Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters










Publication year range
1.
Placenta ; 31(4): 327-33, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20149452

ABSTRACT

OBJECTIVE: Wingless-type mouse mammary tumor virus integration site family, member 5A (WNT5A), is expressed in mouse decidua and is thought to play an important role in decidualization. We examined expression of the receptor for WNT5A, receptor tyrosine kinase-like orphan receptor 2 (ROR2), in the uteri of cycling and pregnant mice. STUDY DESIGN: Reverse transcription (RT)-PCR and immunohistochemistry were performed. RESULTS: RT-PCR revealed that transcripts for Ror2, Wnt3a, Wnt5a and inhibitor of WNT signaling, Dickkopf homolog 1 (Dkk1), were present in the pregnant uterus. Immunohistochemistry revealed that in the virgin uterus, ROR2 is expressed in stromal cells and on the basal side of uterine gland and endometrial epithelial cells. During pregnancy, both the luminal and basal side of uterine gland epithelial cells expressed ROR2, stromal cell expression of ROR2 became more frequent and ROR2 expressing uterine Natural Killer (NK) cells and cells lining the maternal vascular space emerged. Immunofluorescence imaging and flow cytometry revealed that although uterine NK cells expressed ROR2, NK cells of the spleen were ROR2 negative. CONCLUSION: The expression of ROR2 by endometrial epithelial cells may suggest WNT signaling has roles in uterine epithelial cell polarity or implantation. Expression of ROR2 by uterine NK cells may suggest WNT signaling regulates uterine NK cell functions such angiogenesis and regulation of trophoblast migration. In summary, our results show that ROR2 expression by maternal uterine cells is influenced by pregnancy.


Subject(s)
Receptor Tyrosine Kinase-like Orphan Receptors/biosynthesis , Uterus/metabolism , Animals , Female , Intercellular Signaling Peptides and Proteins/genetics , Killer Cells, Natural/metabolism , Mice , Pregnancy , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Wnt Proteins/genetics , Wnt-5a Protein , Wnt3 Protein , Wnt3A Protein
2.
Neurogastroenterol Motil ; 21(8): 880-e64, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19239624

ABSTRACT

Visceral inflammation evokes hyperexcitability in nociceptive dorsal root ganglia (DRG) neurons and these changes are associated with increased voltage-gated sodium channel (Na(v)) 1.8 current density, but the molecular determinants of these changes are unclear. This study used Western blotting to measure changes in Na(v) 1.7, 1.8 and 1.9 protein expression during trinitrobenzenesulphonic acid (TNBS) colitis and quantitative polymerase chain reaction (PCR) to examine corresponding changes in mRNA. Colonic neurons were labelled with the retrograde tracer Fast Blue injected into the wall of the distal colon and quantitative PCR performed on laser-captured labelled colonic neurons from ganglia at T9-13 or unlabelled DRG neurons from the upper spinal cord. Immunohistochemistry and western blots were performed on whole DRG from the same sites. Fast Blue-labelled neurons demonstrated Na(v) 1.7, 1.8 and 1.9 immunoreactivity. On day 7 of colitis, which correlated with electrophysiological studies, there was a threefold increase in Na(v) 1.8 protein in ganglia from T9 to 13, but Na(v) 1.7 and 1.9 levels were unchanged. There was no corresponding change in the Na(v) 1.8 alpha-subunit mRNA levels. However, on days 2 and 4, Na(v) 1.8 mRNA was decreased 10-fold. Na(v) 1.8 protein and mRNA levels were unchanged in neurons isolated from ganglia in the upper spinal cord, where colonic neurons are not found. These findings suggest that the TNBS evoked increase in Na(v) 1.8 currents is associated with increased numbers of channels. The absence of corresponding changes in transcript suggests a translational or post-translational mechanism, but the 10-fold recovery of transcript preceding this time point also demonstrates a complex transcriptional regulation.


Subject(s)
Colitis/chemically induced , Ganglia, Spinal/cytology , Neurons/metabolism , Sodium Channels/metabolism , Trinitrobenzenesulfonic Acid/toxicity , Animals , Colitis/physiopathology , Colon/innervation , Colon/metabolism , Colon/pathology , Mice , NAV1.8 Voltage-Gated Sodium Channel , NAV1.9 Voltage-Gated Sodium Channel , Neuropeptides/genetics , Neuropeptides/metabolism , Protein Subunits/genetics , Protein Subunits/metabolism , Sodium Channels/genetics
3.
Gut ; 58(2): 166-73, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18978176

ABSTRACT

BACKGROUND: Oesophagitis is characterised by basal cell hyperplasia and activated eosinophils, which release mediators including major basic protein (MBP). MBP and its mimetic polyarginine activate the calcium sensing receptor (CaSR) on oesophageal epithelium. Fibroblast growth factor 9 (FGF9) is implicated in epithelial homeostasis and proliferative response to injury, but has not been characterised in the oesophagus. OBJECTIVE: To characterise FGF9 in oesophageal epithelium and oesophagitis, as the result of MBP activation of the CaSR. METHODS: Human oesophageal epithelial cells (HET-1A) were used to compare affects of calcium, polyarginine and MBP-peptide on FGF9. HET-1A were transfected with interfering RNA (siRNA(CaSR)). FGF9, FGF receptors 2 and 3, bone morphogenetic protein (BMP)-2, BMP-4 and noggin mRNA expression were detected by reverse transcriptase polymerase chain reaction. FGF9 was measured from HET-1A and from normal, gastro-oesophageal reflux and eosinophilic oesophagitis (EoE) patient biopsies using ELISA and immunohistochemistry. HET-1A proliferation was studied using bromodeoxyuridine and MTT. RESULTS: FGF9 was secreted by HET-1A cells treated with polyarginine and MBP-peptide, but not calcium. This effect was abrogated by siRNA(CaSR). FGF9 receptor mRNA was present. HET-1A cells proliferated following rhFGF9, but not MBP-peptide treatment, and rhFGF9 altered transcription of downstream proliferation-related genes (noggin, BMP-2 and BMP-4). FGF9 was increased in biopsies from patients with eosinophilic oesophagitis, which correlated with basal hyperplasia. CONCLUSION: Eosinophil-released MBP acts on the CaSR to increase FGF9 in oesophageal epithelial cells, leading to proliferation. Increased FGF9 is found in biopsies of EoE patients and may play a role in the pathogenesis of oesophagitis.


Subject(s)
Eosinophilia/metabolism , Esophagitis/immunology , Esophagus/immunology , Fibroblast Growth Factor 9/pharmacology , Adolescent , Bone Morphogenetic Proteins/genetics , Calcium/metabolism , Calcium/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Child , Child, Preschool , Eosinophil Major Basic Protein/metabolism , Eosinophil Major Basic Protein/pharmacology , Eosinophilia/pathology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Esophagitis/metabolism , Esophagitis/pathology , Esophagus/metabolism , Esophagus/pathology , Female , Fibroblast Growth Factor 9/analysis , Fibroblast Growth Factor 9/genetics , Humans , Male , Peptides/metabolism , Peptides/pharmacology , RNA, Messenger/analysis , RNA, Small Interfering/pharmacology , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 3/genetics , Receptors, Calcium-Sensing/metabolism , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction/methods , Transcription, Genetic/drug effects
4.
Am J Physiol Endocrinol Metab ; 285(2): E329-37, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12700162

ABSTRACT

Elevated extracellular calcium ([Ca2+]o) and other agonists potentially acting via the calcium-sensing receptor (CaR) increase parathyroid hormone-related peptide (PTHrP) release from H-500 Leydig cells. Here, we provide strong evidence for the CaR's involvement by using a dominant negative CaR that attenuates high [Ca2+]o-induced PTHrP release. This effect is likely transcriptional, because high [Ca2+]o upregulates the PTHrP transcript, an effect that is abolished by actinomycin D. Regulation of PTHrP release by the CaR involves activation of PKC as well as ERK1/2, p38 MAPK, and JNK pathways. However, we show for the first time that high [Ca2+]o-induced activation of the stress-activated protein kinase SEK1 is PKC independent, because there is an additive effect of a PKC inhibitor in combination with the JNK inhibitor on [Ca2+]o-stimulated PTHrP release. Furthermore, high [Ca2+]o, in a PKC-independent fashion, induces phosphorylation of ERK1/2, SEK1, p38 MAPK, and its downstream transcription factor ATF-2. We conclude that CaR regulation of PTHrP release in H-500 cells involves activation of PKC as well as the ERK1/2, p38 MAPK, and JNK pathways.


Subject(s)
JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Peptide Hormones/metabolism , Protein Kinase C/metabolism , Receptors, Cell Surface/physiology , Animals , Calcium/pharmacology , Enzyme Activation/drug effects , Leydig Cell Tumor , MAP Kinase Kinase 4 , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Parathyroid Hormone-Related Protein , Phosphorylation , Rats , Rats, Inbred F344 , Receptors, Calcium-Sensing , Tumor Cells, Cultured , p38 Mitogen-Activated Protein Kinases
5.
Am J Physiol Endocrinol Metab ; 284(1): E219-27, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12388161

ABSTRACT

Several mesenchymally derived cells, including osteoblasts, secrete hepatocyte growth factor (HGF). 1alpha,25(OH)(2)-vitamin D(3) [1,25(OH)(2)D(3)] inhibits proliferation and induces differentiation of MG-63 osteoblastic cells. Here we show that MG-63 cells secrete copious amounts of HGF and that 1,25(OH)(2)D(3) inhibits HGF production. MG-63 cells also express HGF receptor (c-Met) mRNA, suggesting an autocrine action of HGF. Indeed, although exogenous HGF failed to stimulate cellular proliferation, neutralizing endogenous HGF with a neutralizing antibody inhibited MG-63 cell proliferation; moreover, inhibiting HGF synthesis with 1,25(OH)(2)D(3) followed by addition of HGF rescued hormone-induced inhibition of proliferation. Nonneutralized cells displayed constitutive phosphorylation of c-Met and the mitogen-activated protein kinases mitogen/extracellular signal-regulated kinase (MEK) 1 and extracellular signal-regulated kinase (Erk) 1/2, which were inhibited by anti-HGF antibody. Constitutive phosphorylation of Erk1/2 was also abolished by 1,25(OH)(2)D(3). Addition of HGF to MG-63 cells treated with neutralizing HGF antibody induced rapid phosphorylation of c-Met, MEK1, and Erk1/2. Thus endogenous HGF induces a constitutively active, autocrine mitogenic loop in MG-63 cells. The known antiproliferative effect of 1,25(OH)(2)D(3) on MG-63 cells can be accounted for by the concomitant 1,25(OH)(2)D(3)-induced inhibition of HGF production.


Subject(s)
Calcitriol/pharmacology , Hepatocyte Growth Factor/biosynthesis , Hepatocyte Growth Factor/metabolism , Osteosarcoma/metabolism , Antibodies/pharmacology , Cell Differentiation/drug effects , Cell Division/drug effects , Gene Expression , Hepatocyte Growth Factor/immunology , Humans , Kinetics , MAP Kinase Kinase 1 , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Osteoblasts/pathology , Osteosarcoma/pathology , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Tumor Cells, Cultured
6.
J Biol Chem ; 276(37): 34880-7, 2001 Sep 14.
Article in English | MEDLINE | ID: mdl-11390380

ABSTRACT

The G protein-coupled, extracellular calcium-sensing receptor (CaR) regulates parathyroid hormone secretion and parathyroid cellular proliferation as well as the functions of diverse other cell types. The CaR resides in caveolae-plasma membrane microdomains containing receptors and associated signaling molecules that are thought to serve as cellular "message centers." An additional mechanism for coordinating cellular signaling is the presence of scaffold proteins that bind and organize components of signal transduction cascades. With the use of the yeast two-hybrid system, we identified filamin-A (an actin-cross-linking, putative scaffold protein that binds mitogen-activated protein kinase (MAPK) components activated by the CaR) as an intracellular binding partner of the CaR's carboxyl (COOH)-terminal tail. A direct interaction of the two proteins was confirmed by an in vitro binding assay. Moreover, confocal microscopy combined with two color immunofluorescence showed co-localization of the CaR and filamin-A within parathyroid cells as well as HEK-293 cells stably transfected with the CaR. Deletion mapping localized the sites of interaction between the two proteins to a stretch of 60 amino acid residues within the distal portion of the CaR's COOH-terminal tail and domains 14 and 15 in filamin-A, respectively. Finally, introducing the portion of filamin-A interacting with the CaR into CaR-transfected HEK-293 cells using protein transduction with a His-tagged, Tat-filamin-A fusion protein nearly abolished CaR-mediated activation of ERK1/2 MAPK but had no effect on ERK1/2 activity stimulated by ADP. Therefore, the binding of the CaR's COOH-terminal tail to filamin-A may contribute to its localization in caveolae, link it to the actin-based cytoskeleton, and participate in CaR-mediated activation of MAPK.


Subject(s)
Contractile Proteins/metabolism , Microfilament Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Receptors, Cell Surface/physiology , Adenosine Diphosphate/pharmacology , Amino Acid Sequence , Cell Line , Contractile Proteins/analysis , Enzyme Activation , Filamins , Humans , Immunohistochemistry , Microfilament Proteins/analysis , Molecular Sequence Data , Receptors, Calcium-Sensing , Receptors, Cell Surface/analysis
7.
Am J Physiol Renal Physiol ; 280(2): F291-302, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11208605

ABSTRACT

Regulation of the extracellular signal-regulated kinase 1 and 2 (ERK1/2) pathway by the extracellular calcium (Ca2+o)-sensing receptor (CaR) was investigated in bovine parathyroid and CaR-transfected human embryonic kidney (HEKCaR) cells. Elevating Ca2+o or adding the selective CaR activator NPS R-467 elicited rapid, dose-dependent phosphorylation of ERK1/2. These phosphorylations were attenuated by pretreatment with pertussis toxin (PTX) or by treatment with the phosphotyrosine kinase (PTK) inhibitors genistein and herbimycin, the phosphatidylinositol-specific phospholipase C (PI-PLC) inhibitor U-73122, or the protein kinase C (PKC) inhibitor GF109203X and were enhanced by the PKC activator phorbol 12-myristate 13-acetate. Combined treatment with PTX and inhibitors of both PKC and PTK nearly abolished high Ca2+o-evoked ERK1/2 activation in HEKCaR cells, demonstrating CaR-mediated coupling via both Gq and G(i). High Ca2+o increased serine phosphorylation of the 85-kDa cytosolic phospholipase A2 (cPLA2) in both parathyroid and HEKCaR cells. The selective mitogen-activated protein kinase (MAPK) inhibitor PD98059 abolished high-Ca2+o)-induced ERK1/2 activation and reduced cPLA2 phosphorylation in both cell types, documenting MAPK's role in cPLA2 activation. Thus our data suggest that the CaR activates MAPK through PKC, presumably through Gq/11-mediated activation of PI-PLC, as well as through G(i)- and PTK-dependent pathway(s) in bovine parathyroid and HEKCaR cells and indicate the importance of MAPK in cPLA2 activation.


Subject(s)
Calcium/metabolism , Kidney/metabolism , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Parathyroid Glands/metabolism , Receptors, Cell Surface/metabolism , Animals , Calcium/pharmacology , Cattle , Enzyme Inhibitors/pharmacology , Humans , Kidney/cytology , Kidney/drug effects , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 1/drug effects , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/drug effects , Parathyroid Glands/cytology , Parathyroid Glands/drug effects , Pertussis Toxin , Phospholipases A/drug effects , Phospholipases A/metabolism , Phospholipases A2 , Phosphorylation , Receptors, Calcium-Sensing , Receptors, Cell Surface/drug effects , Virulence Factors, Bordetella/pharmacology
8.
Physiol Rev ; 81(1): 239-297, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11152759

ABSTRACT

The cloning of a G protein-coupled extracellular Ca(2+) (Ca(o)(2+))-sensing receptor (CaR) has elucidated the molecular basis for many of the previously recognized effects of Ca(o)(2+) on tissues that maintain systemic Ca(o)(2+) homeostasis, especially parathyroid chief cells and several cells in the kidney. The availability of the cloned CaR enabled the development of DNA and antibody probes for identifying the CaR's mRNA and protein, respectively, within these and other tissues. It also permitted the identification of human diseases resulting from inactivating or activating mutations of the CaR gene and the subsequent generation of mice with targeted disruption of the CaR gene. The characteristic alterations in parathyroid and renal function in these patients and in the mice with "knockout" of the CaR gene have provided valuable information on the CaR's physiological roles in these tissues participating in mineral ion homeostasis. Nevertheless, relatively little is known about how the CaR regulates other tissues involved in systemic Ca(o)(2+) homeostasis, particularly bone and intestine. Moreover, there is evidence that additional Ca(o)(2+) sensors may exist in bone cells that mediate some or even all of the known effects of Ca(o)(2+) on these cells. Even more remains to be learned about the CaR's function in the rapidly growing list of cells that express it but are uninvolved in systemic Ca(o)(2+) metabolism. Available data suggest that the receptor serves numerous roles outside of systemic mineral ion homeostasis, ranging from the regulation of hormonal secretion and the activities of various ion channels to the longer term control of gene expression, programmed cell death (apoptosis), and cellular proliferation. In some cases, the CaR on these "nonhomeostatic" cells responds to local changes in Ca(o)(2+) taking place within compartments of the extracellular fluid (ECF) that communicate with the outside environment (e.g., the gastrointestinal tract). In others, localized changes in Ca(o)(2+) within the ECF can originate from several mechanisms, including fluxes of calcium ions into or out of cellular or extracellular stores or across epithelium that absorb or secrete Ca(2+). In any event, the CaR and other receptors/sensors for Ca(o)(2+) and probably for other extracellular ions represent versatile regulators of numerous cellular functions and may serve as important therapeutic targets.


Subject(s)
Calcium Signaling/physiology , Receptors, Cell Surface/metabolism , Alternative Splicing , Animals , Apoptosis/physiology , Calcium Signaling/drug effects , Cell Differentiation/physiology , Cell Division/physiology , GTP-Binding Proteins/metabolism , Gene Expression Regulation , Homeostasis/physiology , Humans , Organ Specificity , Polymorphism, Genetic , Receptors, Calcium-Sensing , Receptors, Cell Surface/genetics , Signal Transduction/physiology , Structure-Activity Relationship
9.
J Membr Biol ; 172(1): 47-58, 1999 Nov 01.
Article in English | MEDLINE | ID: mdl-10552013

ABSTRACT

We studied the relationship between changes in intracellular pH (pH(i)), intracellular Ca(2+)([Ca(2+)](i)) and charybdotoxin sensitive (CTX) maxi-K(+) channels occurring after modest 'physiological' swelling in guinea pig jejunal villus enterocytes. Villus cell volume was assessed by electronic cell sizing, and pH(i) and [Ca(2+)](i) by fluorescence spectroscopy with 2,7, biscarboxyethyl-5-6-carboxyfluorescein and Indo-1, respectively. In a slightly (0.93 x isotonic) hypotonic medium, villus cells swelled to the same size they would reach during d-glucose or l-alanine absorption; the subsequent Regulatory Volume Decrease (RVD) was prevented by CTX. After the large volume increase in a more hypotonic (0.80 x isotonic) medium, RVD was unaffected by CTX. After modest swelling associated with 0.93 x isotonic dilution, the pH(i) alkalinized but N-5-methyl-isobutyl amiloride (MIA) prevented this DeltapH(i) and the subsequent RVD. Even in the presence of MIA, alkalinization with added NH(4)Cl permitted complete RVD which could be inhibited by CTX. The rate of (86)Rb efflux which also increased after this 0.93 x isotonic dilution was inhibited an equivalent amount by CTX, MIA or Na(+)-free medium. Modest swelling transiently increased [Ca(2+)](i) and Ca(2+)-free medium or blocking alkalinization by MIA or Na(+)-free medium diminished this transient increase an equivalent amount. RVD after modest swelling was prevented in Ca(2+)-free medium but alkalinization still occurred. After large volume increases, alkalinization of cells increased [Ca(2+)](i) and volume changes became sensitive to CTX. We conclude that both alkalinization of pH(i) and increased [Ca(2+)](i) observed with 'physiological' volume increase are essential for the activation of CTX-sensitive maxi-K(+) channels required for RVD.


Subject(s)
Calcium/metabolism , Jejunum/cytology , Jejunum/metabolism , Potassium Channels/metabolism , Amiloride/analogs & derivatives , Amiloride/pharmacology , Animals , Cell Size , Charybdotoxin/pharmacology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Guinea Pigs , Hydrogen-Ion Concentration , In Vitro Techniques , Intracellular Fluid/metabolism , Jejunum/drug effects , Male , Potassium Channels/drug effects , Rubidium/pharmacokinetics
10.
J Membr Biol ; 172(1): 59-66, 1999 Nov 01.
Article in English | MEDLINE | ID: mdl-10552014

ABSTRACT

To assess the activation of the charybdotoxin-insensitive K(+) channel responsible for Regulatory Volume Decrease (RVD) after substantial volume increases, we measured intracellular pH (pH(i)), intracellular calcium ([Ca(2+)](i)) and inhibitors of kinases and phosphoprotein phosphatases in guinea pig jejunal villus enterocytes in response to volume changes. Fluorescence spectroscopy was used to measure pH(i) and [Ca(2+)](i) of cells in suspension, loaded with 2, 7,bis-carboxyethyl-5-6-carboxyfluorescein and Indo-1, respectively, and cell volume was assessed using electronic cell sizing. A modest 7% volume increase or substantial 15 to 20% volume increase caused [Ca(2+)](i) to increase proportionately but the 7% increase caused alkalinization while the larger increases resulted in acidification of approximately 0.14 pH units. Following a 15% volume increase, 1-N-0-bis (5-isoquinoline-sulfonyl)-N-methyl-l-4-phenyl-piperazine (KN-62, 50 microm), an inhibitor of Ca(2+)/calmodulin kinase II, blocked RVD. Gramicidin (0.5 microm bypassed this inhibition suggesting that the K(+) channel had been affected by the KN-62. RVD after a modest 7% volume increase was not influenced by KN-62 unless the cell was acidified. Okadaic acid, an inhibitor of phosphoprotein phosphatases 1 and 2A, accelerated RVD after a 20% volume increase; inhibition of RVD generated by increasing the K(+) gradient was bypassed by okadaic acid. Tyrosine kinase inhibitor, genistein (100 microm) had no effect on RVD after 20% volume increases. We conclude that activation of charybdotoxin-insensitive K(+) channels utilized for RVD after substantial (>7%) 'nonphysiological' volume increases requires phosphorylation mediated by Ca(2+)/calmodulin kinase II and that increases in cytosolic acidification rather than larger increases in [Ca(2+)](i) are a critical determinant of this activation.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Jejunum/metabolism , Potassium Channels/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Cell Size , Charybdotoxin/pharmacology , Enzyme Inhibitors/pharmacology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Guinea Pigs , Hydrogen-Ion Concentration , Hypotonic Solutions , In Vitro Techniques , Intracellular Fluid/metabolism , Jejunum/cytology , Jejunum/drug effects , Male , Phosphoprotein Phosphatases/antagonists & inhibitors , Potassium Channel Blockers , Protein-Tyrosine Kinases/antagonists & inhibitors
11.
J Pharmacol Exp Ther ; 291(2): 511-6, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10525065

ABSTRACT

5-Oxo-6,8,11,14-eicosatetraenoic acid (5-oxo-ETE) is a recently discovered arachidonate metabolite that is a potent activator of eosinophils and neutrophils and may be an important mediator of inflammation. The objective of the present investigation was to determine whether 5-oxo-ETE affects the isotonic volume of Cl(-) secretory intestinal crypt epithelial cells. 5-Oxo-ETE caused rapid shrinkage of guinea pig jejunal crypt epithelial cells to a reduced but stable volume, which was measured electronically. This effect was prevented by Cl(-) and K(+) channel blockers and inhibitors of protein kinase C. 5-Oxo-ETE (EC(50) = 20 pM) was more potent than any of the other agonists tested, including its precursor, 5-hydroxy-6,8,11,14-eicosatetraenoic acid (EC(50) = 5 nM); leukotriene D(4) (EC(50) = 1 nM); vasoactive intestinal peptide (EC(50) = 200 pM); and bradykinin (EC(50) = 50 nM). Leukotriene B(4) had no effect on crypt cell volume. In contrast to its effects on crypt cells, 5-oxo-ETE had no effect on the volume of jejunal villus cells. These results indicate that 5-oxo-ETE induces an isotonic volume reduction in intestinal crypt epithelial cells that appears to be dependent on Cl(-) secretion and activation of protein kinase C.


Subject(s)
8,11,14-Eicosatrienoic Acid/pharmacology , Chloride Channels/antagonists & inhibitors , Intestinal Mucosa/drug effects , Jejunum/drug effects , Protein Kinase C/antagonists & inhibitors , Animals , Bradykinin/pharmacology , Dose-Response Relationship, Drug , Guinea Pigs , Leukotrienes/pharmacology , Male , Potassium Channel Blockers , Time Factors , Vasoactive Intestinal Peptide/pharmacology
12.
Gastroenterology ; 116(6): 1342-7, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10348817

ABSTRACT

BACKGROUND & AIMS: When rice is incorporated into oral rehydration therapy for patients with secretory diarrhea, clinical outcomes improve. We have shown that a factor purified from boiled rice (RF) blocks the secretory response of intestinal crypt cells to adenosine 3',5'-cyclic monophosphate (cAMP). Now we report that the cystic fibrosis transmembrane conductance regulator (CFTR) chloride channel is the cellular target for this rice inhibitor. METHODS: We used RF, the same previously described extract prepared from boiled rice, to assess chloride channel activation in vitro, measuring (1) cell volume regulation of guinea pig intestinal crypt epithelial cell suspensions using standard Coulter counter technology, (2) transepithelial chloride current in monolayers of T84 cells mounted in Ussing chambers, and (3) whole-cell and single-channel currents using the patch-clamp technique in cells transfected to express CFTR. RESULTS: RF inhibited activation by cAMP of CFTR chloride channels in all experimental preparations; RF did not block volume-stimulated Cl- secretion, suggesting that its effect might be specific for CFTR chloride channels. RF inhibited transepithelial cAMP-stimulated Cl- current in T84 cells and inhibited forskolin (i.e., cAMP)-induced current in cells transfected with CFTR. Excised patch and single-channel patch-clamp recordings supported the view that the response was a direct effect on CFTR rather than on cAMP signal transduction. CONCLUSIONS: RF exerts a specific inhibitory effect on CFTR chloride channels, blocking activation from the luminal surface of the cell and reversing established activation. Many major diarrheal states are based on cAMP-induced CFTR activation, leading to excessive gut secretion; our findings could have clinical relevance.


Subject(s)
Chlorides/metabolism , Cooking , Intestinal Mucosa/metabolism , Oryza , Animals , CHO Cells , Cell Line/drug effects , Cell Line/metabolism , Chlorides/physiology , Cricetinae , Cyclic AMP/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/antagonists & inhibitors , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Electric Conductivity , Guinea Pigs , Humans , Intestines/cytology , Oryza/chemistry , Patch-Clamp Techniques , Plant Extracts/pharmacology , Transfection
13.
Can J Psychiatry ; 44(2): 144-50, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10097834

ABSTRACT

OBJECTIVE: To compare the identification of psychiatric disorder as informed by parents versus teachers in children aged 6-11 years and parents versus adolescents in youth aged 12-16 years in clinic versus community samples. METHOD: Study data come from parallel surveys in Hamilton, Ontario, of children aged 6-16 years. The surveys included consecutive referrals (N = 1150) between 1989 and 1991 to the region's 2 agencies providing outpatient child mental health services. Also, a simple random sample (N = 1689) was used, drawn in 1989 from students attending public schools. Conduct disorder, hyperactivity, emotional disorder, and somatization disorder were assessed by informants using the original Ontario Child Health Study scales. RESULTS: The percentage of children identified with a disorder was markedly higher in the clinic sample, irrespective of the type of disorder, the age and sex of the child, and who provided the assessment. Also, there was a statistically significant differential shift between parents and teachers in the percentage of children identified with disorder. The ratio of children aged 6-11 years identified with conduct disorder or hyperactivity by parents versus teachers was higher in the clinic sample than in the community sample. Among youth aged 12-16 years, a similar pattern emerged for parents as informants versus the adolescents themselves, but it was statistically nonsignificant. CONCLUSIONS: The data suggest that the relative contribution of informants to the identification of childhood psychiatric disorder varies by sample type: clinic and community. If risk factors for child disorder are influenced by contextually specific factors wedded to informants, then studies conducted in clinic versus community samples may lead to discrepant information about the determinants of psychopathology. The extent of this problem needs to be assessed by comparing the results of parallel studies conducted in clinic versus community samples.


Subject(s)
Caregivers/psychology , Mental Disorders/diagnosis , Parents/psychology , Teaching , Adolescent , Attention Deficit and Disruptive Behavior Disorders/diagnosis , Attention Deficit and Disruptive Behavior Disorders/psychology , Behavioral Symptoms/classification , Behavioral Symptoms/diagnosis , Behavioral Symptoms/psychology , Chi-Square Distribution , Child , Cross-Sectional Studies , Female , Health Surveys , Humans , Male , Mental Disorders/psychology , Neurotic Disorders/diagnosis , Neurotic Disorders/psychology , Sampling Studies , Self-Assessment , Somatoform Disorders/diagnosis , Somatoform Disorders/psychology
14.
J Biol Chem ; 273(32): 20046-51, 1998 Aug 07.
Article in English | MEDLINE | ID: mdl-9685343

ABSTRACT

We have used a sensitive bioassay of calcium-mediated volume changes in mammalian absorptive intestinal epithelial cells to screen extracts of the skin of the amphibian Xenopus laevis for the presence of factors affecting ion transport. A 66-residue peptide, purified using reversed-phase high performance liquid chromatography techniques, caused isotonic volume reduction of guinea pig jejunal villus cells in suspension. This volume reduction required extracellular Ca2+ and was prevented by the dihydropyridine-sensitive Ca2+ channel blocker niguldipine. Structural analysis demonstrated the presence of eight cysteines and a primary structure homologous to that of the neurotoxin/cytotoxin family found in the venom of certain poisonous snakes. The structure of the peptide was identical to that of xenoxin-1 purified from dorsal gland secretions of X. laevis (Kolbe, M., Huber A., Cordier, P., Rasmussen, U., Bouchon, B., Jaquinod, M., Blasak, R., Detot, E., and Kreil, G. (1993) J. Biol. Chem. 268, 16458-16464). Xenoxin-1 (10 nM) caused volume changes that required extracellular Ca2+ and were comparable in magnitude and direction to changes caused by BayK-8644 (100 nM), a dihydropyridine-sensitive Ca2+ channel agonist. The initial rate of dihydropyridine-sensitive 45Ca2+ influx was substantially increased by xenoxin-1. Staurosporine (10 nM) prevented volume changes caused by ATP (250 microM) but had no effect on volume changes caused by BayK-8644 or xenoxin-1. We conclude that xenoxin-1 directly activated dihydropyridine-sensitive Ca2+ channels in villus cells and that a mammalian homologue to xenoxin-1 may exist.


Subject(s)
Amphibian Venoms/chemistry , Calcium Channel Agonists/chemistry , Neurotoxins/chemistry , Peptides/chemistry , Skin/chemistry , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Adenosine Triphosphate/pharmacology , Amino Acid Sequence , Animals , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Cells, Cultured , Dihydropyridines/pharmacology , Guinea Pigs , Jejunum/drug effects , Molecular Sequence Data , Neurotoxins/pharmacology , Staurosporine/pharmacology , Xenopus laevis
16.
J Biol Chem ; 271(38): 23138-45, 1996 Sep 20.
Article in English | MEDLINE | ID: mdl-8798507

ABSTRACT

Epithelial cell volume increases that occur because of the uptake of Na+-cotransported solutes or hypotonic dilution are followed by a regulatory volume decrease (RVD) due to the activation of K+ and Cl- channels. We studied the relationship of Na+/H+ exchange (NHE) to this RVD in suspended guinea pig jejunal villus cells, using electronic sizing to measure cell volume changes and fluorescent spectroscopy of cells loaded with 2', 7'-bis(carboxyethyl)-5()-carboxyfluorescein to monitor intracellular pH (pHi). When the volume increase achieved by these cells during Na+ solute absorption was duplicated by a modest 5-7% hypotonic dilution, their pHi first acidified and then alkalinized. This alkalinization was blocked by 5-(N-methyl-N-isobutyl) amiloride (MIA; 1 microM), an inhibitor of NHE. The RVD subsequent to 5-7% hypotonic dilution was prevented by Na+-free medium and by amiloride and non-amiloride derivatives. The order of potency of these inhibitors was as follows: MIA > 5-(N,N-dimethyl) amiloride > cimetidine > clonidine, in keeping with the pattern attributable to NHE-1 as the isoform of NHE responsible for increase in pHi after modest volume increases. A substantial 30% hypotonic dilution caused acidification, and RVD following this larger volume increase was not affected by MIA. To assess the effect of hypotonicity on the activity of NHE, we measured the rate of MIA-sensitive pHi recovery from an acid load (dpHi/dt) in 5 and 30% hypotonic media. pHi recovery was faster in 5% hypotonic medium compared with isotonic medium and slowest in 30% hypotonic medium, which suggested that the activity of NHE was stimulated in the slightly hypotonic medium, but inhibited in the 30% hypotonic medium. To determine the role of activated NHE in RVD after a modest volume increase, cells were hypotonically diluted 7% in MIA to prevent RVD and then alkalinized by NH4Cl or acidified by propionic acid addition. Only after alkalinization was there complete volume regulation. We conclude that in Na+-absorbing enterocytes, the NHE-1 isoform of Na+/H+ exchange is stimulated by volume increases that duplicate the "physiological" volume increase occurring when these cells absorb Na+-cotransported solutes. The subsequent alkalinization of pHi is a required determinant of the osmolyte loss that underlies this distinct volume regulatory mechanism.


Subject(s)
Cell Size/physiology , Jejunum/metabolism , Protons , Sodium/metabolism , Alkalies/metabolism , Amiloride/analogs & derivatives , Ammonium Chloride/pharmacology , Animals , Biological Transport/drug effects , Cell Size/drug effects , Cimetidine/pharmacology , Clonidine/pharmacology , Dose-Response Relationship, Drug , Epithelial Cells , Epithelium/metabolism , Guinea Pigs , Hydrogen-Ion Concentration , Jejunum/cytology , Jejunum/drug effects , Male
17.
J Biol Chem ; 271(18): 10654-9, 1996 May 03.
Article in English | MEDLINE | ID: mdl-8631871

ABSTRACT

We report the isolation and characterization of RK-1, a novel peptide found in the kidney. RK-1 is related to the corticostatin/defensins and has the sequence MPC-SCKKYCDPWEVIDGSCGLFNSKYCCREK but differs from the very cationic corticostatins/defensins in having only one arginine and a calculated charge at pH 7 of +1. Like some myeloid corticostatin/defensins RK-1 inhibits the growth of Escherichia coli. Since corticostatin/defensins effect ion flux in responsive epithelia we used volume changes in villus enterocytes as a model system to study the effects of RK-1 on ion channels in epithelial cells. At concentrations > or = 10(-9) M RK-1 decreased enterocyte volume in a dose-dependent manner through a pathway that requires extracellular calcium and is inhibited by niguldipine, a dihydropyridine-sensitive "L"-type Ca(2+)-channel blocker. In other assay systems for corticostatin-defensins, such as the inhibition of adrenocorticotropin-stimulated steroidogenesis, or cell lysis, RK-1 was inactive or only weakly active. These results demonstrate the existence of a novel system of biologically active peptides in the kidney represented by RK-1 which is antimicrobial and can activate epithelial ion channels in vitro.


Subject(s)
Kidney/chemistry , Proteins/chemistry , Proteins/isolation & purification , Adrenal Cortex Hormones/biosynthesis , Adrenocorticotropic Hormone/pharmacology , Amino Acid Sequence , Animals , Blood Proteins/chemistry , Blood Proteins/isolation & purification , Blood Proteins/pharmacology , CHO Cells , Cell Survival/drug effects , Chromatography, High Pressure Liquid , Cricetinae , Defensins , Escherichia coli/drug effects , Escherichia coli/growth & development , Molecular Sequence Data , Proteins/pharmacology , Sequence Homology, Amino Acid , Sodium/chemistry , Tumor Cells, Cultured
18.
Am J Physiol ; 270(3 Pt 1): C786-93, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8638658

ABSTRACT

Prior studies show that ileal HCO3- secretion is of crypt origin, possibly involving Na+-HCO3- cotransport. To test for the latter, we isolated crypt cells from guinea pig ileum and determined effects of medium HCO3-, Na+, K+, disulfonic stilbenes, and gramicidin on intracellular pH [pHi;2',7'-bis(carboxyethyl)-5(6)-carboxyfluorescein fluorescence], cell volume (electronic sizing), and Na+ efflux from 22Na+ -preloaded cells. Ileal crypt cells alkalinized when placed in sodium gluconate-HCO3- medium containing N-5-methyl-5-isobutyl amiloride (1 microM), bumetanide (10 microM) and 4,4'-diisothiocyanostilbene-2,2'-disulfonic acid (250 microM which blocks Cl-/HCO3- exchange but not Na+ dependent HCO3- uptake). Depolarization with either gramicidin (50 microM) or 50 mM K+ caused a further 4-acetamido-4'-isothiocyanostilbene-2,2'-disulfonic acid (SITS)-inhibitable increase in pHi. Gramicidin also caused SITS-inhibitable cell swelling. Both gramicidin effects were Na+ dependent: at 0 mM Na+, gramicidin acidified and did not alter cell volume; at 25 mM, gramicidin also acidified; at 90 and 140 mM, gramicidin alkalinized and induced cell swelling. HCO3- -dependent SITS-inhibitable Na+ efflux from 22Na+ -preloaded cells was also seen. We conclude that ileal crypt cells engage in electrogenic Na+ -HCO3- symport.


Subject(s)
Carrier Proteins/metabolism , Intestinal Mucosa/metabolism , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , 4-Acetamido-4'-isothiocyanatostilbene-2,2'-disulfonic Acid/pharmacology , Animals , Bicarbonates/pharmacology , Biological Transport/drug effects , Bumetanide/pharmacology , Gramicidin/pharmacology , Guinea Pigs , Hydrogen-Ion Concentration , Ileum , In Vitro Techniques , Intestinal Mucosa/cytology , Kinetics , Male , Potassium/pharmacology , Sodium/metabolism , Sodium-Bicarbonate Symporters , Time Factors
19.
J Pharmacol Exp Ther ; 276(2): 728-36, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8632343

ABSTRACT

Leukotriene B4 (LTB4) is metabolized by beta-oxidation, omega-oxidation and the 12-hydroxyeicosanoid dehydrogenase/delta 10-reductase pathway. We have investigated the effects of metabolites formed by the latter pathway on calcium mobilization and migration in human neutrophils and have compared their potencies with those of other LTB4 derivatives. 12-Oxo-LTB4 and 10,11-dihydro-LTB4 were 60 to 100 times less potent than LTB4 in stimulating neutrophils, whereas 10,11-dihydro-12-oxo-LTB4 and 10,11-dihydro-12-epi-LTB4 exhibited still lower potencies. The 6-trans isomers of 12-oxo-LTB4 and 10,11-dihydro-12-oxo-LTB4 were much less potent than the 6-cis compounds. The EC50 values for biologically and chemically (6-cis) synthesized 12-oxo-LTB4 were similar, indicating that the 6,7-double bond is retained in the cis configuration in the biologically formed compound. Methylation of LTB4 markedly reduced its effect on cytosolic calcium levels, whereas addition of a 3-hydroxyl group had a much more modest effect. Modifications of the omega end of the molecule also resulted in lower potencies for calcium mobilization. Nearly all of the compounds tested desensitized neutrophils to LTB4-induced calcium mobilization, which suggests that their effects were mediated by receptors for the latter compound. However, modifications in the carboxyl end of the molecule had smaller effects on desensitization than on calcium mobilization, whereas the reverse was true for modifications in the omega end of the molecule. This suggests that the structural requirements for agonist-induced desensitization to LTB4 may differ to some extent from the requirements for calcium mobilization.


Subject(s)
Calcium/metabolism , Leukotriene B4/metabolism , Leukotriene B4/pharmacology , Neutrophils/drug effects , Cell Movement/drug effects , Humans , Leukotriene B4/analogs & derivatives , Neutrophils/physiology , Structure-Activity Relationship
20.
J Immunol ; 156(1): 336-42, 1996 Jan 01.
Article in English | MEDLINE | ID: mdl-8598482

ABSTRACT

5-Oxo-6,8,11,14-eicosatetraenoic acid (5-oxo-ETE) is a recently discovered metabolite of arachidonic acid that activates human neutrophils by a mechanism independent of the receptor for leukotriene B4 (LTB4). The objectives of this study were to identify the major metabolites of 5-oxo-ETE in neutrophils and to compare the biologic activities of 5-oxo-ETE with those of its metabolites and other 5-oxoeicosanoids. Neutrophils rapidly converted 5-oxo-ETE to its omega-oxidation product, 5-oxo-20-hydroxy-6E,8Z,11Z,14Z- eicosatetraenoic acid. This compound was nearly 100 times less potent than 5-oxo-ETE in elevating cytosolic calcium levels in neutrophils. Methylation of the carboxyl group of 5-oxo-ETE resulted in a 20-fold loss of potency, whereas replacement of the 8,9-cis double bond by a trans double bond reduced potency by about sixfold. Similar results were obtained for the effects of the above compounds on neutrophil migration. 5-Oxo-20-hydroxy-6E,8Z,11Z,14Z- eicosatetraenoic acid, 5-oxo-8-trans-ETE, and 5-oxo-ETE methyl ester desensitized neutrophils to 5-oxo-ETE. 5-Oxo-ETE-induced calcium mobilization was inhibited by pretreatment of the cells with pertussis toxin. 5-Oxo metabolites of 6-trans-LTB4 and 12-epi-6-trans-LTB4 had weak stimulatory effects on calcium levels and migration that appeared to be mediated primarily by stimulation of LTB4 receptors. These studies indicate that the 5-oxo group, the omega-end of the molecule, and the carboxyl group are all important for the biologic activity of 5-oxo-ETE, which may be mediated by a G protein-linked receptor. The biologic activity of 5-oxo-ETE can be terminated by omega-oxidation.


Subject(s)
Arachidonic Acids/metabolism , Arachidonic Acids/pharmacology , Neutrophils/drug effects , Neutrophils/metabolism , Arachidonic Acids/blood , Calcium/blood , Calcium/metabolism , Cell Movement/drug effects , Cytosol/drug effects , Cytosol/metabolism , Humans , Leukotriene B4/metabolism , Leukotriene B4/pharmacology , Pertussis Toxin , Stereoisomerism , Tetradecanoylphorbol Acetate/pharmacology , Virulence Factors, Bordetella/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...