Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
PLoS One ; 7(7): e41708, 2012.
Article in English | MEDLINE | ID: mdl-22848573

ABSTRACT

Epidemiological studies and interventional clinical trials indicate that consumption of long chain n-3 polyunsaturated fatty acids (LC n-3 PUFA) such as docosahexaenoic acid (DHA) lengthen gestational duration. Although the mechanisms are not well understood, prostaglandins (PG) of the 2-series are known to play a role in the initiation and progress of labor. In animal studies, modest DHA provision has been shown to reduce placental and uterine PGE(2) and PGF(2α), matrix metalloproteinase (MMP)-2 and MMP-9 expression, and placental collagenase activity. However, modulation of PG biosynthesis may not account for all the effects of LC n-3 PUFAs in labor. We investigated one potential PG-independent mechanism of LC PUFA action using cultured pregnant human myometrial smooth muscle cells. Our goal was to characterize the effect of LC PUFA treatment on oxytocin signaling, a potent uterotonic hormone involved in labor. The addition of 10 µM-100 µM DHA or arachidonic acid (AA) to the culture media for 48 h resulted in dose dependent enrichment of these fatty acids in membrane lipid. DHA and AA significantly inhibited phosphatidylinositol turnover and [Ca(2+)](i) mobilization with oxytocin stimulation compared to bovine serum albumin control and equimolar oleic acid. DHA and AA significantly reduced oxytocin receptor membrane concentration without altering binding affinity or rate of receptor internalization. These findings demonstrate a role for LC n-3 PUFAs in regulation of oxytocin signaling and provide new insight into additional mechanisms pertaining to reports of dietary fish and fish oil consumption prolonging gestation.


Subject(s)
Fatty Acids, Unsaturated/chemistry , Fatty Acids, Unsaturated/pharmacology , Myocytes, Smooth Muscle/cytology , Myometrium/cytology , Oxytocin/metabolism , Receptors, Oxytocin/metabolism , Signal Transduction/drug effects , Animals , Calcium/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Culture Media/chemistry , Dose-Response Relationship, Drug , Fatty Acids, Unsaturated/metabolism , Female , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Ligands , Muscle Contraction/drug effects , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Phosphatidylinositols/metabolism , Pregnancy , Protein Transport/drug effects
2.
Biol Reprod ; 85(2): 315-26, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21565997

ABSTRACT

To explore the relationship between signal-stimulated increases in intracellular calcium ([Ca(2+)](i)) and depletion and refilling of the endoplasmic reticulum (ER) Ca(2+) stores ([Ca(2+)](L)) in human myometrial cells, we measured simultaneous changes in [Ca(2+)](i) and [Ca(2+)](L) using Fura-2 and Mag-fluo-4, respectively, in PHM1-41 immortalized and primary cells derived from pregnant myometrium and in primary cells derived from nonpregnant tissue. Signal- and extracellular Ca(2+)-dependent increases in [Ca(2+)](i) (SRCE) and ER refilling stimulated by oxytocin and cyclopiazonic acid were not inhibited by voltage-operated channel blocker nifedipine or mibefradil, inhibition of Na(+)/Ca(2+) exchange with KB-R7943, or zero extracellular Na(+) in PHM1-41 cells. Gadolinium-inhibited oxytocin- and cyclopiazonic acid-induced SRCE and slowed ER store refilling. TRPC1 mRNA knockdown specifically inhibited oxytocin-stimulated SRCE but had no statistically significant effect on ER store refilling and no effect on either parameter following cyclopiazonic acid treatment. Dominant negative STIMΔERM expression attenuated oxytocin- and thapsigargin-stimulated SRCE. Both STIM1 and ORAI1-ORAI3 mRNA knockdowns significantly attenuated oxytocin- and cyclopiazonic acid-stimulated SRCE. The data also suggest that reduction in STIM1 or ORAI1-ORAI3 mRNA can impede the rate of ER store refilling following removal of SERCA inhibition. These data provide evidence for both distinct and overlapping influences of TRPC1, STIM1, and ORAI1-ORAI3 on SRCE and ER store refilling in human myometrial cells that may contribute to the regulation of myometrial Ca(2+) dynamics. These findings have important implications for understanding the control of myometrial Ca(2+) dynamics in relation to myometrial contractile function.


Subject(s)
Calcium Channels/metabolism , Calcium Signaling/physiology , Calcium/metabolism , Endoplasmic Reticulum/metabolism , Myometrium/cytology , Calcium Channels/genetics , Cells, Cultured , Female , Gene Expression Regulation , Gene Silencing , Humans , Pregnancy , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Time Factors
3.
Reprod Sci ; 17(7): 696-704, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20457802

ABSTRACT

We have previously shown that pregnant rat myometrial plasma membrane-associated cyclic adenosine monophosphate (cAMP)-dependent protein kinase A (PKA) decreases prior to delivery, coincident with a decline in the inhibitory effect of cAMP on contractant-stimulated parameters. We now find that rat myometrial membrane-associated PKA concentrations in early to mid-pregnancy are equivalent to those in cycling rats. Following the decline associated with parturition, membrane PKA recovers within 1 to 2 days postpartum. Treatment with the antiprogestin onapristone caused a decrease in myometrial membrane PKA catalytic and regulatory subunits compared to untreated controls by 12 hours. This coincided temporally with recently reported increases in electrical and contractile activity. In unilaterally pregnant rats, the decline in plasma membrane PKA was observed in both nonpregnant and pregnant horns but was more rapid in the pregnant horns. These data indicate that the myometrial plasma membrane PKA pattern before and during most of pregnancy is not consistent with progesterone exerting a primary influence on PKA membrane localization. Rather, the fall in membrane PKA associated with parturition may contribute to or be influenced by the increased contractile and electrical activity of labor that is a consequence of the loss of progesterone influence and is not absolutely dependent on the presence of fetuses.


Subject(s)
Cell Membrane/enzymology , Cyclic AMP-Dependent Protein Kinases/metabolism , Myometrium/enzymology , Parturition/metabolism , Animals , Estrous Cycle/physiology , Female , Pregnancy , Progesterone/metabolism , Rats , Rats, Sprague-Dawley
4.
Endocrinology ; 151(1): 406-16, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19940041

ABSTRACT

An increase in intracellular Ca(2+) ([Ca(2+)](i)) as a result of release of Ca(2+) from intracellular stores or influx of extracellular Ca(2+) contributes to the regulation of smooth muscle contractile activity. Human uterine smooth muscle cells exhibit receptor-, store-, and diacylglycerol (OAG)-mediated extracellular Ca(2+)-dependent increases in [Ca(2+)](i) (SRCE) and express canonical transient receptor potential-like channels (TRPC) mRNAs (predominantly TRPC1, -4, and -6) that have been implicated in SRCE. To determine the role of TRPC6 in human myometrial SRCE, short hairpin RNA constructs were designed that effectively targeted a TRPC6 mRNA reporter for degradation. One sequence was used to produce an adenovirus construct (TC6sh1). TC6sh1 reduced TRPC6 mRNA but not TRPC1, -3, -4, -5, or -7 mRNAs in PHM1-41 myometrial cells. Compared with uninfected cells or cells infected with empty vector, the increase in [Ca(2+)](i) in response to OAG was specifically inhibited by TC6sh1, whereas SRCE responses elicited by either oxytocin or thapsigargin were not changed. Similar findings were observed in primary pregnant human myometrial cells. When PHM1-41 cells were activated by OAG in the absence of extracellular Na(+), the increase in [Ca(2+)](i) was partially reduced. Furthermore, pretreatment with nifedipine, an L-type calcium channel blocker, also partially reduced the OAG-induced [Ca(2+)](i) increase. Similar effects were observed in primary human myometrial cells. These findings suggest that OAG activates channels containing TRPC6 in myometrial cells and that these channels act via both enhanced Na(+) entry coupled to activation of voltage-dependent Ca(2+) entry channels and a nifedipine-independent Ca(2+) entry mechanism to promote elevation of intracellular Ca(2+).


Subject(s)
Calcium/metabolism , Diglycerides/pharmacology , Myometrium/drug effects , TRPC Cation Channels/genetics , Calcium Signaling/drug effects , Cell Culture Techniques , Cells, Cultured , Down-Regulation/drug effects , Down-Regulation/genetics , Down-Regulation/physiology , Efficiency , Female , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Myometrium/metabolism , Pregnancy , RNA, Small Interfering/pharmacology , Sodium/metabolism , TRPC Cation Channels/antagonists & inhibitors , TRPC Cation Channels/metabolism , TRPC Cation Channels/physiology , TRPC6 Cation Channel
5.
Cell Calcium ; 46(1): 73-84, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19523685

ABSTRACT

Canonical transient receptor potential (TRPC) proteins may play a role in regulating changes in intracellular calcium ([Ca(2+)](i)). Human myometrium expresses TRPC4, TRPC1 and TRPC6 mRNAs in greatest relative abundance. Contributions of TRPC4 to increases in [Ca(2+)](i) were assessed in PHM1-41 and primary human uterine smooth muscle (UtSMC) cells using short hairpin RNAs (shRNAs). Based on a reporter assay screen, one shRNA was selected to construct an adenoviral expression vector (TC4sh1). TC4sh1 induced both mRNA and protein TRPC4 knockdown in PHM1-41 cells without affecting expression of other TRPCs. Signal-regulated Ca(2+) entry (SRCE), defined as a stimulus- and extracellular Ca(2+)-dependent increase in [Ca(2+)](i), was measured in PHM1-41 cells treated with oxytocin (G-protein coupled receptor (GPCR)-stimulated), thapsigargin (store depletion-stimulated), and OAG (diacylglycerol-stimulated), using Fura-2. Cells infected with TC4sh1 exhibited attenuated oxytocin-, ATP- and PGF2alpha-mediated SRCE, but no change in thapsigargin- or OAG-stimulated SRCE. Similar results were obtained in primary uterine smooth muscle cells. Additionally, cells expressing TC4sh1 exhibited a significantly smaller increase in channel activity in response to oxytocin administration than did cells infected with empty virus. These data show that, in human myometrial cells, knockdown of endogenous TRPC4 specifically attenuates GPCR-stimulated, but not thapsigargin- or OAG-stimulated extracellular calcium-dependent increases in [Ca(2+)](i). These data imply that, in this cellular context, the mechanisms regulating extracellular Ca(2+)-dependent increases in [Ca(2+)](i) are differentially affected by different signaling pathways.


Subject(s)
Calcium/metabolism , Myometrium/metabolism , Receptors, G-Protein-Coupled/metabolism , TRPC Cation Channels/metabolism , Calcium/antagonists & inhibitors , Calcium Signaling/drug effects , Cell Line , Cells, Cultured , Female , Humans , Muscle, Smooth/metabolism , Oxytocin/pharmacology , RNA Interference/drug effects , Receptors, G-Protein-Coupled/antagonists & inhibitors , TRPC Cation Channels/antagonists & inhibitors , TRPC Cation Channels/genetics , Thapsigargin/pharmacology
6.
Biol Reprod ; 78(6): 1007-17, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18322273

ABSTRACT

Phospholipase CB3 (PLCB3) serine(1105) (S(1105)), a substrate for multiple protein kinases, represents a potential point of convergence of several signaling pathways in the myometrium. To explore this hypothesis, the regulation of PLCB3-S(1105) phosphorylation (P-S(1105)) was studied in immortalized and primary human myometrial cells. 8-[4-chlorophenylthio] (CPT)-cAMP and calcitonin gene-related peptide (CALCA) transiently increased P-S(1105). Relaxin also stimulated P-S(1105); this effect was partially blocked by the protein kinase A (PRKA) inhibitor, Rp-8-CPT-cAMPS. Oxytocin, which stimulates Galphaq-mediated pathways, also rapidly increased P-S(1105), as did prostaglandin F2alpha and ATP. Oxytocin-stimulated phosphorylation was blocked by protein kinase C (PRKC) inhibitor Go6976 and by pretreatment overnight with a phorbol ester. Cypermethrin, a PP2B phosphatase inhibitor, but not okadaic acid, a PP1/PP2A inhibitor, prolonged the effect of CALCA on P-S(1105), whereas the reverse was the case for the oxytocin-stimulated increase in P-S(1105). PLCB3 was the predominant PLC isoform expressed in the myometrial cells and PLCB3 short hairpin RNA constructs significantly attenuated oxytocin-stimulated increases in intracellular calcium. oxytocin-induced phosphatidylinositol (PI) turnover was inhibited by CPT-cAMP and okadaic acid, but was enhanced by pretreatment with Go6976. CPT-cAMP inhibited oxytocin-stimulated PI turnover in the presence of overexpressed PLCB3, but not overexpressed PLCB3-S(1105)A. These data demonstrate that both negative crosstalk from the cAMP/PRKA pathway and a negative feedback loop in the oxytocin/G protein/PLCB pathway involving PRKC operate in myometrial cells and suggest that different protein phosphatases predominate in mediating P-S(1105) dephosphorylation in these pathways. The integration of multiple signal components at the level of PLCB3 may be important to its function in the myometrium.


Subject(s)
Myometrium/enzymology , Phospholipase C beta/metabolism , Amino Acid Substitution , Base Sequence , Binding Sites/genetics , Calcitonin/pharmacology , Calcitonin Gene-Related Peptide , Cell Line , Cyclic AMP-Dependent Protein Kinases/metabolism , DNA Primers/genetics , Feedback, Physiological , Female , Humans , Mutagenesis, Site-Directed , Myometrium/cytology , Myometrium/drug effects , Oxytocin/pharmacology , Phospholipase C beta/chemistry , Phospholipase C beta/genetics , Phosphorylation , Protein Kinase C/metabolism , Protein Precursors/pharmacology , RNA Interference , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serine/chemistry , Signal Transduction
7.
Semin Cell Dev Biol ; 18(3): 305-14, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17627855

ABSTRACT

Understanding the basis for the control of myometrial contractant and relaxant signaling pathways is important to understanding how to manage myometrial contractions. Signaling pathways are influenced by the level of expression of the signals and signal pathway components, the location of these components in the appropriate subcellular environment, and covalent modification. Crosstalk between these pathways regulates the effectiveness of signal transduction and represents an important way by which hormones can regulate phenotype. This review deals primarily with signaling pathways that control Ca2+ entry and intracellular release, as well as the interplay between these pathways.


Subject(s)
Calcium Signaling , Hormones/metabolism , Myometrium/metabolism , Signal Transduction , Uterine Contraction/metabolism , Animals , Female , Humans , Ion Channels/metabolism , Pregnancy
9.
Am J Physiol Endocrinol Metab ; 292(4): E977-84, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17148753

ABSTRACT

Previously, residue K6.30 in the COOH-terminal region of the third intracellular domain (3iC) of the oxytocin (OT) receptor (OTR) was identified as important for receptor function leading to phospholipase C activation in both OTR and the vasopressin V(2) receptor (V(2)R) chimera V(2)ROTR3iC. Substitution of either A6.28K or V6.30K in wild-type V(2)R did not recapitulate the increase in phosphatidylinositide (PI) turnover observed in V(2)ROTR3iC. Hence, the role of K6.30 may be context-specific. Deletion of two NH(2)-terminal OTR3iC segments in the V(2)ROTR3iC chimera did not diminish vasopressin-stimulated PI turnover, whereas deletion of RVSSVKL (residues 6.19-6.25) reduced receptor expression. Deletion of this sequence in wild-type OTR reduced expression by 50% without affecting affinity for [(3)H]OT. This OTR mutant was unable to activate PI turnover or extracellular signal-regulated kinase 1/2 phosphorylation. The effects of alanine substitution for individual residues in RVSSVKL indicated differential importance for OTR function. The R6.19A substitution lost high-affinity sites for [(3)H]OT and the ability to stimulate PI turnover. Affinity for [(3)H]OT and membrane expression was not affected by any other substitutions. OTR-V6.20A and OTR-K6.24A mutants functioned as well as wild-type OTR, whereas OTR S6.21A, S6.22A, and V6.23A mutants exhibited impaired abilities to activate PI turnover (20-40% of OTR), and the OTR-L6.25A mutant exhibited constitutive activity. In conclusion, specific amino acids in the RVSSVKL segment in the COOH-terminal region of the third intracellular domain of OTR influence the ability of OTR to activate G protein-mediated actions.


Subject(s)
Intracellular Membranes/metabolism , Receptors, Oxytocin/genetics , Receptors, Oxytocin/metabolism , Alanine , Amino Acid Sequence , Amino Acid Substitution , Arginine , Cell Line , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Deletion , Humans , Molecular Sequence Data , Mutation , Phosphatidylinositols/metabolism , Phosphorylation , Protein Structure, Tertiary , Receptors, Vasopressin/genetics , Recombinant Fusion Proteins/metabolism , Vasopressins/pharmacology
10.
Pharmacol Rev ; 58(1): 7-31, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16507880

ABSTRACT

Although the hormone relaxin was discovered 80 years ago, only in the past 5 years have the receptors for relaxin and three other receptors that respond to related peptides been identified with all four receptors being G-protein-coupled receptors. In this review it is suggested that the receptors for relaxin (LGR7) and those for the related peptides insulin-like peptide 3 (LGR8), relaxin-3 (GPCR135), and insulin-like peptide 5 (LGPCR142) be named the relaxin family peptide receptors 1 through 4 (RXFP1-4). RXFP1 and RXFP2 are leucine-rich repeat-containing G-protein-coupled receptors with complex binding characteristics involving both the large ectodomain and the transmembrane loops. RXFP1 activates adenylate cyclase, protein kinase A, protein kinase C, phosphatidylinositol 3-kinase, and extracellular signaling regulated kinase (Erk1/2) and also interacts with nitric oxide signaling. RXFP2 activates adenylate cyclase in recombinant systems, but physiological responses are sensitive to pertussis toxin. RXFP3 and RXFP4 resemble more conventional peptide liganded receptors and both inhibit adenylate cyclase, and in addition RXFP3 activates Erk1/2 signaling. Physiological studies and examination of the phenotypes of transgenic mice have established that relaxin has roles as a reproductive hormone involved in uterine relaxation (some species), reproductive tissue growth, and collagen remodeling but also in the cardiovascular and renal systems and in the brain. The connective tissue remodeling properties of relaxin acting at RXFP1 receptors have potential for the development of agents effective for the treatment of cardiac and renal fibrosis, asthma, and scleroderma and for orthodontic remodelling. Agents acting at RXFP2 receptors may be useful for the treatment of cryptorchidism and infertility, whereas antagonists may be used as contraceptives. The brain distribution of RXFP3 receptors suggests that actions at these receptors have the potential for the development of antianxiety and antiobesity drugs.


Subject(s)
Receptors, G-Protein-Coupled/classification , Receptors, Peptide/classification , Relaxin/classification , Amino Acid Sequence , Animals , Humans , Molecular Sequence Data , Peptides/chemistry , Peptides/classification , Peptides/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Peptide/genetics , Receptors, Peptide/metabolism , Relaxin/chemistry , Relaxin/metabolism , Structure-Activity Relationship , Terminology as Topic
11.
J Soc Gynecol Investig ; 13(3): 217-25, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16527499

ABSTRACT

OBJECTIVE: Cation channels comprised of transient receptor potential (TrpC) proteins may play a role in signal-regulated calcium entry and calcium homeostasis in myometrium. The objective of this study was to determine the relative abundance of specific TrpC mRNAs expressed in human myometrium and determine if TrpC mRNA and protein concentrations differ in fundal myometrium before and after the onset of labor. METHODS: A quantitative real-time polymerase chain reaction (Q-RT-PCR) procedure was developed for determining the concentration of TrpC mRNA expression in immortalized and primary human myometrial cells and myometrial fundus tissues from patients before and after the onset of labor. The corresponding TrpC proteins were detected by Western blot analysis and immunohistochemistry. RESULTS: hTrpC1, 3, 4, 5, 6, and 7 mRNAs were expressed in two lines of immortalized human myometrial cells and in primary human myocytes. In all of these cells, hTrpC1 and hTrpC4 mRNAs were the most abundant, followed by hTrpC6. A similar distribution was observed in fundal myometrium samples from patients before and after the onset of labor. hTrpC4 mRNA was significantly lower after the onset of labor; there were no significant changes in the concentrations of other TrpC mRNAs. Immunohistochemistry identified hTrpC1, 3, 4, and 6 proteins in myometrial smooth muscle cells. Western blot analysis of myometrial membranes demonstrated no statistically significant changes in hTrpC1, 3, 4, and 6 proteins between samples collected before and after the onset of labor. CONCLUSIONS: We have demonstrated that hTrpC1 and hTrpC4 are the most abundant TrpC mRNAs in human myometrium, with TrpC6 being the next most abundant. There was no increase in TrpC mRNA or protein in fundal myometrium with the onset of labor. Nonetheless, these isoforms may play significant roles in signal regulated calcium entry in human myometrium.


Subject(s)
Calcium/metabolism , Labor, Obstetric/metabolism , Myometrium/metabolism , TRPC Cation Channels/biosynthesis , Cell Culture Techniques , Female , Gene Expression Profiling , Humans , Immunoblotting , Pregnancy , Protein Isoforms , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , TRPC Cation Channels/physiology , TRPC6 Cation Channel
12.
Biol Reprod ; 74(4): 699-705, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16371591

ABSTRACT

The MAF (proto-)oncogene family of basic-leucine zipper transcription factors plays crucial roles in the control of mammalian gene expression and development. Here we analyzed the regulation of the human MAFF gene, coding for a small MAF transcription factor, in uterine smooth muscle cells. We found that MAFF transcript levels are induced by proinflammatory cytokines in PHM1-31 myometrial cells. We observed an important induction by interleukin 1 beta (IL1B) and a weaker upregulation by tumor necrosis factor (TNF), whereas interleukin 6 (IL6) treatment had no effect. Time course experiments revealed a rapid induction of MAFF transcripts within 30 min following IL1B treatment. The presence of actinomycin D inhibited the upregulation, suggesting that regulation of MAFF mRNA levels occurs at the transcriptional level. We generated a MAFF-specific antiserum and determined that MAFF protein was also induced by TNF and IL1B in PHM1-31 cells. In contrast, it was particularly interesting that the transcript and protein levels of the highly homologous MAFG and MAFK genes are not modulated by these cytokines. Our results suggest a possible specific role for MAFF in proinflammatory cytokine-mediated control of myometrial gene expression and provide the first link between a small MAF transcription factor and the inflammatory response.


Subject(s)
Cytokines/physiology , MafF Transcription Factor/metabolism , Myometrium/metabolism , Cells, Cultured , Female , Humans , Interleukin-1/physiology , Interleukin-6/physiology , Maf Transcription Factors, Small/metabolism , Proto-Oncogene Mas , Transcription, Genetic , Tumor Necrosis Factors/physiology , Up-Regulation
13.
J Soc Gynecol Investig ; 12(7): 479-87, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16202924

ABSTRACT

Cellular mechanisms regulating myometrial intracellular free calcium (Ca2+(i)) are addressed in this review, with emphasis on G-protein-coupled receptor pathways. An increase in myometrial Ca2+(i) results in phosphorylation of myosin light chain, an increase in myosin adenosine monophosphatase (ATPase) activity and contraction. Dephosphorylation of myosin light chain and a decline in Ca2+(i) are associated with relaxation. Increases in Ca2+(i) are controlled by multiple signaling pathways, including receptor-mediated activation of phospholipase Cbeta (PLCbeta), leading to release of Ca2+ from intracellular stores. Ca2+ also enters myometrial cells through plasma membrane Ca2+ channels. Conversely, adenosine triphosphate (ATP)-dependent Ca2+ pumps lower Ca2+(i) concentrations and potassium channels promote hyperpolarization that can decrease Ca2+ entry. Receptor-coupled pathways that promote uterine relaxation primarily involve activation of cyclic adenosine monophosphate (cAMP)- or cyclic guanosine monophosphate (cGMP)-stimulated protein kinases that phosphorylate proteins regulating Ca2+ homeostasis. cAMP has inhibitory effects on myometrial contractile activity, agonist-stimulated phosphatidylinositide turnover and increases in Ca2+(i). Some of these effects require association of protein kinase A (PKA) with a plasma membrane-associated A-kinase-anchoring-protein (AKAP). Near term in the rat, there is a decline in the plasma membrane localization of PKA associated with this anchoring protein. This correlates with changes in the regulation of signaling pathways controlling Ca2+(i). L-type voltage-operated Ca2+ entry is an important regulator of myometrial contraction. In addition, putative signal-regulated or capacitative Ca2+ channel proteins, TrpCs, are expressed in myometrium, and signal-regulated Ca2+ entry is observed in human myometrial cells. This Ca2+ entry mechanism may play a significant role in the control of myometrial Ca2+(i) dynamics and myometrial contraction. The regulation of myometrial Ca2+(i) is complex. Understanding the mechanisms involved may lead to design of tocolytics that target multiple pathways and achieve improved suppression of premature labor.


Subject(s)
Calcium/pharmacokinetics , Myometrium/physiology , Pregnancy/physiology , Receptors, G-Protein-Coupled/physiology , Adult , Cyclic AMP-Dependent Protein Kinases/metabolism , Female , Humans , Signal Transduction , Tocolysis/methods , Type C Phospholipases/metabolism
14.
Ann N Y Acad Sci ; 1041: 61-76, 2005 May.
Article in English | MEDLINE | ID: mdl-15956688

ABSTRACT

Recent studies have identified four receptors that are the physiological targets for relaxin family peptides. All are class I (rhodopsin like) G-protein-coupled receptors with LGR7 (RXFP1) and LGR8 (RXFP2) being type C leucine-rich repeat-containing receptors, whereas GPCR135 (RXFP3) and GPCR142 (RXFP4) resemble receptors that respond to small peptides such as somatostatin and angiotensin II. The cognate ligands for the receptors have been identified: relaxin for RXFP1; INSL3 for RXFP2; relaxin 3 for RXFP3 and INSL5 for RXFP4. RXFP1 and RXFP2 receptors produce increases in intracellular cAMP levels upon stimulation, although the response is complex and contains a component sensitive to PI-3-kinase inhibitors. There is also evidence that RXFP1 can activate Erk1/2 and nitric oxide synthase, and relaxin has been reported to enter cells and activate glucocorticoid receptors. In contrast, RXFP3 and RXFP4 couple to Gi by a pertussis toxin-sensitive mechanism to cause inhibition of cAMP production. Now that the receptors for relaxin family peptides and their cognate ligands have been identified, we suggest a nomenclature for both the peptides and the receptors that we hope will be helpful to researchers in this rapidly advancing field.


Subject(s)
Receptors, Peptide/metabolism , Relaxin/classification , Relaxin/metabolism , Animals , Gene Expression , Humans , Ligands , Receptors, G-Protein-Coupled , Receptors, Peptide/agonists , Receptors, Peptide/antagonists & inhibitors , Receptors, Peptide/genetics , Signal Transduction
15.
Ann N Y Acad Sci ; 1041: 300-4, 2005 May.
Article in English | MEDLINE | ID: mdl-15956722

ABSTRACT

Relaxin exhibits pleiotropic effects on reproductive and nonreproductive tissues; the signaling mechanisms underlying these functions are still not well understood. Activation of protein kinase A and several other signal-regulated protein kinases results in the phosphorylation of phospholipase C (PLC)-beta3 and inhibit Galpha(q)-stimulated PLC activity. Therefore, PLCbeta3 may be targeted by both contractant and relaxant signaling pathways in myometrium and play a critical role in the balance between them. PHM1 cells express mRNA for relaxin receptor LGR7, and relaxin inhibits oxytocin-stimulated PLC activity in these cells. Thus, this model system may be useful in delineating signaling pathways used by relaxin. Here, we present evidence that relaxin stimulates phosphorylation of PLCbeta3 in PHM1 cells.


Subject(s)
Isoenzymes/metabolism , Myometrium/enzymology , Relaxin/pharmacology , Signal Transduction/drug effects , Type C Phospholipases/metabolism , Amino Acid Sequence , Animals , Cell Line , Female , Humans , Isoenzymes/chemistry , Models, Biological , Molecular Sequence Data , Myometrium/drug effects , Myometrium/metabolism , Phospholipase C beta , Phosphorylation/drug effects , Sequence Alignment , Type C Phospholipases/chemistry
16.
J Soc Gynecol Investig ; 12(6): 421-7, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15914039

ABSTRACT

OBJECTIVE: We have previously shown that the association of protein kinase A (PKA) with purified myometrial plasma membrane declined at the end of pregnancy in the rat. This study was designed to determine if a similar decline in PKA occurred in pregnant human myometrium. METHODS: Myometrial plasma membranes were isolated from lower uterine segment tissues from not-in-labor (NIL) and in-labor (IL) patients undergoing cesarean delivery. Membrane proteins were subjected to Western blot analysis to detect PKA-catalytic (PKA-cat) and PKA-regulatory (PKA-reg) subunits, the PKA binding protein A-kinase anchoring protein 79 (AKAP79), protein phosphatase 2B (PP2B), and Galphaq, a guanosine triphosphate (GTP)-binding protein. Protein levels were expressed relative to caveolin-1, which was invariant between the two groups. RESULTS: The amount of PKA-cat, PKA-reg, AKAP79, and PP2B in plasma membranes from myometrium of women in early labor decreased significantly compared with that in tissues from women not in labor. In contrast, Galphaq did not change. All proteins were localized to myometrial smooth muscle cells by immunohistochemistry. CONCLUSIONS: Expression of PKA, PP2B, and AKAP79 is consistent with the presence of a functional AKAP-mediated signaling complex in pregnant human myometrial membranes. A small but significant decrease in PKA, AKAP79, and PP2B in myometrial tissues from women in labor may contribute to a decrease in negative feedback on and enhancement of contractant signals at term.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/biosynthesis , Myometrium/physiology , Pregnancy/physiology , A Kinase Anchor Proteins , Adaptor Proteins, Signal Transducing/biosynthesis , Animals , Calcineurin/biosynthesis , Cell Membrane/physiology , Cyclic AMP-Dependent Protein Kinases/analysis , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Humans , Labor, Obstetric/physiology , Pregnancy, Animal/physiology , Rats , Signal Transduction , Uterine Contraction/physiology
17.
Circulation ; 110(12): 1612-9, 2004 Sep 21.
Article in English | MEDLINE | ID: mdl-15381659

ABSTRACT

BACKGROUND: Preeclampsia is a serious disorder of pregnancy characterized by hypertension, proteinuria, edema, and coagulation and vascular abnormalities. At the cellular level, abnormalities include increased calcium concentration in platelets, lymphocytes, and erythrocytes. Recent studies have shown that antibodies directed against angiotensin II type I (AT1) receptors are also highly associated with preeclampsia. METHODS AND RESULTS: We tested the hypothesis that AT1 receptor-agonistic antibodies (AT1-AAs) could activate AT1 receptors, leading to an increased intracellular concentration of free calcium and to downstream activation of Ca2+ signaling pathways. Sera of 30 pregnant patients, 16 diagnosed with severe preeclampsia and 14 normotensive, were examined for the presence of IgG capable of stimulating intracellular Ca2+ mobilization. IgG from all preeclamptic patients activated AT1 receptors and increased intracellular free calcium. In contrast, none of the normotensive individuals had IgG capable of activating AT1 receptors. The specific mobilization of intracellular Ca2+ by AT1-AAs was blocked by losartan, an AT1 receptor antagonist, and by a 7-amino-acid peptide that corresponds to a portion of the second extracellular loop of the AT1 receptor. In addition, we have shown that AT1-AA-stimulated mobilization of intracellular Ca2+ results in the activation of the transcription factor, nuclear factor of activated T cells. CONCLUSIONS: These results suggest that maternal antibodies capable of activating AT1 receptors are likely to account for increased intracellular free Ca2+ concentrations and changes in gene expression associated with preeclampsia.


Subject(s)
Autoantibodies/pharmacology , Calcium Signaling/drug effects , Immunoglobulin G/pharmacology , Pre-Eclampsia/immunology , Receptor, Angiotensin, Type 1/immunology , Adult , Animals , Autoantibodies/immunology , Autoantibodies/isolation & purification , Autoantigens/immunology , CHO Cells/drug effects , Cricetinae , DNA-Binding Proteins/genetics , Dose-Response Relationship, Immunologic , Epitopes/immunology , Female , Gene Expression Regulation/drug effects , Genes, Reporter , Humans , Immunoglobulin G/immunology , Immunoglobulin G/isolation & purification , Luciferases, Renilla/biosynthesis , Luciferases, Renilla/genetics , NFATC Transcription Factors , Nuclear Proteins/genetics , Peptide Fragments/immunology , Pregnancy , Rats , Receptor, Angiotensin, Type 1/agonists , Receptor, Angiotensin, Type 1/genetics , Transcription Factors/genetics , Transcription, Genetic/drug effects , Transfection
18.
Cell Calcium ; 36(2): 157-64, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15193863

ABSTRACT

Stimulation of G-protein coupled membrane receptors linked to phospholipase C results in production of the second messengers diacylglycerol and inositol-1,4,5-trisphosphate (IP3). IP3 releases Ca2+ from the endoplasmic reticulum, which triggers increased Ca2+ influx across the plasma membrane, so-called capacitative calcium entry. DAG can also activate plasma membrane calcium-permeable channels but the mechanism is still not fully understood. In the pregnant human myometrial cell line PHM1 and in primary myometrial cells, 1-oleoyl-2-acetyl-sn-glycerol (OAG), a membrane-permeant analogue of diacylglycerol, induced variable oscillatory patterns of intracellular free Ca2+. Similar behavior was seen with Sr2+ entry. The Ca2+ oscillations were not blocked by a broad spectrum of protein kinase C inhibitors, including chelerytrine, bisindolylmaleimide I and calphostin C, and were enhanced and prolonged by RHC-80267, an inhibitor of diacylglycerol lipase. The OAG-induced oscillatory response was not dependent on Ca2+ release from the endoplasmic reticulum but required extracellular Ca2+. Our results indicate that diacylglycerol directly activates cation channels in PHM1 and primary myometrial cells and promotes intracellular Ca2+ oscillations by actions independent of intracellular Ca2+ -ATPase activity and protein kinase C involvement.


Subject(s)
Calcium/metabolism , Diglycerides/pharmacology , Myometrium/drug effects , Diglycerides/metabolism , Female , Humans , Lipoprotein Lipase/antagonists & inhibitors , Myometrium/metabolism , Protein Kinase C/antagonists & inhibitors , Time Factors
19.
Biochemistry ; 43(12): 3490-8, 2004 Mar 30.
Article in English | MEDLINE | ID: mdl-15035619

ABSTRACT

Oxytocin receptor (OTR) activates the GTP-binding protein Galpha(q). To investigate whether the N-terminal region of the fourth intracellular domain of this receptor, which forms putative helix 8, plays a role in coupling, its hydrophilic residues (H7.59, H7.62, E7.63, Q7.66, and R7.67) were mutated individually to alanine. In COSM6 cells, these mutants were expressed at equivalent concentrations, but at lower concentrations than OTR. Alanine substitution for H7.62 or Q7.66 did not substantially affect the affinity for OT (K(d) = 0.63 and 0.48 nM, respectively, vs 0.52 nM for the wild type), whereas substitution for H7.59, E7.63, or R7.67 reduced the affinity 5-6-fold. When expressed at equal concentrations, OTR-H7.62/A and OTR-Q7.66/A stimulated phosphatidylinositide turnover as well as OTR, whereas OTR-H7.59/A, OTR-E7.63/A, and OTR-R7.67/A exhibited an impaired ability to respond to OT. Therefore, residues H7.59, E7.63, and R7.67 within the putative hydrophilic interface appeared to influence both the OTR conformation and Galpha(q) coupling. To explore this further, five multiple alanine substitution mutants were constructed. Alanine modification at H7.62 and Q7.66 did not substantially affect the affinity for OT (K(d) = 0.75 nM), whereas any combination of alanine substitutions for H7.59, E7.63, and R7.67 produced mutant receptors that lost high-affinity ligand binding. While OTR-(H7.62,Q7.66)/A exhibited PLC activation equivalent to that of OTR, receptors with two or more changes in H7.59, E7.63, and R7.67 lost the ability to respond to OT in a dose-dependent manner. Five residues (L7.60, F7.61, L7.64, V7.65, and F7.68) in the opposite hydrophobic interface were also mutated to alanine. None of these substitutions affected ligand binding; only OTR-(L7.60,F7.61)/A had a somewhat weaker ability to activate PLC. These data are consistent with the prediction that these residues lie within an amphipathic alpha-helix and emphasize the importance of this hydrophilic interface, and particularly of H7.59, E7.63, and R7.67, in OTR function.


Subject(s)
Receptors, Oxytocin/chemistry , Receptors, Oxytocin/physiology , Alanine/genetics , Amino Acid Sequence , Amino Acid Substitution/genetics , Animals , Arginine/genetics , COS Cells , Cattle , Glutamic Acid/genetics , Histidine/genetics , Humans , Hydrophobic and Hydrophilic Interactions , Molecular Sequence Data , Oxytocin/physiology , Protein Binding/genetics , Protein Structure, Secondary/genetics , Rats , Receptors, Oxytocin/genetics , Receptors, Vasopressin/chemistry , Signal Transduction/genetics , Transfection
20.
J Clin Endocrinol Metab ; 89(2): 1010-3, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14764828

ABSTRACT

We tested the hypothesis that prostaglandin (PGs), PGE2, and PGF2 alpha, stimulate labor and delivery in women, in part, by inducing functional progesterone withdrawal in myometrial cells by increasing the progesterone receptor (PR)-A/PR-B expression ration. PHM1-31 cells (an immortal pregnant human myometrial cell line) were exposed to PGE2, PGF2 alpha, cyclic-8-bromoadenosine monophosphate (8-Br-cAMP) and phorbol 12-myristate 13-acetate (PMA) at various concentrations for 24h. Effects on PR-A and PR-B expression were then assessed by quantitative RT-PCR. PGF2 alpha dose dependently increased PR-A mRNA and the PR-A/PR-B expression ration but did not effect PR-B mRNA. PGE2 dose-dependently increased mRNAs encoding PR-A and PR-B. The PGE2 dose-threshold for PR-A (0.01 nM) was lower than that for PR-B (0.1 nM), which resulted in an initial rise then a gradual fall in PR-A/PR-B expression ration to basal levels in response to PGE2. Activation of the protein kinase (PK)-A signaling pathway with 8-Br-cAMP coordinately increased expression of PR-A and PR-B and therefore did not alter the PR-A/PR-B expression ration. In contrast, activation of the PKC signaling pathway with PMA increased expression of PR-A without affecting PR-B and therefore significantly (P<0.05) increased the PR-A/PR-B expression ration. These data demonstrate differential control of myometrial PR-A and PR-B expression by PGE2 and PGF2 alpha and by specific intracellular signaling pathways. We conclude that PGs acting via the PKC pathway facilitate functional progesterone withdrawal by increasing the myometrial PR-A/PR-B expression ratio.


Subject(s)
Dinoprost/physiology , Dinoprostone/physiology , Myometrium/metabolism , Progesterone/antagonists & inhibitors , Receptors, Progesterone/metabolism , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Cell Line, Transformed , Dinoprost/administration & dosage , Dinoprostone/administration & dosage , Dose-Response Relationship, Drug , Female , Humans , Myometrium/cytology , RNA, Messenger/metabolism , Receptors, Progesterone/genetics , Tetradecanoylphorbol Acetate/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...