND: Cyclooxygenase-2 (COX-2) and the bombesin (BBS)-like peptide, gastrin-releasing peptide (GRP), have been implicated in the progression of hormone-refractory prostate cancer; however, a mechanistic link between the bioactive peptide and COX-2 expression in prostate cells has not been made. RESULTS: We report that BBS stimulates COX-2 mRNA and protein expression, and the release of prostaglandin E2 from the GRP receptor (GRPR)-positive, androgen-insensitive prostate cancer cell line, PC-3. BBS-stimulated COX-2 expression is mediated, in part, by p38MAPK and PI3 kinase (PI3K)/Akt pathways, and blocked by a GRPR antagonist. The PI3K/Akt pathway couples GRPR to the transcription factor, activator protein-1 (AP-1), and enhanced COX-2 promoter activity. Although BBS stimulates nuclear factor-kappaB (NF-κB) in PC-3, NF-κB does not regulate GRPR-mediated COX-2 expression. The p38MAPK pathway increases BBS-stimulated COX-2 expression by slowing the degradation of COX-2 mRNA. Expression of recombinant GRPR in the androgen-sensitive cell line LNCaP is sufficient to confer BBS-stimulated COX-2 expression via the p38MAPK and PI3K/Akt pathways. CONCLUSIONS: Our study establishes a mechanistic link between GRPR activation and enhanced COX-2 expression in prostate cancer cell lines, and suggests that inhibiting GRPR may, in the future, provide an effective therapeutic alternative to non-steroidal anti-inflammatory drugs for inhibiting COX-2 in patients with recurrent prostate cancer.


Subject(s)
Bombesin/metabolism , Cyclooxygenase 2/genetics , Dinoprostone/metabolism , Gene Expression Regulation, Neoplastic , Neurotransmitter Agents/metabolism , Prostatic Neoplasms/metabolism , Receptors, Bombesin/metabolism , Active Transport, Cell Nucleus , Cell Line, Tumor , Chromones/pharmacology , Cyclooxygenase 2/metabolism , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , MAP Kinase Kinase Kinases/metabolism , Male , Morpholines/pharmacology , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Prostatic Neoplasms/genetics , RNA, Messenger/genetics , Signal Transduction , Transcription Factor AP-1/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
4.
Int J Cancer ; 126(4): 864-75, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-19697327

ABSTRACT

Expression of gastrin and cholecystokinin 2 (CCK(2)) receptor splice variants (CCK(2)R and CCK(2i4sv)R) are upregulated in human colonic adenomas where they are thought to contribute to tumor growth and progression. To determine the effects of ectopic CCK(2) receptor variant expression on colonic epithelial cell growth in vitro and in vivo, we employed the non-tumorigenic colonic epithelial cell line, NCM356. Receptor expression was induced using a retroviral expression vector containing cDNAs for either CCK(2i4sv)R or CCK(2)R. RT-PCR and intracellular Ca(2+) ([Ca(2+)](i)) imaging of RIE/CCK(2)R cells treated with conditioned media (CM) from NCM356 revealed that NCM356 cells express gastrin mRNA and secrete endogenous, biologically active peptide. NCM356 cells expressing either CCK(2)R or CCK(2i4sv)R (71 and 81 fmol/mg, respectively) grew faster in vitro, and exhibited an increase in basal levels of phosphorylated ERK (pERK), compared with vector. CCK(2) receptor selective antagonist, YM022, partially inhibited the growth of both receptor-expressing NCM356 cells, but not the control cells. Inhibitors of mitogen activated protein kinase pathway (MEK/ERK) or protein kinase C (PKC) isozymes partially inhibited the elevated levels of basal pERK and in vitro growth of receptor-expressing cells. Vector-NCM356 cells did not form tumors in nude mice, whereas, either CCK(2) receptor-expressing cells formed large tumors. Autocrine activation CCK(2) receptor variants are sufficient to increase in vitro growth and tumorigenicity of non-transformed NCM356 colon epithelial cells through a pathway involving PKC and the MEK/ERK axis. These findings support the hypothesis that expression of gastrin and its receptors in human colonic adenomas contributes to tumor growth and progression.


Subject(s)
Colon/physiology , Colorectal Neoplasms/pathology , Intestinal Mucosa/physiology , Receptor, Cholecystokinin B/genetics , Adenoma/pathology , Animals , Calcium/metabolism , Carcinoma/genetics , Cell Culture Techniques/methods , Cell Division/genetics , Colon/metabolism , Colon/pathology , Colorectal Neoplasms/genetics , DNA Primers , Disease Progression , Gastrins/genetics , Gastrins/metabolism , Genetic Variation , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Mutation , Neoplasm Staging , Reverse Transcriptase Polymerase Chain Reaction
5.
J Surg Res ; 156(1): 26-31, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19631337

ABSTRACT

BACKGROUND: Breast cancers aberrantly express gastrin-releasing peptide (GRP) hormone and its cognate receptor, gastrin-releasing peptide receptor (GRP-R). Experimental evidence suggests that bombesin (BBS), the pharmacological homologue of GRP, promotes breast cancer growth and progression. The contribution of GRP-R to other poor prognostic indicators in breast cancer, such as the expression of the EGF-R family of growth factors and hormone insensitivity, is unknown. MATERIALS AND METHODS: Two estrogen receptor (ER)-negative breast cancer cell lines were used. MDA-MB-231 overexpress both EGFR and GRPR, whereas SK-BR-3 cells express EGF-R but lack GRP-R. Cellular proliferation was assessed by Coulter counter. Chemotactic migration was performed using Transwell chambers, and the migrated cells were quantified. Northern blot and real-time PCR were used to evaluate proangiogenic factor interleukin-8 (IL-8) mRNA expression. RESULTS: In MDA-MB-231 cells, GRP-R and EGF-R synergize to regulate cell migration, IL-8 expression, but not cell proliferation. In SK-BR-3 cells, ectopic expression of GRP-R was sufficient to increase migration and IL-8 mRNA. CONCLUSIONS: These data suggest relevant roles for GRP-R in ER-negative breast cancer progression. Future mechanistic studies to define the molecular role of GRP-R in breast cancer metastasis provide novel targets for the treatment of ER-negative breast cancers.


Subject(s)
Breast Neoplasms/metabolism , Cell Movement , ErbB Receptors/metabolism , Interleukin-8/metabolism , Receptors, Bombesin/metabolism , Bombesin/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Epidermal Growth Factor/pharmacology , Female , Humans , Neurotransmitter Agents/pharmacology , RNA, Messenger/metabolism , Up-Regulation/drug effects
6.
J Biol Chem ; 284(4): 2459-71, 2009 Jan 23.
Article in English | MEDLINE | ID: mdl-19028687

ABSTRACT

The protein kinase D (PKD) family of serine/threonine kinases, which can be activated by gastrointestinal hormones, consists of three distinct isoforms that modulate a variety of cellular processes including intracellular protein transport as well as constitutive and regulated secretion. Although isoform-specific functions have been identified in a variety of cell lines, the expression and function of PKD isoforms in normal, differentiated secretory tissues is unknown. Here, we demonstrate that PKD isoforms are differentially expressed in the exocrine and endocrine cells of the pancreas. Specifically, PKD3 is the predominant isoform expressed in exocrine cells of the mouse and human pancreas, whereas PKD1 and PKD2 are more abundantly expressed in the pancreatic islets. Within isolated mouse pancreatic acinar cells, PKD3 undergoes rapid membrane translocation, trans-activating phosphorylation, and kinase activation after gastrointestinal hormone or cholinergic stimulation. PKD phosphorylation in pancreatic acinar cells occurs viaaCa2+-independent, diacylglycerol- and protein kinase C-dependent mechanism. PKD phosphorylation can also be induced by physiologic concentrations of secretagogues and by in vivo stimulation of the pancreas. Furthermore, activation of PKD3 potentiates MEK/ERK/RSK (RSK, ribosomal S6 kinase) signaling and significantly enhances cholecystokinin-mediated pancreatic amylase secretion. These findings reveal a novel distinction between the exocrine and endocrine cells of the pancreas and further identify PKD3 as a signaling molecule that promotes hormone-stimulated amylase secretion.


Subject(s)
Amylases/metabolism , Hormones/pharmacology , Pancreas, Exocrine/enzymology , Pancreas, Exocrine/metabolism , Protein Kinase C/metabolism , Animals , Cell Line , Chemokines, CC/metabolism , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Mice , Organ Specificity , Pancreas, Exocrine/drug effects , Phosphorylation , Protein Kinase C/genetics , Protein Transport , Rats , Salivary Glands/enzymology , Signal Transduction
7.
Clin Cancer Res ; 12(18): 5346-55, 2006 Sep 15.
Article in English | MEDLINE | ID: mdl-17000667

ABSTRACT

PURPOSE: Neurotensin, a gut tridecapeptide, acts as a potent cellular mitogen for various colorectal and pancreatic cancers that possess high-affinity neurotensin receptors. Cytokine/chemokine proteins are increasingly recognized as important local factors that play a role in the metastasis and invasion of multiple cancers. The purpose of this study was to (a) determine the effect of neurotensin on cytokine/chemokine gene expression and cell migration in human cancer cells and (b) assess the effect of curcumin, a natural dietary product, on neurotensin-mediated processes. EXPERIMENTAL DESIGN: The human colorectal cancer, HCT116, was treated with neurotensin, with or without curcumin, and interleukin (IL)-8 expression and protein secretion was measured. Signaling pathways, which contribute to the effects of neurotensin, were assessed. Finally, the effect of curcumin on neurotensin-mediated HCT116 cell migration was analyzed. RESULTS: We show that neurotensin, acting through the native high-affinity neurotensin receptor, induced IL-8 expression in human colorectal cancer cells in a time- and dose-dependent fashion. This stimulation involves Ca2+-dependent protein kinase C, extracellular signal-regulated kinase-dependent activator protein-1, and extracellular signal-regulated kinase-independent nuclear factor-kappaB pathways. Curcumin inhibited neurotensin-mediated activator protein-1 and nuclear factor-kappaB activation and Ca2+ mobilization. Moreover, curcumin blocked neurotensin-stimulated IL-8 gene induction and protein secretion and, at a low concentration (i.e., 10 micromol/L), blocked neurotensin-stimulated colon cancer cell migration. CONCLUSIONS: Neurotensin-mediated induction of tumor cell IL-8 expression and secretion may contribute to the procarcinogenic effects of neurotensin on gastrointestinal cancers. Furthermore, a potential mechanism for the chemopreventive and chemotherapeutic effects of curcumin on colon cancers may be through the inhibition of gastrointestinal hormone (e.g., neurotensin)-induced chemokine expression and cell migration.


Subject(s)
Cell Movement/drug effects , Curcumin/pharmacology , Interleukin-8/biosynthesis , Neurotensin/pharmacology , Calcium/metabolism , Colonic Neoplasms/metabolism , Extracellular Signal-Regulated MAP Kinases/physiology , Gene Expression Regulation/drug effects , HCT116 Cells , Humans , Interleukin-8/metabolism , NF-kappa B/physiology , Protein Kinase C/metabolism , Transcription Factor AP-1/physiology , Transcriptional Activation
8.
J Biol Chem ; 280(39): 33368-73, 2005 Sep 30.
Article in English | MEDLINE | ID: mdl-16079138

ABSTRACT

The third intracellular loop domain of G protein-coupled receptors regulates their desensitization, internalization, and resensitization. Colorectal and pancreatic cancers, but not the nonmalignant tissue, express a splice variant of the cholecystokinin 2 receptor (CCK2R) called CCK(2i4sv)R that, because of intron 4 retention, contains an additional 69 amino acids within its third intracellular loop domain. This structural alteration is associated with agonist-independent activation of Src kinase (Olszewska-Pazdrak, B., Townsend, C. M., Jr., and Hellmich, M. R. (2004) J. Biol. Chem. 279, 40400-40404). The purpose of the study was to determine the roles of intron 4 retention and Src kinase on CCK(2i4sv)R desensitization, internalization, and resensitization. Gastrin1-17 (G17) binds to both CCK2R and CCK(2i4sv)R and induces intracellular Ca2+ ([Ca2+]i) increases. Agonist-induced increases in [Ca2+]i were used to assess receptor activity. Src kinase activity was inhibited by transducing cells with a retrovirus containing a dominant-negative mutant Src (A430V). The subcellular location of enhanced green fluorescent protein-tagged receptors was monitored using laser scanning confocal microscopy. Both receptor variants desensitized at the same rate; however, CCK(2i4sv)R resensitized five times faster than CCK2R. Without agonist, 80% of CCK(2i4sv)R is located in an intracellular compartment. In contrast, 80% of CCK2R was located on the plasma membrane. Treatment with inverse agonist (YM022) or expression of dominant-negative Src blocked the constitutive internalization of CCK(2i4sv)R, resulting in its accumulation on the plasma membrane. Expression of dominant-negative Src slowed the rate of CCK(2i4sv)R resensitization. Inhibition of Src did not affect G17-induced internalization of either receptor variant. Constitutive internalization of CCK(2i4sv)R increases its rate of resensitization by creating an intracellular pool of receptors that can rapidly recycle back to the plasma membrane.


Subject(s)
Alternative Splicing , Genetic Variation , Receptor, Cholecystokinin B/genetics , Receptor, Cholecystokinin B/metabolism , src-Family Kinases/metabolism , Amino Acid Sequence , Benzodiazepines/pharmacology , Calcium/metabolism , Carbocyanines , Cell Line , Cell Membrane/metabolism , Clone Cells , Fluorescent Dyes , Gastrins/metabolism , Gastrins/pharmacology , Green Fluorescent Proteins/metabolism , Hormone Antagonists/pharmacology , Humans , Introns , Kinetics , Microscopy, Confocal , Models, Biological , Protein Structure, Tertiary , Receptor, Cholecystokinin B/agonists , Receptor, Cholecystokinin B/chemistry , Retroviridae/genetics , Transduction, Genetic , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/genetics
9.
J Surg Res ; 129(2): 313-21, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16051276

ABSTRACT

BACKGROUND: In this study, the prevalence and identity of the cells expressing functional receptors for the gastrointestinal (GI) peptide hormones: gastrin, bombesin, and neurotensin in dissociated cells from 20 freshly resected human primary colorectal carcinomas were determined. MATERIALS AND METHODS: GI peptide hormone-induced increases in the concentration of free intracellular Ca(2+) ([Ca(2+)](i)) were used as an assay for the detection of functional receptors. Reverse-transcription polymerase chain reaction (RT-PCR) was performed in a subset of tumor samples. Agonist-responsive cells were identified as either of epithelial or stromal origin by immunocytochemistry with cytokeratin and vimentin antibodies, respectively. RESULTS: Overall, expression of GI peptide hormone receptors was more frequent in stromal cells when compared to epithelial cells. Of the three receptors, expression of bombesin receptor (95%) was most prevalent in vimentin-positive (stromal) cells; whereas, gastrin receptor expression by cytokeratin-positive (epithelial) cells was more common (39%). A single gastrin receptor splice variant differentially regulates [Ca(2+)](i) in a cell-type specific manner. The gastrin receptor-expression profile in the 11 colon cancer-derived cell lines did not reflect the prevalence of expression in primary human cancers. CONCLUSIONS: The Ca(2+) assay is a sensitive method for detecting functional GI peptide hormone receptor expression by colon cancer cells. Because this approach utilizes living cells, it is amenable to further functional analyses of signal transduction mechanisms at the single cell level. Importantly, our data provide a rationale for examining of the role of these GI peptide hormones and their cognate receptors in mesenchymal cell biology.


Subject(s)
Calcium/metabolism , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Immunohistochemistry/methods , Receptors, Gastrointestinal Hormone/metabolism , Bombesin/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gastrins/metabolism , Homeodomain Proteins/metabolism , Humans , In Vitro Techniques , Mesoderm/metabolism , Mesoderm/pathology , Neurotensin/metabolism , Proto-Oncogene Proteins/metabolism , Receptor, Cholecystokinin B/genetics , Receptor, Cholecystokinin B/metabolism , Receptors, Bombesin/genetics , Receptors, Bombesin/metabolism , Receptors, Gastrointestinal Hormone/genetics , Receptors, Neurotensin/genetics , Receptors, Neurotensin/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sensitivity and Specificity
10.
J Biol Chem ; 279(3): 1853-60, 2004 Jan 16.
Article in English | MEDLINE | ID: mdl-14602717

ABSTRACT

Small differences in amplitude, duration, and temporal patterns of change in the concentration of free intracellular Ca2+ ([Ca2+](i)) can profoundly affect cell physiology, altering programs of gene expression, cell proliferation, secretory activity, and cell survival. We report a novel mechanism for amplitude modulation of [Ca2+](i) that involves mitogen-activated protein kinase (MAPK). We show that epidermal growth factor (EGF) potentiates gastrin-(1-17) (G17)-stimulated Ca2+ release from intracellular Ca2+ stores through a MAPK-dependent pathway. G17 activation of the cholecystokinin/gastrin receptor (CCK(2)R), a G protein-coupled receptor, stimulates release of Ca2+ from inositol 1,4,5-triphosphate-sensitive Ca2+ stores. Pretreating rat intestinal epithelial cells expressing CCK(2)R with EGF increased the level of G17-stimulated Ca2+ release from intracellular stores. The stimulatory effect of EGF on CCK(2)R-mediated Ca2+ release requires activation of the MAPK kinase (MEK)1,2/extracellular signal-regulated kinase (ERK)1,2 pathway. Inhibition of the MEK1,2/ERK1,2 pathway by either serum starvation or treatment with selective MEK1,2 inhibitors PD98059 and U0126 or expression of a dominant-negative mutant form of MEK1 decreased the amplitude of the G17-stimulated Ca2+ release response. Activation of the MEK1,2/ERK1,2 pathway either by pretreating cells with EGF or by expression of constitutively active K-ras (K-rasV12G) or MEK1 (MEK1*) increased the amplitude of G17-stimulated Ca2+ release. Although EGF, MEK1*, and K-rasV12G activated the MEK1,2/ERK1,2 pathway, they did not increase [Ca2+](i) in the absence of G17. These data demonstrate that the activation state of the MEK1,2/ERK1,2 pathway can modulate the amplitude of the CCK(2)R-mediated Ca2+ release response and identify a novel mechanism for cross-talk between EGF receptor- and CCK(2)R-regulated signaling pathways.


Subject(s)
Calcium/metabolism , Epidermal Growth Factor/pharmacology , Mitogen-Activated Protein Kinases/physiology , Receptors, Cholecystokinin/physiology , Animals , Enzyme Activation , Gastrins/pharmacology , Humans , Phosphatidylinositol 3-Kinases/physiology , Phosphorylation , Rats
11.
Mol Pharmacol ; 64(4): 890-904, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14500746

ABSTRACT

Acute desensitization of many guanine nucleotide-binding protein-coupled receptors (GPCRs) requires receptor phosphorylation and subsequent binding of an arrestin. GPCRs are substrates for phosphorylation by several classes of kinases. Gastrin-releasing peptide receptor (GRPr) is phosphorylated by a kinase other than protein kinase C (PKC) after exposure to agonist and is also a substrate for PKC-dependent phosphorylation after treatment with 12-O-tetradecanoylphorbol 13-acetate (TPA). Using GRPr mutants, we examined receptor domains required for agonist- and TPA-induced phosphorylation of GRPr and consequences of these phosphorylation events on GRPr signaling via Gq. Agonist- and TPA-stimulated GRPr phosphorylation in cells require an intact carboxyl terminal domain (CTD). GRPr is phosphorylated in vitro by GPCR kinase 2 (GRK2) and multiple PKC isoforms. An intact DRY motif is required for agonist-stimulated phosphorylation in cells, and agonist-dependent GRK2 phosphorylation in vitro. Although GRPr CTD mutants do not show enhanced in vitro coupling to Gq relative to intact GRPr, CTD mutants have more potent Gq-dependent signaling in cells. Acute desensitization involves CTD-independent processes because desensitization can precede ligand binding in intact GRPr and CTD mutants. TPA-mediated impairment of GRPr-Gq signaling in cells also requires an intact CTD. Similar to GRK2 phosphorylation, PKC phosphorylation reduces GRPr-Gq coupling by approximately 80% in vitro. Arrestin translocation to plasma membrane requires agonist, an intact DRY motif, and GRPr phosphorylation. Therefore, agonist- and PKC-induced GRPr phosphorylation sites are in nearby regions of the receptor, and phosphorylation at both sites has similar functional consequences for Gq signaling.


Subject(s)
Protein Kinase C/metabolism , Receptors, Bombesin/metabolism , 3T3 Cells , Amino Acid Sequence , Animals , Arrestin/metabolism , Biological Transport , Bombesin/metabolism , Calcium/metabolism , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Mutation , Phosphorylation , Protein Structure, Tertiary , Signal Transduction
12.
Biol Reprod ; 66(5): 1230-6, 2002 May.
Article in English | MEDLINE | ID: mdl-11967182

ABSTRACT

Although oxytocin and its receptor have been identified in human ovary, its regulatory role in granulosa cell or corpus luteum function has not been clearly defined. To better understand oxytocin action in the human ovary, we have characterized the expression and function of oxytocin receptors in an immortalized human granulosa-lutein cell line, HGL5. Expression of oxytocin receptor mRNA was demonstrated by reverse transcriptase-polymerase chain reaction analysis, and by specific binding of an iodinated oxytocin antagonist (apparent dissociation constant of 131 +/- 0.15 pM, and a B(max) of 12 +/- 0.5 fmol/microg DNA). Receptor levels were down-regulated by serum starvation, and rapidly up-regulated by serum restoration. Stimulation of protein kinase C activity increased oxytocin receptor levels in a concentration-dependent manner. Conversely, protein kinase C inhibition blocked up-regulation of oxytocin receptors. Treatment of cells with 10 nM oxytocin resulted in a rapid, transient increase in intracellular Ca(2+), and the response was blocked by an oxytocin antagonist. Because HGL5 cells secrete progesterone and estradiol in response to agents that elevate intracellular cAMP concentrations, we studied the effect of oxytocin on steroid production. Oxytocin enhanced the effects of forskolin on progesterone production. These results suggest that oxytocin augments the activity of luteotropins in vivo. Our studies are the first to show an ovarian cell line that expresses functional oxytocin receptors. These cells can serve as a useful model for studying oxytocin signal pathways and their cross-talk with respect to progesterone synthesis. These cells also will be useful in the analysis of mechanisms of oxytocin receptor regulation, including regulation of its gene.


Subject(s)
Granulosa Cells/physiology , Lutein/metabolism , Receptors, Oxytocin/physiology , Calcium/metabolism , Cell Line , DNA/biosynthesis , DNA/genetics , Female , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Granulosa Cells/metabolism , Humans , Kinetics , Ligands , Myometrium/metabolism , Phorbol Esters/pharmacology , Progesterone/metabolism , Prostaglandins/metabolism , RNA, Messenger/biosynthesis , Radioimmunoassay , Receptors, Oxytocin/biosynthesis , Receptors, Vasopressin/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...