Regulation of electrolyte transport across cultured endometrial epithelial cells by prolactin

in Journal of Endocrinology
Authors:
Chatsri Deachapunya
Search for other papers by Chatsri Deachapunya in
Current site
Google Scholar
PubMed
Close
,
Sutthasinee Poonyachoti Department of Physiology, Department of Physiology, Consortium for Calcium and Bone Research, Faculty of Medicine, Srinakharinwirot University, Sukhumvit 23, Wattana, Bangkok 10110, Thailand

Search for other papers by Sutthasinee Poonyachoti in
Current site
Google Scholar
PubMed
Close
, and
Nateetip Krishnamra Department of Physiology, Department of Physiology, Consortium for Calcium and Bone Research, Faculty of Medicine, Srinakharinwirot University, Sukhumvit 23, Wattana, Bangkok 10110, Thailand
Department of Physiology, Department of Physiology, Consortium for Calcium and Bone Research, Faculty of Medicine, Srinakharinwirot University, Sukhumvit 23, Wattana, Bangkok 10110, Thailand

Search for other papers by Nateetip Krishnamra in
Current site
Google Scholar
PubMed
Close

Free access

Sign up for journal news

The effect of prolactin (PRL) on ion transport across the porcine glandular endometrial epithelial cells was studied in primary cell culture using the short-circuit current technique. Addition of 1 μg/ml PRL either to the apical solution or to the basolateral solution produced a peak followed by a sustained increase in Isc, but with a lesser response when PRL was added apically. Basolateral addition of PRL increased the Isc in a concentration-dependent manner with a maximum effect at 1 μg/ml and an effective concentration value of 120 ng/ml. The PRL-stimulated Isc was significantly reduced by pretreatment with an apical addition of 5-nitro-2-(3-phenylpropylamino) benzoic acid (200 μM), diphenylamine-2-carboxylic acid (1 mM) or 4,4′-diisothiocyanatostilbene-2,2′-disulfonic acid (200 μM), Cl channel blockers, but not by amiloride (10 μM), a Na+ channel blocker. In addition, pretreatment with bumetanide (200 μM), a Na+–K+–2Cl cotransporter inhibitor, in the basolateral solution significantly reduced the PRL-stimulated Isc. Replacement of Cl or in the bathing solutions also decreased the Isc response to PRL. Pretreatment of the monolayer with AG490 (50 μM), an inhibitor of JAK2 activity significantly inhibited the PRL-induced increase in Isc. Western blot analysis of the porcine endometrial epithelial cells revealed the presence of short isoform of PRL receptor (PRLR-S) that could be regulated by 17β-estradiol. The results of this investigation showed that PRL acutely stimulated anion secretion across the porcine endometrial epithelial cells possibly through PRLR-S present in both apical and basolateral membranes. The PRL response appeared to be mediated by the JAK2-dependent pathway.

Abstract

The effect of prolactin (PRL) on ion transport across the porcine glandular endometrial epithelial cells was studied in primary cell culture using the short-circuit current technique. Addition of 1 μg/ml PRL either to the apical solution or to the basolateral solution produced a peak followed by a sustained increase in Isc, but with a lesser response when PRL was added apically. Basolateral addition of PRL increased the Isc in a concentration-dependent manner with a maximum effect at 1 μg/ml and an effective concentration value of 120 ng/ml. The PRL-stimulated Isc was significantly reduced by pretreatment with an apical addition of 5-nitro-2-(3-phenylpropylamino) benzoic acid (200 μM), diphenylamine-2-carboxylic acid (1 mM) or 4,4′-diisothiocyanatostilbene-2,2′-disulfonic acid (200 μM), Cl channel blockers, but not by amiloride (10 μM), a Na+ channel blocker. In addition, pretreatment with bumetanide (200 μM), a Na+–K+–2Cl cotransporter inhibitor, in the basolateral solution significantly reduced the PRL-stimulated Isc. Replacement of Cl or in the bathing solutions also decreased the Isc response to PRL. Pretreatment of the monolayer with AG490 (50 μM), an inhibitor of JAK2 activity significantly inhibited the PRL-induced increase in Isc. Western blot analysis of the porcine endometrial epithelial cells revealed the presence of short isoform of PRL receptor (PRLR-S) that could be regulated by 17β-estradiol. The results of this investigation showed that PRL acutely stimulated anion secretion across the porcine endometrial epithelial cells possibly through PRLR-S present in both apical and basolateral membranes. The PRL response appeared to be mediated by the JAK2-dependent pathway.

Introduction

Endometrial epithelial cells play an important role in the regulation of fluid and electrolyte volume and composition within the uterine cavity, providing an optimal intrauterine environment for implantation and embryo development. The transport-related activities of the surface and glandular epithelial cells have been shown to be regulated by several hormones, growth factors, cytokines, and a number of signaling molecules. Electrophysiological studies of cultured human endometrial epithelial cells (Matthews et al. 1993) and the intact porcine endometrial epithelium (Vetter & O'Grady 1996) have provided direct evidence for the regulation of Na+ absorption and K+ secretion. In the primary culture of mouse and porcine endometrial epithelial cells, prostaglandins (PGs) especially PGE2, adrenaline, ATP, and UTP were found to activate anion secretion (Chan et al. 1997, Fong et al. 1998, Deachapunya & O'Grady 1998, Palmer-Densmore et al. 2002). These epithelial cells also exhibited Na+ transport that was activated by insulin and insulin-like growth factor, and inhibited by epidermal growth factor (Deachapunya et al. 1999, Deachapunya & O'Grady 2001).

Prolactin (PRL) is synthesized and secreted from the anterior pituitary gland, as well as the extrapituitary tissues including myometrium, deciduas, and mammary epithelial cells (Freeman et al. 2000). It exerts a wide variety of biological actions, such as the regulation of water and electrolyte balance, growth of mammary gland, milk production, and secretion. Recently, PRL has been reported to stimulate the intestinal Ca2+ absorption (Jantarajit et al. 2007), especially under conditions of high calcium demand such as pregnancy and lactation (Charoenphandhu & Krishnamra 2007).

In human endometrium, PRL receptors (PRL-R) and its mRNA have been identified in glandular epithelial and stromal cells (Jabbour et al. 1998, Tseng & Zhu 1998). PRL-R belongs to the superfamily of the cytokine class-1 receptor (Kelly et al. 1991). Several isoforms, i.e. short, intermediate, and long isoforms and the soluble PRL-binding protein have been identified in many tissues (Clevenger & Kline 2001). Binding of PRL to its transmembrane receptors induces receptor dimerization, tyrosine phosphorylation, and activation of the JAK, which leads to phosphorylation of other associated regulatory proteins especially the STAT proteins. The phosphorylated STAT proteins dimerize and translocate to the nucleus to bind to the PRL-responsive genes, resulting in target gene transcription and biological responses. Other signaling pathways involving mitogen-activated protein kinase (MAPK), insulin-receptor substrate (IRS-1), phosphoinositide 3 (PI-3) kinase, PLC, PKC, and intracellular Ca2+ have also been reported to mediate PRL actions (Bole-Feysot et al. 1998, Gubbay et al. 2002).

Although PRL is known as an important regulator of water and electrolyte transport in lower vertebrates (Bern 1975, Sakamoto & McCormick 2006), there were very few reports on its transport-related effect in the mammalian epithelial cells. Several studies using the everted intestinal sac technique have demonstrated the stimulatory effect of PRL on fluid and NaCl absorption in the rat, hamster, and guinea pig jejunum, but not in guinea pig ileum or rat colon (Mainoya et al. 1974). In the rabbit mammary glands, PRL decreased epithelial membrane permeability to sucrose, suggesting a decrease in the permeability of the tight junction (Linzell et al. 1975). From the studies using the mouse mammary epithelial cells grown on floating collagen gels, PRL treatment for 3 days was found to increase the short-circuit current (Isc) and transepithelial potential difference (PD), which indicated an increase in net active Na+ absorption, probably with some Cl secretion (Bisbee et al. 1979). In the mouse mammary epithelial cell line HC11, PRL acutely increased Cl transport through the JAK–STAT system (Selvaraj et al. 2000). Eventhough PRL seemed to play an important role in the regulation of transepithelial ion transport in a variety of epithelia, its effect on the ion transport function of the endometrial epithelium has not been investigated. Since we have previously shown that the primary cultured porcine endometrial epithelial cells possessed the transport machinery capable of Na+ absorption and Cl secretion (Deachapunya & O'Grady 1998, Deachapunya et al. 1999), the objectives of the present study were to investigate the regulatory mechanism of PRL on the ion transport across these epithelial cells.

Materials and Methods

Materials

PRL, insulin, amiloride, 5-nitro-2-(3-phenylpropylamino) benzoic acid (NPPB), diphenylamine-2-carboxylic acid (DPC), 4,4′-diisothiocyanatostilbene-2,2′-disulfonic acid (DIDS), bumetanide, acetazolamide, PGE2, 8-chloro-phenyl-thio-3′,5′-cyclicmonophosphate (8cpt-cAMP), non-essential amino acids, and high-purity grade salts were obtained from Sigma Chemical Co. Dulbecco's modified Eagle's medium (DMEM), Dulbecco's PBS (DPBS), fetal bovine serum (FBS), collagenase (type 1), kanamycin, penicillin–streptomycin, and fungizone were purchased from Gibco (Grand Island, NY).

Cell isolation and culture

Porcine uterine tissues collected from 5- to 6-month-old pig were obtained from the Metropolitan slaughterhouse, Klongtoey, Bangkok, under the supervision of the Department of Livestock Development, Ministry of Agriculture and Cooperatives, Thailand. The tissue was placed in an ice-cold porcine Ringer solution containing (mM): 130 NaCl, 6 KCl, 3 CaCl2, 0.7 MgCl2, 20 NaHCO3, 0.3 NaH2PO4, and 1.3 Na2HPO4 (pH 7.4). After removal of the serosal muscle layer, the tissue fragments were cut into small pieces and digested overnight with collagenase. The epithelial glands were then isolated as described previously (Deachapunya & O'Grady 1998), and suspended in DMEM supplemented with 3.7 g/l NaHCO3, 10% FBS, 850 nM (5 μg/ml) insulin, 1% non-essential amino acid, 5 μg/ml fungizone, 100 U/ml penicillin, 100 μg/ml streptomycin, and 100 μg/ml kanamycin (standard media). They were then plated onto the cell culture dishes and incubated at 37 °C in a humidified atmosphere of 5%CO2 in air. Culture medium was changed after 24 h and then every 2–3 days. After 80% confluence (within 2–3 days), the epithelial cells were subcultured onto 24 mm (4.5 cm2) transparent permeable membrane filters (Costar, Cambridge, MA, USA). Using this method of isolation, the purity of epithelial cells was greater than 90% as assessed by staining the isolated cells with cytokeratin (Deachapunya & O'Grady 1998). Cell monolayers were fed every 2 days and maintained in the standard media for about 7 days before the beginning of the experiment.

Measurement of electrical parameters

Before studying ion transport, the transepithelial resistance of the cell monolayer was measured with an epithelial voltohmmeter (EVOM) coupled to Ag/AgCl ‘chopstick’ electrodes (World Precision Instruments, Serasota, FL, USA). Monolayer with high resistance (≈3000 Ωcm2) was then mounted in Ussing Chamber, bathed in both sides with the standard porcine Ringer solution, which was maintained at 37 °C and bubbled with 95%O2–5%CO2. Transepithelial PD and Isc were measured with the use of voltage-clamp circuitry (EVC-4000, World Precision Instruments) with Ag/AgCl electrodes connected to the bathing solution via agar bridges. Tissue conductance (G) was calculated using Ohm's law (G=Isc/PD). The monolayer was continuously short circuited, except for a brief interval of open-circuited readings for PD measurement before and after adding any chemical. Data from the voltage clamp were connected to a MacLab 4S A/D converter and recorded with a 400 MHz PowerPC Macintosh. After mounting, the cell monolayer was equilibrated for at least 20 min to achieve a stable Isc before addition of chemicals. Positive Isc corresponded to the movement of anions in the serosal to mucosal direction or the movement of cations in the mucosal to serosal direction or a combination of both. In the anion replacement experiments, gluconate salts were substituted for chloride and HEPES were substituted for bicarbonate. The experiment under free condition was performed in the presence of 100 μM acetazolamide and bubbled with 100% O2.

Western blot analysis

Porcine endometrial epithelial cells seeded in the 100 mm cell culture dish were allowed to grow in the standard medium up to 80% confluence. In some experiments, the medium was switched to the serum-free and phenol red-free DMEM alone or supplemented with 10−8 M 17β-estradiol for 48 h. Cells were then harvested and suspended in lysis buffer containing 50 mM Tris–HCl, 1% NP-40, 0.25% sodium deoxycholate, 150 mM NaCl, 1 mM EGTA, 1 mM phenylmethylsulfonyl fluoride, 20 μg aprotinin, and 1 mM NaF (pH 7.4). The supernatant was collected and protein concentrations were determined using the BCA protein assay kit (Pierce Biotechnology, Inc., Rockford, IL, USA). Protein samples (20 μg) were separated by 10% SDS-PAGE and electrically transferred to a polyvinylidene difluoride membrane (Hybond-P, Amersham Biosciences) in Tris–glycine buffer. Blotted membranes were washed and then blocked with 5% nonfat powdered milk in Tris-buffered saline for 4 h at room temperature with constant agitation. The membrane was incubated overnight at 4 °C with 1 μg/ml primary antibody, which is anti-rat PRL receptor monoclonal antibody generated against the extracellular domain of PRL receptor (clone U5, Affinity BioReagents). The membrane was then washed and incubated for 1 h at room temperature with a 1:10 000 horseradish peroxidase (HRP)-conjugated goat anti-mouse secondary antibody (Zymed Laboratories Inc., San Francisco, CA, USA). After washing, the immunoreactive protein bands were visualized using the enhanced chemiluminescence (ECL) detection system (Santa Cruz Biotechnology Inc., Santa Cruz, CA, USA) according to the manufacturer's instructions. The membranes were exposed to film (Hyperfilm-ECL, Amersham Biosciences) for adequate duration to visualize the chemiluminescent bands. To confirm equal loading, the membranes were stripped and reprobed with a 1:300 000 anti-β-actin monoclonal antibody (clone AC-15, Sigma Co.) followed by a 1:10 000 HRP-conjugated anti-mouse antibody. The intensity of the protein bands was determined using densiotometry analysis (Scion Image; Scion Cooperation, Frederick, MD, USA). Band intensity of the PRL-R from each treatment was normalized to the β-actin intensity and expressed as the PRL-R/β-actin ratio.

Data analyses

All values are presented as mean±s.e.m. and n is the number of monolayers from at least three different uterine tissue cultures. The differences between control and experimental means were analyzed using a Student's t-test or ANOVA where appropriate. The difference between treatment and control means following a significant ANOVA was identified by Dunnett's test (Prism 3.0, GraphPad Software, Inc., San Diego, CA, USA). A value of P<0.05 was considered statistically significant. The effective concentration (EC50) value was determined using a four-parameter logistic function to fit the data (Prism 3.0; GraphPad Software Inc.).

Results

Effect of PRL on Isc

Under basal condition, after an equilibrating period of 30 min, the porcine endometrial epithelial monolayer exhibited average Isc, PD (lumen negative), and tissue conductance of 30.68±2.49 μA, −27.22±3.89 mV, and 1.47±0.25 mS (n=17) respectively. Addition of 1 μg/ml PRL to the apical or basolateral solution produced an increase in Isc that reached a peak within 2–3 min before decreasing slightly and was maintained at a level above baseline (Fig. 1A). In some experiments, the PRL-stimulated Isc gradually decreased to the baseline level. The peak Isc response to apical addition of 1 μg/ml PRL was 3.45±0.94 μA (n=4), while the subsequent basolateral addition of 1 and 5 μg/ml PRL led to a peak Isc of 11.00±0.98 μA (n=17) and 14.36±1.37 μA (n=12) respectively. Basolateral addition of PRL increased the Isc in a concentration-dependent manner with a maximum response with 1 μg/ml PRL and a half maximum EC50 value of 120 ng/ml (n=6, Fig 1B).

Figure 1
Figure 1

Effect of PRL on the basal Isc in the endometrial epithelial monolayers. (A) A representative Isc tracing responded to an addition of 1 μg/ml PRL to (a) the apical solution followed by 1 and 5 μg/ml PRL added to (b) the basolateral solution. (B) Concentration–response relationships showing the increase in Isc following basolateral treatment with various concentrations of PRL. The EC50 value was 120 ng/ml (n=6).

Citation: Journal of Endocrinology 197, 3; 10.1677/JOE-08-0077

Ionic basis of the PRL-stimulated Isc

To determine the ionic basis of the Isc response induced by PRL, we examined the effect of PRL in the presence of pharmacological ion channel blockers and ion substitution solutions. An apical application of the Na+ channel blocker amiloride at 10 μM inhibited the basal Isc by 35% (Fig. 2A). However, it did not affect the Isc response induced by PRL (1 μg/ml), which was 12.21±1.39 μA (n=5), when compared with the control value of 11.44±1.22 μA (n=12) (Fig. 2E). By contrast, the PRL-stimulated Isc was significantly decreased in the presence of Cl channel blockers, NPPB, DPC, and DIDS in the apical solution. NPPB and DPC have been widely used to block CFTR, whereas DIDS blocks the Ca2+-activated Cl channels with no effect on the activity and conductance of CFTR (Anderson et al. 1992). As shown in Fig. 2B and C, pretreatment with 100 μM NPPB or 1 mM DPC reduced the basal Isc by 60 and 75% and decreased the Isc response to PRL to 1.00±0.71 μA (n=4) and 3.65±0.95 μA (n=5) respectively. An apical addition of 200 μM DIDS reduced the basal Isc by 17% and reduced the Isc response to PRL to 6.43±1.20 μA (n=4) (Fig. 2D). In addition, pretreatment with 200 μM bumetanide, a Na+–K+–2Cl cotransporter inhibitor, in the basolateral solution abolished most of the PRL-induced increase in Isc from 11.44±1.22 μA (n=12) to 2.48±1.17 μA (P<0.01, n=4) (Fig. 3). Replacement of Cl or in both the apical and the basolateral solutions markedly reduced the maximal Isc response to 1 μg/ml PRL to 1.63±1.06 μA (n=5) and 0.9±0.3 μA (n=3) respectively (Fig. 4).

Figure 2
Figure 2

Effect of ion channel blockers on the PRL-stimulated Isc. (A) A representative Isc tracing showing that an apical addition of amiloride (Amil, 10 μM) produced a small decrease in basal Isc. A subsequent addition of PRL (1 μg/ml) into the basolateral solution produced an increase in Isc. (B) An apical addition of NPPB (100 μM), (C) DPC (1 mM), or (D) DIDS (200 μM) decreased the basal Isc and reduced the PRL response. (E) Bar graph illustrating the average maximal increases in Isc response produced by prolactin alone (control) and in the presence of amiloride, NPPB, DPC, or DIDS. Values represent means±s.e.m. *P<0.01 when compared with the control value by ANOVA.

Citation: Journal of Endocrinology 197, 3; 10.1677/JOE-08-0077

Figure 3
Figure 3

Effect of Na+–K+–2Cl cotransporter blockers on the PRL-stimulated Isc. (A) A representative Isc tracing showing that a basolateral addition of bumetanide (200 μM) produced a decrease in basal Isc. A subsequent addition of PRL (1 μg/ml) into the basolateral solution produced a slightly increase in Isc. (B) Bar graph illustrating the average maximal increases in Isc response produced by prolactin alone (control) and in the presence of bumetanide. Values represent means±s.e.m. *P<0.01 when compared with the control value by Student's t-test.

Citation: Journal of Endocrinology 197, 3; 10.1677/JOE-08-0077

Figure 4
Figure 4

Effects of anion substitution on the maximal Isc response to PRL. In standard Ringer's solution, PRL (1 μg/ml) produced a mean increase in Isc of 11.44±1.22 μA (n=12). Replacement of Cl and in both the apical and basolateral solutions significantly inhibited the maximal Isc response to PRL by 86% (n=4) and 92% (n=4) respectively. *P<0.01 when compared with the control value by ANOVA.

Citation: Journal of Endocrinology 197, 3; 10.1677/JOE-08-0077

Intracellular signaling pathways of PRL-induced increase in Isc

The major signaling pathway involved in PRL action is the JAK–STAT pathway which was shown to mediate Cl secretion in the mammary cell line, HC11 (Selvaraj et al. 2000). Phosphorylations of JAK2, STAT1, and STAT5 have been demonstrated in response to PRL stimulation (200 ng/ml) in human endometrium (Jabbour et al. 1998). To determine whether JAK2 was involved in the PRL-induced increase in Isc, we examined the effect of AG490, an inhibitor of JAK2 activity, on PRL action. As shown in Fig 5A, pretreatment with 50 μM AG490, added to both the basolateral and apical solutions, reduced the basal Isc by 55% from 26.21±4.99 to 11.75±4.01 μA (P<0.01, n=5). Sequential additions of 1 and 5 μg/ml PRL slightly increased the Isc response by 3.70±3.09 and 0.80±0.66 μA (n=5) respectively. However, the presence of AG490 did not affect the Isc response to 100 μM 8cpt-cAMP, which was 22.80±2.14 μA (n=4) when compared with the control value of 18.99±1.59 μA (n=5), but slightly decreased the Isc response to 3 μM PGE2 to 12.08±0.87 μA (n=4), which was not statistically significant from that of control (16.84±1.55 μA, n=7, Fig. 5B).

Figure 5
Figure 5

Effect of JAK2 activity inhibitor on the PRL-stimulated Isc. (A) A representative Isc tracing showing that an addition of AG490 (50 μM), an inhibitor of JAK2 activity, to both apical and basolateral solutions completely inhibited both basal and PRL-induced increase in Isc. (B) Bar graph illustrating the average maximal increases in Isc response produced by PRL (1 μg/ml), 8cpt-cAMP (cAMP, 100 μM), or prostaglandin E2 (PGE, 3 μM) in the absence and presence of AG490 (50 μM). Values represent means ±s.e.m. *P<0.01 when compared with the corresponding control value by Student's t-test.

Citation: Journal of Endocrinology 197, 3; 10.1677/JOE-08-0077

Expression of PRL receptor

To confirm the functional significance of PRL in the regulation of ion transport, the expression of PRL-R was determined using western blot analysis. A representative western blot as presented in Fig. 6A demonstrated the presence of proteins with an approximate molecular mass of 36 kDa in porcine endometrial epithelial cells as well as in the human mammary gland cancer cell MCF-7 and human endometrial cancer cell RL-95. The 36 kDa protein band corresponded to the short form of PRL-R. Replacement of the standard medium of the endometrial epithelial cells with the serum-free and phenol red-free medium reduced the expression of the protein, whereas addition of 17β-estradiol (10−8 M) in the serum-free medium up-regulated the PRL protein expression. Based on the densitometry analysis, the PRL-R/β-actin ratio was significantly decreased from 5.67±0.79 in the standard medium to 1.15±0.09 (P<0.05, n=4) in the serum-free medium. Treatment with 17β-estradiol increased the PRL-R/β-actin ratio by twofold to 2.35±0.12.

Figure 6
Figure 6

Expression of PRL receptor protein in porcine endometrial epithelial cells. (A) Western blot analysis of PRL receptor (PRL-R). A 36 kDa band of PRL-R was detected in human mammary gland cancer cell MCF-7, human endometrial cancer cell RL-95, and porcine endometrial epithelial cells (PE) under standard medium (SM), serum-free medium (SF) alone, or supplemented with 10−8 M 17β-estradiol (SF+E 10 nM). The internal control of 43 kDa band of β-actin was also detected in all samples. (B) Bar graph illustrating the ratio of PRL-R to β-actin protein expression in the endometrial epithelial cells, based on the densitometry of immunoblots obtained from SM, SF, and SF+E10 nM (n=4). Values represent means±s.e.m. *P<0.05 when compared with the serum-free medium condition by Student's t-test.

Citation: Journal of Endocrinology 197, 3; 10.1677/JOE-08-0077

Discussion

A previous study in mouse mammary epithelial cells demonstrated that the PRL-induced increase in Isc was predominately mediated by Na+ absorption (Mainoya et al. 1974). However, in another study in the mouse mammary epithelial cell line HC11, it was Cl transport that was acutely stimulated by PRL (Selvaraj et al. 2000). Using cultured porcine endometrial epithelial cells that possessed the machinery for Na+ and Cl transports, we showed that PRL acutely stimulated anion secretion without affecting Na+ absorption. This was further supported by the findings that the PRL-induced increase in Isc was blocked by Cl channel blockers, NPPB, DPC, and DIDS, but not by Na+ channel blocker, amiloride. In addition, the basolateral pretreatment with bumetanide, a blocker of Na+–K+–2Cl cotransporter, or replacement of Cl or significantly inhibited the PRL-stimulated Isc. PRL added to the basolateral solution of the high-resistance monolayer produced a greater increase in the Isc response than when added to the apical solution, suggesting that PRL receptors were predominately located at the basolateral membrane. Western blot analysis revealed the expression of a 36 kDa protein band, which corresponded to the short form of PRL-R, indicating the role of this PRL receptor isoform in the mediation of transepithelial anion secretion in the porcine endometrial epithelium.

The basal electrical properties of cultured epithelial cells used in the present study have been described previously (Deachapunya & O'Grady 1998). Under the basal condition, these cells exhibited substantial Isc that was due to a greater Cl secretion than Na+ absorption. Application of PRL produced a concentration-dependent increase in the anion transport with a maximal effect seen at 1 μg/ml PRL and an EC50 value of 120 ng/ml. In addition, the maximal PRL response observed within 3–5 min after application, implied a non-genomic action. At PRL concentration of 1 μg/ml, which could be considered a hyperprolactinemic level, PRL was also found to maximally stimulate Cl transport in the mammary epithelial HC11 cells (Selvaraj et al. 2000). This effective concentration of PRL was comparable with the circulating levels during pregnancy and lactation in the human and the rat (Handwerger & Freemark 1987, Arbogast & Voogt 1998). Like in many other epithelia, Cl secretion across the endometrial epithelial cells requires activation of the apical membrane Cl channels and the basolateral membrane K+ channels with the basolateral Na+–K+–2Cl cotransporters serving as the Cl loading step. Since two types of Cl channels, cAMP-activated cystic fibrosis transmembrane conductance regulator (CFTR) and Ca2+-activated Cl channels have been identified in a variety of epithelia including endometrial epithelial cells (Deachapunya & O'Grady 1998, Palmer-Densmore et al. 2002), NPPB and DPC, inhibitors of anion channels including CFTR, and DIDS, an inhibitor of Ca2+-activated Cl channels, were used to elucidate PRL action. As the present results showed that the PRL-induced increase in Isc was nearly completely inhibited by DPC and NPPB, it was most likely that CFTR was the primary target of the PRL-stimulated Cl secretion. However, the 40% inhibition of the PRL-stimulated Isc by DIDS indicated that the Ca2+-activated Cl channels may also be partially involved in the PRL activation of Cl secretion. In addition, the PRL-stimulated increase in Isc was also diminished by bumetanide, an inhibitor of Na+–K+–2Cl cotransporter, which was not surprising since the bumetanide-sensitive Cl uptake probably served as the Cl loading step for PRL-stimulated Cl secretion. The finding that Isc response to PRL was abolished in the Clfree solution further confirmed the PRL activation of Cl secretion. Since the electronic secretion used the cAMP- and Ca2+-activated Cl channels (Illek et al. 1999), a marked reduction of Isc response to PRL in the free solution suggested a substantial contribution of to the PRL-induced increase in Isc. Altogether, the results suggest a possible involvement of exchangers as well as cotransporters in the PRL-dependent secretion. However, it was noted that the replacement of itself, could have produced the intracellular acidification, which could affect the transport pathways or the signaling mechanisms that regulate anion secretion. Taken together, it could be stated that PRL-induced anion secretion by stimulating the apical Cl efflux through CFTR as the major channel type and through some Ca2+-activated Cl channels, concurrently with the increase in the basolateral Cl uptake through the bumetanide-sensitive Na+–K+–2Cl cotransporter. Although not being investigated in this study, the basolateral K+ channels that provide the driving force for Cl exit across the apical membrane, could also be a target of PRL action, and are subject to further investigation.

Several signaling pathways mediating the multiple actions of PRL have been demonstrated in a variety of tissues with the JAK–STAT pathway being most extensively studied. Previous evidence of the phosphorylation of JAK2, STAT1, and STAT5 in response to PRL stimulation in the human endometrium (Jabbour et al. 1998) and the PRL-stimulated Cl transport through the JAK2 cascade pathway in mouse mammary epithelial cell line HC11 (Selvaraj et al. 2000) suggested that JAK2 is a likely mediator of PRL-stimulating effect on the anion transport in the porcine endometrial epithelial cells. In the present study, pretreatment with AG490, an inhibitor of JAK2 activity inhibited both the basal Isc and the PRL-induced increase in Isc. Regarding the basal Isc, since the basal Isc of the cultured porcine endometrial epithelial cells has been shown to be generated mainly by Cl secretion (Deachapunya & O'Grady 1998), the fact that AG490 could inhibit the basal Isc within 10 min suggested that a constitutive JAK2 activity was responsible for the basal active Cl secretion. Although no direct association between JAK2 activity and Cl transport mechanism has been reported, the fact that tyrosine-phosphorylated proteins could regulate the basal Cl secretion in human colonic epithelial cell line, T84 (Uribe et al. 1996) suggested that the JAK2 inhibitors inhibited Cl secretion by interfering with the tyrosine phosphorylation of the regulatory transport proteins. Furthermore, based on the findings that i) Cl secretion could still be activated by 8cpt-cAMP in the presence of JAK2 inhibitor, ii) the inhibition of the basal Isc did not affect the PGE2-stimulated Isc response, and iii) the PGE2-stimulated Cl secretion was via the cAMP-dependent pathway (Deachapunya & O'Grady 1998), it is likely that the cAMP-dependent Cl secretion did not involve JAK2 pathway.

In contrast to the PGE2-stimulated Cl secretion, the PRL-stimulated Cl secretion was probably mediated by the JAK2 pathway because the PRL-induced increase in Isc was significantly inhibited by AG490. These findings were consistent with a report of AG490 blocking phosphorylation of STAT5 and PRL-induced Cl secretion, but not the PGE1-induced Cl secretion in mouse mammary cell line (Selvaraj et al. 2000). The effect of AG490 was more specific to PRL action, since tyrosine kinase inhibitor genistein had no effect on the PRL-stimulated Isc (data not shown).

PRL-R and its mRNA have been identified in human glandular epithelial and stromal cells (Jabbour et al. 1998, Tseng & Zhu 1998). Two isoforms of PRL-R, short and long, have been identified in several rat tissues including liver, ovary, thymus, and spleen (Gunes & Mastro 1996, Telleria et al. 1997). In the present study, we examined the expression of PRL-R protein in the endometrial epithelial cells by western blot analysis. The monoclonal antibody used in the present study detected protein with molecular mass of about 36 kDa, which corresponded to the short form of the PRL-R, similar to the predominant short isoforms expressed in the rat spleen and brain (Shingo et al. 2003). By contrast, the long form of PRL-R is the major receptor isoforms in the rat liver and mammary gland (Jahn et al. 1991, Selvaraj et al. 2000). Although at least two isoforms of PRL-R mRNA have been found in the reproductive tissues (Telleria et al. 1997), the present data indicated that the short form PRL-R was the functional PRL-R in the porcine endometrial epithelial cells, and that the PRL-stimulated Cl secretion in the porcine endometrial epithelial cells was mediated through the short form of PRL receptors located predominately at the basolateral membrane.

Generally, PRL is synthesized by the decidualized endometrial stromal cells during the late secretory phase of the menstrual cycle and throughout pregnancy. The level of PRL is apparently much higher in the blood and amniotic fluid during pregnancy (Golander et al. 1978, Daly et al. 1983). After conception, a continuous increase in PRL production in the decidual cells leads to an accumulation of PRL in the amniotic fluid up to 2–3 μg/ml (Golander et al. 1978, Daly et al. 1983). Concomitantly, the PRL receptor expression and its mRNA are up-regulated toward the secretory phase of the menstrual cycle (Jabbour et al. 1998, Jones et al. 1998) and maintained throughout pregnancy (Maaskant et al. 1996). The level of PRL receptor mRNA is much higher in the glandular cells than in the stromal cells (Jabbour et al. 1998, Jones et al. 1998). Although the exact role of PRL in the human endometrium remains to be clarified, the pattern of secretion and expression supports a role of PRL in implantation and placentation. In agreement with those reports, the presence of PRL-R protein that was up-regulated by 17β-estradiol (Fig. 6) in the cultured porcine endometrial epithelial cells strongly suggested the physiological role of PRL in pregnancy. This speculation was consistent with the report that the blastocyst implantation and the maintenance of pregnancy were impaired in the PRL and PRL-R knockout mice (Jikihara et al. 1996). The next question is what is the exact role of PRL in the pregnant uterus. It is known that specific concentrations of electrolytes and pH within the uterine lumen are important for implantation and embryo development. In the rhesus monkey, PRL has been shown to regulate the amniotic and fetal extracellular fluid and electrolyte balance by decreasing the water flux from the amniotic side of the fetal membrane (Josimovich et al. 1977). Concomitant with the present finding of PRL role in the stimulation of anion secretion across the endometrial epithelial cells, and that active secretion of Cl and provides the driving force for fluid secretion and the regulation of luminal fluid pH, it is likely that PRL exerts endocrine and paracrine actions to regulate the volume and composition of the fluid within the uterine cavity, thus providing an optimal condition for implantation and development of the embryo.

In conclusion, this study shows for the first time the regulation of the transepithelial anion secretion by PRL in the endometrium. The results showed that PRL acutely stimulated anion secretion across the porcine endometrial epithelial cells through the short isoform of PRL receptor and the JAK–STAT-dependent pathway. The PRL-stimulated anion secretion was mostly a result of the activation of DPC- and NPPB-sensitive Cl channels, and bumetanide-sensitive Na+–K+–2Cl cotransport. Further investigation is required to define the physiological and pharmacological significance of PRL action in the endometrium. In addition, the PRL-signaling mechanisms, i.e. intracellular Ca2+, cAMP, or other signaling molecules remain to be elucidated.

Acknowledgements

The authors wish to thank Miss Norathee Buathong for her help with primary cell preparation and some of the experiments, and Dr Narattaphol Charoenphandhu for his valuable comments of the manuscript. This work was fully supported by the Thailand Research Fund (Contract grant number: TRA4780008) awarded to N M. The authors declare that there is no conflict of interest that would prejudice the impartiality of this scientific work.

References

  • Anderson MP, Sheppard DN, Berger HA & Welsh MJ 1992 Chloride channels in the apical membrane of normal and cystic fibrosis airway and intestinal epithelia. American Journal of Physiology 263 L1L14.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Arbogast LA & Voogt JL 1998 Endogenous opioid peptides contribute to suckling-induced prolactin release by suppressing tyrosine hydroxylase activity and messenger ribonucleic acid levels in tuberoinfundibular dopaminergic neurons. Endocrinology 139 28572862.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bern HA 1975 Prolactin and osmoregulation. American Zoologists 15 937949.

  • Bisbee CA, Machen TE & Bern HA 1979 Mouse mammary epithelial cells on floating collagen gels: transepithelial ion transport and effects of PRL. PNAS 76 536540.

  • Bole-Feysot C, Goffin V, Edery M, Binart N & Kelly PA 1998 Prolactin (PRL) and its receptor: actions, signal transduction pathways and phenotypes observed in PRL receptor knockout mice. Endocrine Reviews 19 225268.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chan HC, Liu CQ, Fong SK, Law SH, Wu LJ, So E, Chung YW, Ko WH & Wong PYD 1997 Regulation of Cl secretion by extracellular ATP in cultured mouse endometrial epithelium. Journal of Membrane Biology 156 4552.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Charoenphandhu N & Krishnamra N 2007 Prolactin is an important regulator of intestinal calcium transport. Canadian Journal of Physiology and Pharmacology 85 569581.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Clevenger CV & Kline JB 2001 Prolactin receptor signal transduction. Lupus 10 706718.

  • Daly DC, Maslar IA & Riddick DH 1983 Prolactin production during in vitro decidualization of proliferative endometrium. American Journal of Obstetrics and Gynecology 145 672678.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Deachapunya C & O'Grady SM 1998 Regulation of chloride secretion across porcine endometrial epithelial cells by prostaglandin E2. Journal of Physiology 508 3147.

  • Deachapunya C & O'Grady SM 2001 EGF regulates the transition from basal sodium absorption to anion secretion in cultured endometrial epithelial cells. Journal of Cell Physiology 186 243250.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Deachapunya C, Palmer-Densmore M & O'Grady SM 1999 Insulin stimulates transepithelial sodium transport by activation of a protein phosphatase that increases Na-K ATPase activity in endometrial epithelial cells. Journal of General Physiology 114 561574.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fong SK, Liu CQ & Chan HC 1998 Cellular mechanisms of adrenaline-stimulated anion secretion by the mouse endometrium epithelium. Biology of Reproduction 59 13421348.

  • Freeman ME, Kanyicsky B, Lerant A & Nagy G 2000 Prolactin: structure, function, and regulation of secretion. Physiological Reviews 80 15231631.

  • Golander A, Hurley T, Barrett J, Hizi A & Handwerger S 1978 Prolactin synthesis by human chorion decidual tissue: a possible source of prolactin in the amniotic fluid. Science 202 311313.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gubbay O, Critchley HOD, Bowen JM, King A & Jabbour HN 2002 Prolactin induces ERK phosphorylation in epithelial and CD56+ natural killer cells of the human endometrium. Journal of Clinical Endocrinology and Metabolism 87 23292335.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gunes H & Mastro AM 1996 Prolectin receptor gene expression in rat splenocytes and thymocytes from birth to adulthood. Molecular and Cellular Endocrinology 117 4152.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Handwerger S & Freemark M 1987 Role of placental lactogen and prolactin in human pregnancy. Advances in Experimental Medicine and Biology 219 399420.

  • Illek B, Tam AW, Fischer H & Machen TE 1999 Anion selectivity of apical membrane conductance of Calu 3 human airway epithelium. Pflügers Archiv 437 812822.

  • Jabbour HN, Critchley HO & Boddy SC 1998 Expression of functional prolactin receptors in nonpregnant human endometrium: janus kinase-2, signal transducer and activator of transcription-1 (STAT1), and STAT5 proteins are phosphorylated after stimulation with prolactin. Journal of Clinical Endocrinology and Metabolism 83 25452553.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jahn GA, Edery M, Belair L, Kelly PA & Djiane J 1991 Prolactin receptor gene expression in rat mammary gland and liver during pregnancy and lactation. Endocrinology 128 29762984.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jantarajit W, Thongon N, Pandaranandaka J, Teerapornpuntakit J, Krishnamra N & Charoenphandhu N 2007 Prolactin-stimulated transepithelial calcium transport in duodenum and Caco-2 monolayer are mediated by the phosphoinositide 3-kinase pathway. American Journal of Physiology. Endocrinology and Metabolism 293 372384.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jikihara H, Kessler CA, Cedars MI & Bra AK 1996 Up-regulation of the human prolactin receptor in the endometrium. Journal of Endocrinology 5 157162.

  • Jones RL, Critchley HO, Brooks J, Jabbour HN & McNeilly AS 1998 Localization and temporal expression of prolactin recptor in human endometrium. Journal of Clinical Endocrinology and Metabolism 83 258262.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Josimovich JB, Merisko K & Boccella L 1977 Amniotic prolactin control over amniotic and fetal extracellular fluid water and electrolytes in the rhesus monkey. Endocrinology 100 564570.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kelly PA, Djiane J, Postel-Vinay MC & Edery M 1991 The prolactin/growth hormone receptor family. Endocrine Reviews 12 235251.

  • Linzell JL, Peaker M & Taylor JC 1975 The effects of prolactin and oxytocin on milk secretion and on the permeability of the mammary epithelium in the rabbit. Journal of Physiology 253 547563.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Maaskant RA, Bogic LV, Gilger S, Kelly PA & Bryant-Greenwood GD 1996 The human prolactin receptor in the fetal membranes, decidua, and placenta. Journal of Clinical Endocrinology Metabolism 81 396405.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mainoya JR, Bern HA & Regan JW 1974 Influence of ovine prolactin on transport of fluid and sodium chloride by the mammalian intestine and gall bladder. Journal of Endocrinology 63 311317.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Matthews CJ, Thomas EJ, Redfern CPF & Hirst BH 1993 Ion transport by human endometrium in vitro. Human Reproduction 8 15101575.

  • Palmer-Densmore M, Deachapunya C & O'Grady SM 2002 UTP-dependent inhibition of Na absorption requires activation of PKC in endometrial epithelial cells. Journal of General Physiology 120 897906.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sakamoto T & McCormick SD 2006 Prolactin and growth hormone in fish osmoregulation. General and Comparative Endocrinology 147 2430.

  • Selvaraj NG, Omi E, Gibori G & Rao MC 2000 Janus kinase 2 (JAK2) regulates prolactin-mediated chloride transport in mouse mammary epithelial cells through tyrosine phosphorylation of Na+–K+–2Cl cotransporter. Molecular Endocrinology 14 20542065.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Shingo T, Gregg C, Enwere E, Fujikawa H, Hassam R, Geary C, Cross JC & Weiss S 2003 Pregnancy-stimulated neurogenesis in the adult female forebrain mediated by prolactin. Science 299 117120.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Telleria CM, Parmer TG, Zhong L, Clarke DL, Albarracin CT, Duan WR, Linzer DI & Gibori G 1997 The different forms of the prolactin receptor in the rat corpus luteum: developmental expression and hormonal regulation in pregnancy. Endocrinology 138 48124820.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Tseng L & Zhu HH 1998 Progestin induces prolactin receptor in human endometrial stromal cells. Journal of the Society for Gynecologic Investigation 5 149155.

  • Uribe JM, Keely SJ, Traynor-Kaplan AE & Barrett KE 1996 Phosphatidylinositol 3-kinase mediates the inhibitory effect of epidermal growth factor on calcium-dependent chloride secretion. Journal of Biological Chemistry 271 2658826595.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Vetter AE & O'Grady SM 1996 Mechanisms of electrolyte transport across the endometrium I. Regulation by PGF2 and cAMP. American Journal of Physiology. Cell Physiology 270 663672.

    • PubMed
    • Search Google Scholar
    • Export Citation

 

  • Collapse
  • Expand
  • Effect of PRL on the basal Isc in the endometrial epithelial monolayers. (A) A representative Isc tracing responded to an addition of 1 μg/ml PRL to (a) the apical solution followed by 1 and 5 μg/ml PRL added to (b) the basolateral solution. (B) Concentration–response relationships showing the increase in Isc following basolateral treatment with various concentrations of PRL. The EC50 value was 120 ng/ml (n=6).

  • Effect of ion channel blockers on the PRL-stimulated Isc. (A) A representative Isc tracing showing that an apical addition of amiloride (Amil, 10 μM) produced a small decrease in basal Isc. A subsequent addition of PRL (1 μg/ml) into the basolateral solution produced an increase in Isc. (B) An apical addition of NPPB (100 μM), (C) DPC (1 mM), or (D) DIDS (200 μM) decreased the basal Isc and reduced the PRL response. (E) Bar graph illustrating the average maximal increases in Isc response produced by prolactin alone (control) and in the presence of amiloride, NPPB, DPC, or DIDS. Values represent means±s.e.m. *P<0.01 when compared with the control value by ANOVA.

  • Effect of Na+–K+–2Cl cotransporter blockers on the PRL-stimulated Isc. (A) A representative Isc tracing showing that a basolateral addition of bumetanide (200 μM) produced a decrease in basal Isc. A subsequent addition of PRL (1 μg/ml) into the basolateral solution produced a slightly increase in Isc. (B) Bar graph illustrating the average maximal increases in Isc response produced by prolactin alone (control) and in the presence of bumetanide. Values represent means±s.e.m. *P<0.01 when compared with the control value by Student's t-test.

  • Effects of anion substitution on the maximal Isc response to PRL. In standard Ringer's solution, PRL (1 μg/ml) produced a mean increase in Isc of 11.44±1.22 μA (n=12). Replacement of Cl and in both the apical and basolateral solutions significantly inhibited the maximal Isc response to PRL by 86% (n=4) and 92% (n=4) respectively. *P<0.01 when compared with the control value by ANOVA.

  • Effect of JAK2 activity inhibitor on the PRL-stimulated Isc. (A) A representative Isc tracing showing that an addition of AG490 (50 μM), an inhibitor of JAK2 activity, to both apical and basolateral solutions completely inhibited both basal and PRL-induced increase in Isc. (B) Bar graph illustrating the average maximal increases in Isc response produced by PRL (1 μg/ml), 8cpt-cAMP (cAMP, 100 μM), or prostaglandin E2 (PGE, 3 μM) in the absence and presence of AG490 (50 μM). Values represent means ±s.e.m. *P<0.01 when compared with the corresponding control value by Student's t-test.

  • Expression of PRL receptor protein in porcine endometrial epithelial cells. (A) Western blot analysis of PRL receptor (PRL-R). A 36 kDa band of PRL-R was detected in human mammary gland cancer cell MCF-7, human endometrial cancer cell RL-95, and porcine endometrial epithelial cells (PE) under standard medium (SM), serum-free medium (SF) alone, or supplemented with 10−8 M 17β-estradiol (SF+E 10 nM). The internal control of 43 kDa band of β-actin was also detected in all samples. (B) Bar graph illustrating the ratio of PRL-R to β-actin protein expression in the endometrial epithelial cells, based on the densitometry of immunoblots obtained from SM, SF, and SF+E10 nM (n=4). Values represent means±s.e.m. *P<0.05 when compared with the serum-free medium condition by Student's t-test.

  • Anderson MP, Sheppard DN, Berger HA & Welsh MJ 1992 Chloride channels in the apical membrane of normal and cystic fibrosis airway and intestinal epithelia. American Journal of Physiology 263 L1L14.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Arbogast LA & Voogt JL 1998 Endogenous opioid peptides contribute to suckling-induced prolactin release by suppressing tyrosine hydroxylase activity and messenger ribonucleic acid levels in tuberoinfundibular dopaminergic neurons. Endocrinology 139 28572862.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Bern HA 1975 Prolactin and osmoregulation. American Zoologists 15 937949.

  • Bisbee CA, Machen TE & Bern HA 1979 Mouse mammary epithelial cells on floating collagen gels: transepithelial ion transport and effects of PRL. PNAS 76 536540.

  • Bole-Feysot C, Goffin V, Edery M, Binart N & Kelly PA 1998 Prolactin (PRL) and its receptor: actions, signal transduction pathways and phenotypes observed in PRL receptor knockout mice. Endocrine Reviews 19 225268.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Chan HC, Liu CQ, Fong SK, Law SH, Wu LJ, So E, Chung YW, Ko WH & Wong PYD 1997 Regulation of Cl secretion by extracellular ATP in cultured mouse endometrial epithelium. Journal of Membrane Biology 156 4552.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Charoenphandhu N & Krishnamra N 2007 Prolactin is an important regulator of intestinal calcium transport. Canadian Journal of Physiology and Pharmacology 85 569581.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Clevenger CV & Kline JB 2001 Prolactin receptor signal transduction. Lupus 10 706718.

  • Daly DC, Maslar IA & Riddick DH 1983 Prolactin production during in vitro decidualization of proliferative endometrium. American Journal of Obstetrics and Gynecology 145 672678.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Deachapunya C & O'Grady SM 1998 Regulation of chloride secretion across porcine endometrial epithelial cells by prostaglandin E2. Journal of Physiology 508 3147.

  • Deachapunya C & O'Grady SM 2001 EGF regulates the transition from basal sodium absorption to anion secretion in cultured endometrial epithelial cells. Journal of Cell Physiology 186 243250.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Deachapunya C, Palmer-Densmore M & O'Grady SM 1999 Insulin stimulates transepithelial sodium transport by activation of a protein phosphatase that increases Na-K ATPase activity in endometrial epithelial cells. Journal of General Physiology 114 561574.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Fong SK, Liu CQ & Chan HC 1998 Cellular mechanisms of adrenaline-stimulated anion secretion by the mouse endometrium epithelium. Biology of Reproduction 59 13421348.

  • Freeman ME, Kanyicsky B, Lerant A & Nagy G 2000 Prolactin: structure, function, and regulation of secretion. Physiological Reviews 80 15231631.

  • Golander A, Hurley T, Barrett J, Hizi A & Handwerger S 1978 Prolactin synthesis by human chorion decidual tissue: a possible source of prolactin in the amniotic fluid. Science 202 311313.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gubbay O, Critchley HOD, Bowen JM, King A & Jabbour HN 2002 Prolactin induces ERK phosphorylation in epithelial and CD56+ natural killer cells of the human endometrium. Journal of Clinical Endocrinology and Metabolism 87 23292335.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Gunes H & Mastro AM 1996 Prolectin receptor gene expression in rat splenocytes and thymocytes from birth to adulthood. Molecular and Cellular Endocrinology 117 4152.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Handwerger S & Freemark M 1987 Role of placental lactogen and prolactin in human pregnancy. Advances in Experimental Medicine and Biology 219 399420.

  • Illek B, Tam AW, Fischer H & Machen TE 1999 Anion selectivity of apical membrane conductance of Calu 3 human airway epithelium. Pflügers Archiv 437 812822.

  • Jabbour HN, Critchley HO & Boddy SC 1998 Expression of functional prolactin receptors in nonpregnant human endometrium: janus kinase-2, signal transducer and activator of transcription-1 (STAT1), and STAT5 proteins are phosphorylated after stimulation with prolactin. Journal of Clinical Endocrinology and Metabolism 83 25452553.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jahn GA, Edery M, Belair L, Kelly PA & Djiane J 1991 Prolactin receptor gene expression in rat mammary gland and liver during pregnancy and lactation. Endocrinology 128 29762984.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jantarajit W, Thongon N, Pandaranandaka J, Teerapornpuntakit J, Krishnamra N & Charoenphandhu N 2007 Prolactin-stimulated transepithelial calcium transport in duodenum and Caco-2 monolayer are mediated by the phosphoinositide 3-kinase pathway. American Journal of Physiology. Endocrinology and Metabolism 293 372384.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Jikihara H, Kessler CA, Cedars MI & Bra AK 1996 Up-regulation of the human prolactin receptor in the endometrium. Journal of Endocrinology 5 157162.

  • Jones RL, Critchley HO, Brooks J, Jabbour HN & McNeilly AS 1998 Localization and temporal expression of prolactin recptor in human endometrium. Journal of Clinical Endocrinology and Metabolism 83 258262.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Josimovich JB, Merisko K & Boccella L 1977 Amniotic prolactin control over amniotic and fetal extracellular fluid water and electrolytes in the rhesus monkey. Endocrinology 100 564570.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Kelly PA, Djiane J, Postel-Vinay MC & Edery M 1991 The prolactin/growth hormone receptor family. Endocrine Reviews 12 235251.

  • Linzell JL, Peaker M & Taylor JC 1975 The effects of prolactin and oxytocin on milk secretion and on the permeability of the mammary epithelium in the rabbit. Journal of Physiology 253 547563.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Maaskant RA, Bogic LV, Gilger S, Kelly PA & Bryant-Greenwood GD 1996 The human prolactin receptor in the fetal membranes, decidua, and placenta. Journal of Clinical Endocrinology Metabolism 81 396405.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Mainoya JR, Bern HA & Regan JW 1974 Influence of ovine prolactin on transport of fluid and sodium chloride by the mammalian intestine and gall bladder. Journal of Endocrinology 63 311317.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Matthews CJ, Thomas EJ, Redfern CPF & Hirst BH 1993 Ion transport by human endometrium in vitro. Human Reproduction 8 15101575.

  • Palmer-Densmore M, Deachapunya C & O'Grady SM 2002 UTP-dependent inhibition of Na absorption requires activation of PKC in endometrial epithelial cells. Journal of General Physiology 120 897906.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Sakamoto T & McCormick SD 2006 Prolactin and growth hormone in fish osmoregulation. General and Comparative Endocrinology 147 2430.

  • Selvaraj NG, Omi E, Gibori G & Rao MC 2000 Janus kinase 2 (JAK2) regulates prolactin-mediated chloride transport in mouse mammary epithelial cells through tyrosine phosphorylation of Na+–K+–2Cl cotransporter. Molecular Endocrinology 14 20542065.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Shingo T, Gregg C, Enwere E, Fujikawa H, Hassam R, Geary C, Cross JC & Weiss S 2003 Pregnancy-stimulated neurogenesis in the adult female forebrain mediated by prolactin. Science 299 117120.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Telleria CM, Parmer TG, Zhong L, Clarke DL, Albarracin CT, Duan WR, Linzer DI & Gibori G 1997 The different forms of the prolactin receptor in the rat corpus luteum: developmental expression and hormonal regulation in pregnancy. Endocrinology 138 48124820.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Tseng L & Zhu HH 1998 Progestin induces prolactin receptor in human endometrial stromal cells. Journal of the Society for Gynecologic Investigation 5 149155.

  • Uribe JM, Keely SJ, Traynor-Kaplan AE & Barrett KE 1996 Phosphatidylinositol 3-kinase mediates the inhibitory effect of epidermal growth factor on calcium-dependent chloride secretion. Journal of Biological Chemistry 271 2658826595.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • Vetter AE & O'Grady SM 1996 Mechanisms of electrolyte transport across the endometrium I. Regulation by PGF2 and cAMP. American Journal of Physiology. Cell Physiology 270 663672.

    • PubMed
    • Search Google Scholar
    • Export Citation