Semin Reprod Med 2007; 25(1): 052-059
DOI: 10.1055/s-2006-956775
Copyright © 2007 by Thieme Publishers, Inc., 333 Seventh Avenue, New York, NY 10001, USA.

Regulation of Oxytocin Receptors and Oxytocin Receptor Signaling

Andrew M. Blanks1 , Anatoly Shmygol1 , Steven Thornton1
  • 1Warwick Medical School, Clinical Sciences Research Institute, University Hospital of Coventry and Warwickshire, Coventry, United Kingdom
Further Information

Publication History

Publication Date:
05 January 2007 (online)

ABSTRACT

The oxytocin (OT) -oxytocin receptor (OTR) system plays a key role in many aspects of mammalian reproduction as well as several other physiological processes such as bond pairing and cardiovascular homeostasis. To manifest these diverse physiological roles, the transcription and expression of the OTR is tightly regulated within reproductive, cardiovascular, and neuronal tissues. The expression of the OTR within the mammalian uterus is regulated during gestation with a peak at the day of delivery. The control of this dramatic increase in expression is mediated in rodent species by a combination of stretch, classical steroid hormone stimulation, and repression. In the human uterus events are less clear, although a prominent role for inflammatory-related rapid-response genes and novel transcription factors such as hMafF (human homologue of chicken musculoaponeurotic fibrosarcoma oncogene family protein F) have been put forward. In the uterus the potent uterotonic actions of OT are mediated by the OTR through G-protein activation to stimulate phospholipase C activity. The activated OTR increases contraction frequency and increases force by sensitizing the contractile apparatus of the myocytes to calcium. This review summarizes the current knowledge of the complex regulation of OTR transcription in the myometrium and the intracellular signaling mechanisms through which OT mediates its potent stimulatory effects.

REFERENCES

  • 1 Bielsky I F, Young L J. Oxytocin, vasopressin, and social recognition in mammals.  Peptides. 2004;  25 1565-1574
  • 2 Cassoni P, Sapino A, Marrocco T, Chini B, Bussolati G. Oxytocin and oxytocin receptors in cancer cells and proliferation.  J Neuroendocrinol. 2004;  16 362-364
  • 3 Gimpl G, Fahrenholz F. The oxytocin receptor system: structure, function, and regulation.  Physiol Rev. 2001;  81 629-683
  • 4 Blanks A M, Thornton S. The role of oxytocin in parturition.  BJOG. 2003;  110(suppl 20) 46-51
  • 5 McCann S M, Antunes-Rodrigues J, Jankowski M, Gutkowska J. Oxytocin, vasopressin and atrial natriuretic peptide control body fluid homeostasis by action on their receptors in brain, cardiovascular system and kidney.  Prog Brain Res. 2002;  139 309-328
  • 6 Lipschitz D L, Crowley W R, Armstrong W E, Bealer S L. Neurochemical bases of plasticity in the magnocellular oxytocin system during gestation.  Exp Neurol. 2005;  196 210-223
  • 7 Fuchs A R, Fuchs F, Husslein P, Soloff M S. Oxytocin receptors in the human uterus during pregnancy and parturition.  Am J Obstet Gynecol. 1984;  150 734-741
  • 8 Kimura T, Tanizawa O, Mori K, Brownstein M J, Okayama H. Structure and expression of a human oxytocin receptor.  Nature. 1992;  356 526-529
  • 9 Kimura T. Investigation of the oxytocin receptor at the molecular level.  Adv Exp Med Biol. 1995;  395 259-268
  • 10 Copland J A, Ives K L, Simmons D J, Soloff M S. Functional oxytocin receptors discovered in human osteoblasts.  Endocrinology. 1999;  140 4371-4374
  • 11 Thibonnier M, Conarty D M, Preston J A et al.. Human vascular endothelial cells express oxytocin receptors.  Endocrinology. 1999;  140 1301-1309
  • 12 Inoue T, Kimura T, Azuma C et al.. Structural organization of the human oxytocin receptor gene.  J Biol Chem. 1994;  269 32451-32456
  • 13 Simmons Jr C F, Clancy T E, Quan R, Knoll J H. The oxytocin receptor gene (OXTR) localizes to human chromosome 3p25 by fluorescence in situ hybridization and PCR analysis of somatic cell hybrids.  Genomics. 1995;  26 623-625
  • 14 Murasawa S, Matsubara H, Kijima K et al.. Structure of the rat V1a vasopressin receptor gene and characterization of its promoter region and complete cDNA sequence of the 3′-end.  J Biol Chem. 1995;  270 20042-20050
  • 15 Terzidou V, Lee Y, Lindstrom T et al.. Regulation of the human oxytocin receptor by nuclear factor-kappaB and CCAAT/enhancer-binding protein-beta.  J Clin Endocrinol Metab. 2006;  91 2317-2326
  • 16 Kubota Y, Kimura T, Hashimoto K et al.. Structure and expression of the mouse oxytocin receptor gene.  Mol Cell Endocrinol. 1996;  124 25-32
  • 17 Bale T L, Dorsa D M. Cloning, novel promoter sequence, and estrogen regulation of a rat oxytocin receptor gene.  Endocrinology. 1997;  138 1151-1158
  • 18 Larcher A, Neculcea J, Breton C et al.. Oxytocin receptor gene expression in the rat uterus during pregnancy and the estrous cycle and in response to gonadal steroid treatment.  Endocrinology. 1995;  136 5350-5356
  • 19 Fuchs A R, Periyasamy S, Alexandrova M, Soloff M S. Correlation between oxytocin receptor concentration and responsiveness to oxytocin in pregnant rat myometrium: effects of ovarian steroids.  Endocrinology. 1983;  113 742-749
  • 20 Fang X, Wong S, Mitchell B F. Relationships among sex steroids, oxytocin, and their receptors in the rat uterus during late gestation and at parturition.  Endocrinology. 1996;  137 3213-3219
  • 21 Fang X, Wong S, Mitchell B F. Effects of RU486 on estrogen, progesterone, oxytocin, and their receptors in the rat uterus during late gestation.  Endocrinology. 1997;  138 2763-2768
  • 22 Young L J, Wang Z, Donaldson R, Rissman E F. Estrogen receptor alpha is essential for induction of oxytocin receptor by estrogen.  Neuroreport. 1998;  9 933-936
  • 23 Parry L J, Bathgate R A, Shaw G, Renfree M B, Ivell R. Evidence for a local fetal influence on myometrial oxytocin receptors during pregnancy in the tammar wallaby (Macropus eugenii).  Biol Reprod. 1997;  56 200-207
  • 24 Ou C W, Orsino A, Lye S J. Expression of connexin-43 and connexin-26 in the rat myometrium during pregnancy and labor is differentially regulated by mechanical and hormonal signals.  Endocrinology. 1997;  138 5398-5407
  • 25 Terzidou V, Sooranna S R, Kim L U et al.. Mechanical stretch up-regulates the human oxytocin receptor in primary human uterine myocytes.  J Clin Endocrinol Metab. 2005;  90 237-246
  • 26 Bale T L, Dorsa D M. Transcriptional regulation of the oxytocin receptor gene.  Adv Exp Med Biol. 1998;  449 307-315
  • 27 Hoare S, Copland J A, Wood I G et al.. Identification of a GABP alpha/beta binding site involved in the induction of oxytocin receptor gene expression in human breast cells, potentiation by c-Fos/c-Jun.  Endocrinology. 1999;  140 2268-2279
  • 28 Lye S J, Mitchell J, Nashman N et al.. Role of mechanical signals in the onset of term and preterm labor.  Front Horm Res. 2001;  27 165-178
  • 29 Metz R, Ziff E. cAMP stimulates the C/EBP-related transcription factor rNFIL-6 to trans-locate to the nucleus and induce c-fos transcription.  Genes Dev. 1991;  5 1754-1766
  • 30 Inoue H, Yokoyama C, Hara S, Tone Y, Tanabe T. Transcriptional regulation of human prostaglandin-endoperoxide synthase-2 gene by lipopolysaccharide and phorbol ester in vascular endothelial cells. Involvement of both nuclear factor for interleukin-6 expression site and cAMP response element.  J Biol Chem. 1995;  270 24965-24971
  • 31 Akira S, Kishimoto T. NF-IL6 and NF-kappa B in cytokine gene regulation.  Adv Immunol. 1997;  65 1-46
  • 32 Romero R, Avila C, Brekus C A, Morotti R. The role of systemic and intrauterine infection in preterm parturition.  Ann N Y Acad Sci. 1991;  622 355-375
  • 33 Dudley D J, Hunter C, Mitchell M D, Varner M W. Clinical value of amniotic fluid interleukin-6 determinations in the management of preterm labour.  Br J Obstet Gynaecol. 1994;  101 592-597
  • 34 Schmid B, Wong S, Mitchell B F. Transcriptional regulation of oxytocin receptor by interleukin-1beta and interleukin-6.  Endocrinology. 2001;  142 1380-1385
  • 35 Nelson D E, Ihekwaba A E, Elliot M et al.. Oscillations in NF-kappaB signaling control the dynamics of gene expression.  Science. 2004;  306 704-708
  • 36 Kimura T, Mizumoto Y, Ivell R. Differential protein-DNA binding analysis identifies a novel enhancer element, US-1, involved in the upregulation of the oxytocin receptor gene in human myometrium at term.  Mol Cell Endocrinol. 1999;  148 137-149
  • 37 Kimura T, Ivell R, Rust W et al.. Molecular cloning of a human MafF homologue, which specifically binds to the oxytocin receptor gene in term myometrium.  Biochem Biophys Res Commun. 1999;  264 86-92
  • 38 Bethin K E, Nagai Y, Sladek R et al.. Microarray analysis of uterine gene expression in mouse and human pregnancy.  Mol Endocrinol. 2003;  17 1454-1469
  • 39 Massrieh W, Derjuga A, Doualla-Bell F et al.. Regulation of the MAFF transcription factor by proinflammatory cytokines in myometrial cells.  Biol Reprod. 2006;  74 699-705
  • 40 Ye X, Li Y, Huang Q et al.. The novel human gene MIP functions as a co-activator of hMafF.  Arch Biochem Biophys. 2006;  449 87-93
  • 41 Ivell R, Bathgate R A, Walther N, Kimura T. The molecular basis of oxytocin and oxytocin receptor gene expression in reproductive tissues.  Adv Exp Med Biol. 1998;  449 297-306
  • 42 Mizumoto Y, Kimura T, Ivell R. A genomic element within the third intron of the human oxytocin receptor gene may be involved in transcriptional suppression.  Mol Cell Endocrinol. 1997;  135 129-138
  • 43 Qian A, Wang W, Sanborn B M. Evidence for the involvement of several intracellular domains in the coupling of oxytocin receptor to G alpha(q/11).  Cell Signal. 1998;  10 101-105
  • 44 Sanborn B M, Dodge K, Monga M et al.. Molecular mechanisms regulating the effects of oxytocin on myometrial intracellular calcium.  Adv Exp Med Biol. 1998;  449 277-286
  • 45 Hoare S, Copland J A, Strakova Z et al.. The proximal portion of the COOH terminus of the oxytocin receptor is required for coupling to g(q), but not g(i). Independent mechanisms for elevating intracellular calcium concentrations from intracellular stores.  J Biol Chem. 1999;  274 28682-28689
  • 46 Ku C Y, Qian A, Wen Y, Anwer K, Sanborn B M. Oxytocin stimulates myometrial guanosine triphosphatase and phospholipase-C activities via coupling to G alpha q/11.  Endocrinology. 1995;  136 1509-1515
  • 47 Arnaudeau S, Lepretre N, Mironneau J. Oxytocin mobilizes calcium from a unique heparin-sensitive and thapsigargin-sensitive store in single myometrial cells from pregnant rats.  Pflugers Arch. 1994;  428 51-59
  • 48 Phaneuf S, Carrasco M P, Europe-Finner G N, Hamilton C H, Lopez Bernal A. Multiple G proteins and phospholipase C isoforms in human myometrial cells: implication for oxytocin action.  J Clin Endocrinol Metab. 1996;  81 2098-2103
  • 49 Berridge M J, Irvine R F. Inositol trisphosphate, a novel second messenger in cellular signal transduction.  Nature. 1984;  312 315-321
  • 50 Word R A, Stull J T, Casey M L, Kamm K E. Contractile elements and myosin light chain phosphorylation in myometrial tissue from nonpregnant and pregnant women.  J Clin Invest. 1993;  92 29-37
  • 51 Oancea E, Meyer T. Protein kinase C as a molecular machine for decoding calcium and diacylglycerol signals.  Cell. 1998;  95 307-318
  • 52 Carrasco M P, Phaneuf S, Asboth G, Lopez Bernal A. Fluprostenol activates phospholipase C and Ca2 + mobilization in human myometrial cells.  J Clin Endocrinol Metab. 1996;  81 2104-2110
  • 53 Khac L D, Arnaudeau S, Lepretre N, Mironneau J, Harbon S. Beta adrenergic receptor activation attenuates the generation of inositol phosphates in the pregnant rat myometrium. Correlation with inhibition of Ca + + influx, a cAMP-independent mechanism.  J Pharmacol Exp Ther. 1996;  276 130-136
  • 54 Molnar M, Rigo Jr J, Hertelendy F. Signal transduction in human myometrial cells.  Acta Physiol Hung. 1996;  84 89-97
  • 55 Baek K J, Kwon N S, Lee H S et al.. Oxytocin receptor couples to the 80 kDa Gh alpha family protein in human myometrium.  Biochem J. 1996;  315(pt 3) 739-744
  • 56 Park E S, Won J H, Han K J et al.. Phospholipase C-delta1 and oxytocin receptor signalling: evidence of its role as an effector.  Biochem J. 1998;  331(pt 1) 283-289
  • 57 Shmygol A, Gullam J, Blanks A, Thornton S. Multiple mechanisms involved in oxytocin-induced modulation of myometrial contractility.  Acta Pharmacol Sin. 2006;  27 827-832
  • 58 Parkington H C, Tonta M A, Davies N K, Brennecke S P, Coleman H A. Hyperpolarization and slowing of the rate of contraction in human uterus in pregnancy by prostaglandins E2 and f2alpha: involvement of the Na + pump.  J Physiol. 1999;  514(pt 1) 229-243
  • 59 Arnaudeau S, Lepretre N, Mironneau J. Chloride and monovalent ion-selective cation currents activated by oxytocin in pregnant rat myometrial cells.  Am J Obstet Gynecol. 1994;  171 491-501
  • 60 Mironneau J. Ion channels and the control of uterine contractility. In: Control of Uterine Contractility. Garfield RE, Tabb TN Boca Raton, FL; CRC Press 1994: 1-20
  • 61 Nakao K, Inoue Y, Okabe K, Kawarabayashi T, Kitamura K. Oxytocin enhances action potentials in pregnant human myometrium-a study with microelectrodes.  Am J Obstet Gynecol. 1997;  177 222-228
  • 62 Kawarabayashi T, Kishikawa T, Sugimori H. Effect of oxytocin on spontaneous electrical and mechanical activities in pregnant human myometrium.  Am J Obstet Gynecol. 1986;  155 671-676
  • 63 McKillen K, Thornton S, Taylor C W. Oxytocin increases the [Ca2 + ]i sensitivity of human myometrium during the falling phase of phasic contractions.  Am J Physiol. 1999;  276 E345-E351
  • 64 Somlyo A P, Somlyo A V. Signal transduction by G-proteins, rho-kinase and protein phosphatase to smooth muscle and non-muscle myosin II.  J Physiol. 2000;  522(Pt 2) 177-185
  • 65 Kimura K, Ito M, Amano H et al.. Regulation of myosin phosphatase by Rho and Rho-associated kinase (Rho-kinase).  Science. 1996;  273 245-248
  • 66 Katoh H, Aoki J, Yamaguchi Y et al.. Constitutively active Galpha12, Galpha13, and Galphaq induce Rho-dependent neurite retraction through different signaling pathways.  J Biol Chem. 1998;  273 28700-28707
  • 67 Moore F, Da Silva C, Wilde J I et al.. Up-regulation of p21- and RhoA-activated protein kinases in human pregnant myometrium.  Biochem Biophys Res Commun. 2000;  269 322-326
  • 68 Kupittayanant S, Burdyga T, Wray S. The effects of inhibiting Rho-associated kinase with Y-27632 on force and intracellular calcium in human myometrium.  Pflugers Arch. 2001;  443 112-114
  • 69 Woodcock N A, Taylor C W, Thornton S. Effect of an oxytocin receptor antagonist and rho kinase inhibitor on the [Ca + + ]i sensitivity of human myometrium.  Am J Obstet Gynecol. 2004;  190 222-228
  • 70 Kitazawa T, Eto M, Woodsome T P, Brautigan D L. Agonists trigger G protein-mediated activation of the CPI-17 inhibitor phosphoprotein of myosin light chain phosphatase to enhance vascular smooth muscle contractility.  J Biol Chem. 2000;  275 9897-9900
  • 71 Koyama M, Ito M, Feng J et al.. Phosphorylation of CPI-17, an inhibitory phosphoprotein of smooth muscle myosin phosphatase, by Rho-kinase.  FEBS Lett. 2000;  475 197-200
  • 72 Gong M C, Kinter M T, Somlyo A V, Somlyo A P. Arachidonic acid and diacylglycerol release associated with inhibition of myosin light chain dephosphorylation in rabbit smooth muscle.  J Physiol. 1995;  486(pt 1) 113-122

 Dr.
Andrew M Blanks

RCUK Fellow in Reproduction, Warwick Medical School, Clinical Sciences Research Institute, University Hospital of Coventry and Warwickshire

Clifford Bridge Road, Coventry CV2 2DX, United Kingdom

Email: Andrew.Blanks@warwick.ac.uk

    >