Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Peptides ; 31(2): 322-31, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19944726

ABSTRACT

Little is known about CRF expression and regulation in the rat colon compared to the brain. We investigated CRF gene expression, cellular location, and regulation by endotoxin and corticosterone in the male rat colon at 6h after intraperitoneal (ip) injection. CRF mRNA level, detected by reverse transcription-polymerase chain reaction (RT-PCR) was 1.3-fold higher in the distal than proximal colon and 3.4-fold higher in the proximal colonic submucosa plus muscle layers than in mucosa. CRF immunoreactivity was located in the epithelia, lamina propria and crypts, and co-localized with tryptophan hydroxylase, a marker for enterochromaffin (EC) cells, and in enteric neurons. Lipopolysaccharide (LPS, 100 microg/kg, ip) increased defecation by 2.9-fold and upregulated CRF mRNA by 2.5-fold in the proximal and 1.1-fold in the distal colon while there was no change induced by corticosterone as monitored by quantitative PCR. LPS-induced increased CRF mRNA expression occurred in the submucosa plus muscle layers (1.5-fold) and the mucosa of proximal colon (0.9-fold). LPS increased significantly CRF immunoreactivity in the submucosal and myenteric plexuses of proximal and distal colon compared to saline groups. These results indicate that in rats, CRF is expressed in both proximal and distal colon and more prominently in enteric neurons of the submucosa plus muscle layers and subject to upregulation at the gene and protein levels by LPS through corticosteroid independent pathways. These data suggests that colonic CRF may be part of the local effector limb of the CRF(1) receptor mediated colonic alterations induced by acute stress.


Subject(s)
Colon/metabolism , Corticotropin-Releasing Hormone/metabolism , Endotoxins/pharmacology , Up-Regulation/drug effects , Animals , Colon/drug effects , Corticotropin-Releasing Hormone/genetics , Defecation/drug effects , Enterochromaffin Cells/cytology , Enterochromaffin Cells/metabolism , Gene Expression/drug effects , Gene Expression/genetics , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Lipopolysaccharides/pharmacology , Male , Mucous Membrane/cytology , Mucous Membrane/metabolism , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Myenteric Plexus/cytology , Myenteric Plexus/drug effects , Myenteric Plexus/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Up-Regulation/genetics
2.
Neurogastroenterol Motil ; 19(11): 923-36, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17973638

ABSTRACT

Intraperitoneal (i.p.) corticotropin releasing factor (CRF) induced a CRF(1) receptor-dependent stimulation of myenteric neurons and motility in the rat proximal colon. We characterize the colonic enteric nervous system response to CRF in conscious rats. Laser capture microdissection combined with reverse transcriptase polymerase chain reaction (RT-PCR) and immunohistochemistry in longitudinal muscle myenteric plexus whole-mount colonic preparations revealed CRF(1) receptor expression in myenteric neurons. CRF (i.p., 10 microg kg(-1)) induced Fos immunoreactivity (IR) (cells per ganglion) selectively in myenteric plexus of proximal (18.3 +/- 2.4 vs vehicle: 0.0 +/- 0.0) and distal colon (16.8 +/- 1.2 vs vehicle: 0.0 +/- 0.0), but not in that of gastric corpus, antrum, duodenum, jejunum and ileum. The selective CRF(1) agonist, stressin(1)-A (i.p., 10 microg kg(-1)) also induced Fos IR in myenteric but not in submucosal plexus of the proximal and distal colon. Fos IR induced by CRF was located in 55 +/- 1.9% and 53 +/- 5.1% of CRF(1) receptor-IR myenteric neurons and in 44 +/- 2.8% and 40 +/- 3.9% of cholinergic neurons with Dogiel type I morphology, and in 20 +/- 1.6% and 80 +/- 3.3% of nitrergic neurons in proximal and distal colon respectively. CRF and stressin(1)-A elicit defecation and diarrhoea. These data support that one mechanism through which peripherally injected CRF ligands stimulate colonic function involves a direct action on colonic cholinergic and nitrergic myenteric neurons expressing CRF(1) receptor.


Subject(s)
Cholinergic Fibers/metabolism , Colon/innervation , Corticotropin-Releasing Hormone/analogs & derivatives , Corticotropin-Releasing Hormone/pharmacology , Myenteric Plexus/metabolism , Nitrergic Neurons/metabolism , Peptides, Cyclic/pharmacology , Receptors, Corticotropin-Releasing Hormone/metabolism , Animals , Cholinergic Fibers/pathology , Colon/pathology , Gastrointestinal Motility/drug effects , Male , Myenteric Plexus/pathology , Nitrergic Neurons/pathology , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Corticotropin-Releasing Hormone/agonists
3.
Neuroscience ; 128(1): 131-41, 2004.
Article in English | MEDLINE | ID: mdl-15450360

ABSTRACT

I.v. injection of secretin activates neurons in brain areas controlling autonomic function and emotion. Peripheral administration of secretin inhibits gastric functions through a central mechanism that is mediated by vagal dependent pathways. We investigated whether the vagus nerve is involved in i.p. injection of secretin-induced brain neuronal activation in conscious rats as monitored by Fos immunohistochemistry. Secretin (40 or 100 microg/kg, i.p., 90 min) induced a dose-related increase in the number of Fos positive neurons in the central nucleus of the amygdala (CeA), and a plateau Fos response in the area postrema (AP), nucleus tractus solitarii (NTS), locus coeruleus (LC), Barrington's nucleus (Bar), external lateral subnucleus of parabrachial nucleus (PBel) and arcuate nucleus, and at 100 microg/kg, in the dorsal motor nucleus of the vagus (DMV) compared with i.p. injection of vehicle. Double immunohistochemistry showed that secretin (40 microg/kg, i.p.) activates tyrosine hydroxylase neurons in the NTS. Subdiaphragmatic vagotomy (7 days) abolished Fos expression-induced by i.p. secretin (40 microg/kg) in the NTS, DMV, LC, Bar, PBel and CeA, while a significant rise in the AP was maintained. In contrast, s.c. capsaicin (10 days) did not influence the Fos induction in the above nuclei. Reverse transcription polymerase chain reaction (RT-PCR) and quantitative real-time PCR showed that secretin receptor mRNA is expressed in the nodose ganglia and levels were higher in the right compared with the left ganglion. These results indicate that peripheral secretin activates catecholaminergic NTS neurons as well as neurons in medullary, pontine and limbic nuclei regulating autonomic functions and emotion through vagal-dependent capsaicin-resistant pathways. Secretin injected i.p. may signal to the brain by interacting with secretin receptors on vagal afferent as well as on AP neurons outside the blood-brain barrier.


Subject(s)
Brain/drug effects , Oncogene Proteins v-fos/drug effects , Secretin/pharmacology , Vagus Nerve/metabolism , Animals , Brain/metabolism , Capsaicin/metabolism , Capsaicin/pharmacology , Functional Laterality , Immunohistochemistry , Male , Neural Pathways/drug effects , Neural Pathways/metabolism , Neurons/drug effects , Neurons/metabolism , Nodose Ganglion/metabolism , Oncogene Proteins v-fos/biosynthesis , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Secretin/metabolism , Vagotomy , Vagus Nerve/surgery
4.
Pancreas ; 25(4): 405-10, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12409837

ABSTRACT

INTRODUCTION AND AIMS: Orexins have been demonstrated to have mainly central physiological functions, including regulation of food and water intake, sleep, and arousal. However, little is known about their direct peripheral effects, if any. As a first step toward understanding the role of Orexin in non-neuronal tissues or cells, we initiated studies to examine expression of Orexin receptors (OXR) in an established pancreatic tumor cell line AR42J. Secondly, we wanted to determine whether Orexins, in various molecular forms, are active to stimulate any known pancreatic cell functions in AR42J cells. METHODOLOGY: Reverse transcription-PCR analysis was performed to identify the presence of specific Orexin receptor subtypes. Intracellular calcium mobilization and cAMP levels were measured following stimulation by Orexin A and B peptides, their respective C-terminal decapeptide fragments, and hypocretin-2-gly (glycine-extended Orexin B). Release of alpha-amylase was measured in conditioned media after acute stimulation with the set of Orexin peptides for 30 minutes. Cell proliferation was determined by H-thymidine incorporation after 24 hours following treatment with Orexins under serum-free condition. RESULTS: RT-PCR and sequencing results showed that Orexin receptor subtype 2 (OX2R) was the main form expressed in AR42J cells. Orexins stimulated dose-dependent increases in intracellular calcium mobilization with EC50 0.05 nM for Orexin A and 0.1 nM for Orexin B but were unable to stimulate any significant cAMP accumulation or DNA synthesis even at micromolar concentrations. Both Orexin-A and -B, but not hypocretin-2-gly, also stimulated dose-dependent increases in amylase release in the AR42J cells. Orexin-A and -B carboxyl-terminal decapeptides elicited significant but much lower calcium and amylase responses. CONCLUSION: Our data demonstrate that OX2R mediates Ca -dependent amylase release in AR42J cells, suggesting that Orexins may have secretory functions in pancreatic tumor cells.


Subject(s)
Amylases/metabolism , Carrier Proteins/pharmacology , Intracellular Signaling Peptides and Proteins , Neuropeptides/pharmacology , Pancreas/enzymology , Receptors, Neuropeptide/physiology , Amino Acid Sequence , Animals , Calcium/metabolism , Carrier Proteins/genetics , Cyclic AMP/analysis , Dose-Response Relationship, Drug , Molecular Sequence Data , Neuropeptides/genetics , Orexin Receptors , Orexins , Pancreas/chemistry , Pancreas/drug effects , RNA, Messenger/biosynthesis , Rats , Receptors, G-Protein-Coupled , Receptors, Neuropeptide/genetics , Sequence Alignment , Tumor Cells, Cultured
5.
Am J Physiol Gastrointest Liver Physiol ; 280(1): G88-94, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11123201

ABSTRACT

Only one secretin receptor has been cloned and its properties characterized in native and transfected cells. To test the hypothesis that stimulatory and inhibitory effects of secretin are mediated by different secretin receptor subtypes, pancreatic and gastric secretory responses to secretin and secretin-Gly were determined in rats. Pancreatic fluid secretion was increased equipotently by secretin and secretin-Gly, but secretin was markedly more potent for inhibition of basal and gastrin-induced acid secretion. In Chinese hamster ovary cells stably transfected with the rat secretin receptor, secretin and secretin-Gly equipotently displaced (125)I-labeled secretin (IC(50) values 5.3 +/- 0.5 and 6.4 +/- 0.6 nM, respectively). Secretin, but not secretin-Gly, caused release of somatostatin from rat gastric mucosal D cells. Thus the equipotent actions of secretin and secretin-Gly on pancreatic secretion appear to result from equal binding and activation of the pancreatic secretin receptor. Conversely, secretin more potently inhibited gastric acid secretion in vivo, and only secretin released somatostatin from D cells in vitro. These results support the existence of a secretin receptor subtype mediating inhibition of gastric acid secretion that is distinct from the previously characterized pancreatic secretin receptor.


Subject(s)
Peptide Fragments/pharmacology , Receptors, Gastrointestinal Hormone/classification , Receptors, Gastrointestinal Hormone/metabolism , Secretin/pharmacology , Animals , CHO Cells , Cricetinae , Dogs , Gastric Acid/metabolism , Gastric Mucosa/chemistry , Gastric Mucosa/metabolism , Gastrins/pharmacology , Glycine , Iodine Radioisotopes , Male , Pancreas/drug effects , Pancreas/metabolism , Peptide Fragments/chemical synthesis , Protein Processing, Post-Translational/physiology , Radioligand Assay , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled , Secretin/analogs & derivatives , Secretin/chemical synthesis
6.
Pancreas ; 19(3): 224-30, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10505752

ABSTRACT

Expression of the long form of the leptin receptor, the isoform that is considered to have full signaling capability, has been reported in the central nervous system and several peripheral cell types. However, only a few cell lines have been shown to express the long form of the receptor. AR42J, a cell line derived from azaserine-treated rat pancreas, is a common model for pancreatic acinar cell secretion. In this study, the presence of leptin-receptor variants and leptin action was evaluated in this cell line. Messenger RNAs for both the long and a short form of the leptin receptor were detected by reverse transcription-polymerase chain reaction (RT-PCR) in AR42J cells, and authenticity of the receptor was confirmed by DNA sequencing. Competitive binding studies demonstrated that binding of radiolabeled leptin was specific and did not cross-react with cholecystokinin (CCK). Biologic effects of leptin on amylase release and intracellular calcium mobilization were further assessed in the presence and the absence of CCK, a known pancreatic secretagogue. Although leptin alone (< or =200 ng/ml) did not affect basal amylase release, it inhibited amylase release stimulated by 1 nM CCK by 48%. Leptin alone had no significant effect on calcium mobilization. However, pretreatment of leptin (10 and 100 ng/ml) enhanced calcium responses stimulated by CCK. These data demonstrate that the rat pancreatic tumor cell line AR42J expresses a functional form of the leptin receptor that modulates the action of CCK in calcium mobilization and amylase release.


Subject(s)
Amylases/metabolism , Cholecystokinin/pharmacology , Leptin/pharmacology , Pancreas/drug effects , Pancreas/metabolism , Receptors, Cell Surface , Amylases/antagonists & inhibitors , Animals , Binding, Competitive/drug effects , Calcium/metabolism , Carrier Proteins/genetics , Cell Line , Cholecystokinin/pharmacokinetics , Dose-Response Relationship, Drug , Gastrointestinal Agents/pharmacology , Intracellular Fluid/metabolism , Leptin/pharmacokinetics , Protein Isoforms/genetics , RNA, Messenger/metabolism , Rats , Receptors, Leptin , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Sincalide/pharmacology
7.
J Gastroenterol Hepatol ; 14(5): 476-80, 1999 May.
Article in English | MEDLINE | ID: mdl-10355513

ABSTRACT

BACKGROUND: Cytotoxin-associated gene A (cagA) has been implicated as a potential pathogenic marker for Helicobacter pylori-induced severe gastroduodenal diseases. Although the prevalence of cagA-positive strains has been reported in patient populations from developed countries, only limited information from developing countries is available. METHODS: Polymerase chain reaction (PCR) in combination with immunoblot analysis was used to determine the prevalence of cagA and its adjacent cagE genes and to evaluate the expression of CagA protein in 55 H. pylori clinical isolates from China. RESULTS: The expected PCR products derived from H. pylori cagA and cagE genes were identified in all Chinese H. pylori clinical isolates. Similarly, the CagA protein was detected in all 40 isolates tested. CONCLUSIONS: These results demonstrated that the presence of the cagA gene correlated well with expression of the CagA protein in all surveyed Chinese H. pylori isolates and that infection with cagA-positive H. pylori strains is highly common in China and independent of clinical presentation.


Subject(s)
Bacterial Proteins/genetics , Helicobacter Infections/epidemiology , Helicobacter pylori/genetics , Helicobacter pylori/isolation & purification , Adolescent , Adult , Antigens, Bacterial/biosynthesis , Antigens, Bacterial/genetics , Bacterial Proteins/biosynthesis , Blotting, Western , China , Female , Gene Expression , Gene Frequency , Helicobacter Infections/microbiology , Helicobacter pylori/metabolism , Humans , Male , Middle Aged , Polymerase Chain Reaction , Prevalence , Sensitivity and Specificity
8.
Mol Pharmacol ; 55(5): 795-803, 1999 May.
Article in English | MEDLINE | ID: mdl-10220557

ABSTRACT

To understand molecular basis of Gs coupling to cholecystokinin (CCK)-A and CCK-B receptor subtypes, we examined cAMP responses in three sets of human CCK receptor mutants expressed in human embryonic kidney (HEK)293 cells. Single or double substitutions of the four nonconserved amino acids in the first intracellular loop of the CCK-BR were made with their CCK-AR counterparts to determine which residues are critical in Gs coupling. Single substitution of Ser82 to Asn, produced maximal cAMP responses comparable with the chimeric CCK-BR containing the entire first intracellular loop of the CCK-AR. Two other single substitutions, Leu81 to Arg and Leu85 to Met, produced significant but smaller cAMP responses. Ser82 was further changed into Asp, Thr, or Ala to determine the specificity of this position in Gs coupling by the CCK-BR. Replacements of Ser to Asp or Thr showed significant cAMP increases but the stimulatory effects were smaller than Ser to Asn, whereas Ser to Ala did not enhance any cAMP response to either CCK or gastrin. Finally, CCK-AR reverse mutants were studied to compare them with their corresponding CCK-BR mutants that showed increased cAMP responses. Substitution of CCK-AR residue Arg68 to Leu resulted in a complete loss of cAMP response, whereas Asn69 to Ser or Met72 to Leu showed markedly diminished cAMP responses. These data identify that specific residues in the first intracellular loop of both CCK receptor subtypes are critical for Gs coupling. Substitution of a single residue Ser82 to Asn in the CCK-BR is sufficient to confer full cAMP responses to agonist stimulation.


Subject(s)
Cholecystokinin/metabolism , Cyclic AMP/metabolism , Gastrins/metabolism , Mutagenesis , Receptors, Cholecystokinin/metabolism , Adenylyl Cyclases/metabolism , Amino Acid Sequence , Amino Acid Substitution , Asparagine/genetics , Binding Sites , Calcium/metabolism , Cells, Cultured , Humans , Molecular Sequence Data , Receptor, Cholecystokinin B , Receptors, Cholecystokinin/biosynthesis , Receptors, Cholecystokinin/genetics , Sequence Homology, Amino Acid , Serine/genetics
9.
Am J Physiol ; 275(4): G660-7, 1998 10.
Article in English | MEDLINE | ID: mdl-9756494

ABSTRACT

The localization of histidine decarboxylase (HDC) activity in the enterochromaffin-like (ECL) cells of the oxyntic mucosa was studied during fasting and refeeding using monoclonal (CURE no. 44178) and polyclonal (CURE no. 94211) antibodies directed against the COOH terminus of HDC (HDC-CT). Changes in HDC immunostaining were correlated with mucosal HDC enzyme activity. Immunoneutralization of circulating gastrin and atropine treatment during refeeding were used to determine the relative importance of gastrin and cholinergic mechanisms in the regulation of HDC activity and immunostaining. Fasting caused a rapid reduction in the number of ECL cells immunostaining for HDC that was correlated with an almost complete loss of mucosal HDC enzyme activity. Refeeding restored both HDC immunostaining and enzyme activity within 2-4 h, and this response was inhibited by gastrin immunoneutralization but not by atropine treatment. Immunostaining was uniformly decreased and restored in the lower half of the oxyntic mucosa, which corresponds to the predominant area of ECL cells in the gastric gland. Histamine immunostaining and mucosal histamine content were not significantly changed during fasting and refeeding or by gastrin antibody and/or atropine treatment during refeeding. These findings indicate that HDC activity correlates with HDC-CT immunostaining and that both HDC activity and HDC-CT immunostaining are regulated by gastrin during refeeding.


Subject(s)
Eating/physiology , Enterochromaffin-like Cells/enzymology , Gastric Mucosa/enzymology , Gastrins/physiology , Histidine Decarboxylase/analysis , Amino Acid Sequence , Animals , Antibodies, Monoclonal/pharmacology , Enterochromaffin-like Cells/cytology , Gastric Mucosa/cytology , Gastric Mucosa/physiology , Gastrins/immunology , Histamine/analysis , Histidine Decarboxylase/chemistry , Immunohistochemistry , Male , Peptide Fragments/chemistry , Peptide Fragments/immunology , Rats , Rats, Sprague-Dawley
10.
Endocrinology ; 139(9): 3730-5, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9724024

ABSTRACT

Cold exposure increases TRH gene expression in hypothalamic and raphe nuclei and results in a vagal activation of gastric function. We investigated the role of medullary TRH receptors in cold (4-6 C, 90 min)-induced stimulation of gastric motor function in fasted conscious rats using intracisternal injections of TRH receptor (TRHr) antisense oligodeoxynucleotides (100 microg twice, -48 and -24 h). The gastric emptying of a methyl-cellulose solution was assessed by the phenol red method. TRH (0.1 microg) or the somatostatin subtype 5-preferring analog, BIM-23052 (1 microg), injected intracisternally increased basal gastric emptying by 34% and 47%, respectively. TRHr antisense, which had no effect on basal emptying, blocked TRH action but did not influence that of BIM-23052. Cold exposure increased gastric emptying by 64%, and the response was inhibited by vagotomy, atropine (0.1 mg/kg, i.p.), and TRHr antisense (intracisternally). Saline or mismatched oligodeoxynucleotides, injected intracisternally under similar conditions, did not alter the enhanced gastric emptying induced by cold or intracisternal injection of TRH or BIM-23052. These results indicate that TRH receptor activation in the brain stem mediates acute cold-induced vagal cholinergic stimulation of gastric transit, and that medullary TRH may play a role in the autonomic visceral responses to acute cold.


Subject(s)
Cisterna Magna/physiology , Cold Temperature , Gastric Emptying/drug effects , Oligonucleotides, Antisense/pharmacology , Receptors, Thyrotropin-Releasing Hormone/genetics , Vagus Nerve/physiology , Animals , Atropine/pharmacology , Male , Oligonucleotides, Antisense/genetics , Rats , Rats, Sprague-Dawley , Somatostatin/analogs & derivatives , Somatostatin/pharmacology , Thyrotropin-Releasing Hormone/pharmacology , Time Factors , Vagotomy
11.
J Comp Neurol ; 386(3): 396-408, 1997 Sep 29.
Article in English | MEDLINE | ID: mdl-9303425

ABSTRACT

Somatostatin exerts multiple effects by activating distinct G protein-coupled receptors. Here we report the cellular sites of expression of the somatostatin subtype 2A (sst2A) receptor in the rat enteric nervous system by using a C-terminus-specific, affinity-purified antiserum and immunohistochemistry. Antibody specificity was confirmed by the cell surface staining of human embryonic kidney 293 cells expressing the sst2A receptor, the lack of staining of cells expressing the somatostatin subtype 2B receptor, and the abolition of staining by preincubating the antiserum with the C-terminus peptide used for immunization, SSt2A(361-369). The SSt2A receptor antibody recognized a broad 80 kDa band on Western blots of membranes prepared from cells transfected with sst2A receptor cDNA; following receptor membrane deglycosylation, the antibody detected an additional 40 kDa band. In the enteric nervous system, the sst2A antibody primarily stained neurons of the myenteric and submucosal plexuses, and abundant fibers distributed to the muscle, mucosa, and vasculature. Immunoreactive staining was also observed in non-neuronal cells, including presumed interstitial cells of Cajal of the intestine and enterochromaffin-like cells of the stomach. Fibers expressing sst2A receptor immunoreactivity were often in close proximity to D cells of the gastric and intestinal mucosa. Colocalization of somatostatin and sst2A receptor immunoreactivities was not observed in endocrine cells nor in enteric neurons. Double-label immunohistochemistry revealed colocalization of sst2A and vasoactive intestinal peptide immunoreactivities in enteric neurons. The multiple types of cells expressing the sst2A receptor, including enteric neurons and non-neuronal structures, in addition to the relationship between somatostatin and sst2A receptor elements, provide evidence that the sst2A receptor mediates somatostatin effects in the gastrointestinal tract via neuronal and paracrine pathways.


Subject(s)
Chromaffin Cells/cytology , Digestive System/innervation , Gastric Mucosa/innervation , Myenteric Plexus/cytology , Neurons/cytology , Receptors, Somatostatin/analysis , Amino Acid Sequence , Animals , Blotting, Western , Cell Line , Cloning, Molecular , Digestive System/cytology , Female , Gastric Mucosa/cytology , Humans , Immunohistochemistry , Kidney , Muscle, Smooth/cytology , Muscle, Smooth/innervation , Nerve Fibers/ultrastructure , Peptide Fragments/chemistry , Peptide Fragments/immunology , Rabbits , Rats , Rats, Sprague-Dawley , Receptors, Somatostatin/biosynthesis , Recombinant Proteins/analysis , Recombinant Proteins/biosynthesis , Transfection
12.
Dig Dis Sci ; 42(5): 1062-9, 1997 May.
Article in English | MEDLINE | ID: mdl-9149063

ABSTRACT

The intestine of the suckling rat has the unique capacity of absorbing immunoglobulins from maternal milk. We investigated intestinal Fc receptor mRNA expression and the absorption of orally administered antibodies to delineate the ontogeny and tissue specificity of this transport system. Duodenal expression of Fc receptor mRNA was at maximum levels between 1 and 19 days of age, but was not detectable during fetal life and in animals after weaning. Along the horizontal axis of the intestine, FcRn mRNA expression was maximum in the proximal duodenum and declined gradually in distal bowel. Similarly, absorption of orally administered antibody was low shortly after birth, but reached maximum levels at 14 days of age. By the time of weaning, antibody uptake had almost completely ceased. These data further delineate the temporal and spatial nature of the intestinal immunoglobulin transport system, and represent additional examples of how the intestinal Fc receptor is transcriptionally regulated.


Subject(s)
Intestines/immunology , Receptors, Fc/biosynthesis , Animals , Animals, Suckling , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacokinetics , Biological Transport , Blotting, Northern , Female , Fetus/immunology , Immunity, Maternally-Acquired/physiology , Intestinal Absorption , Intestines/embryology , Intestines/physiology , Male , Pregnancy , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Receptors, Fc/genetics
13.
Am J Physiol ; 272(3 Pt 1): C1017-24, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9124504

ABSTRACT

The membrane potential (E(m)) was used as an indicator to evaluate the effect of stem cell factor (SCF) on the membrane integrity of peritoneal mast cells (PMCs). PMCs were harvested from the peritoneal lavage of Sprague-Dawley rats, purified more than 95% and cultured with or without the presence of SCF (2 x 10(-8) M). E(m) values were measured with conventional intracellular recording techniques. Results from day 1 to day 4 in culture were compared. Significant differences in average E(m) (aE(m)) (P < 0.01, analysis of variance) were seen on days 3 and 4 (means +/- SE in millivolts): -67.4 +/- 8.0 and -59.4 +/- 4.8 with SCF vs. -24.8 +/- 7.9 and -7.6 +/- 3.9 without SCF, respectively. Moreover, after culture with SCF for >1 wk, the aE(m) values of purified PMCs had a tendency to reach plateau values similar to that of unpurified PMCs on day 1 (at -20 mV). The morphological appearances of PMCs can be correlated with the results of aE(m) measurements. PMCs with a smooth spherical shape and highly refractive appearance, and better tolerance to electrode impalement, showed E(m) with greater negative values and lesser fluctuations. These results indicate that SCF can maintain the membrane properties and viability of purified PMCs in a long-term culture.


Subject(s)
Mast Cells/physiology , Stem Cell Factor/pharmacology , Animals , Cell Survival/drug effects , Cells, Cultured , Female , Image Processing, Computer-Assisted , Mast Cells/drug effects , Membrane Potentials/drug effects , Peritoneal Cavity/cytology , Rats , Rats, Sprague-Dawley , Time Factors
14.
Gastroenterology ; 112(1): 127-35, 1997 Jan.
Article in English | MEDLINE | ID: mdl-8978351

ABSTRACT

BACKGROUND & AIMS: The cellular target for peptide YY (PYY) inhibition of gastric acid secretion is unknown. The aim of this study was to determine whether PYY inhibits histamine release from isolated enterochromaffin-like (ECL) cells by stimulation of a selective Y receptor. METHODS: Isolated rat gastric ECL cells were analyzed in short-term culture for histamine release and for changes in intracellular calcium concentration using video imaging. RESULTS: Gastrin-stimulated histamine release was inhibited with a 50% inhibiting concentration of 2.10(-9) mol/L. Inhibition of histamine release and of calcium entry by PYY and [Pro34]PYY and no effect of PYY(3-36) characterizes an inhibitory Y1 receptor subtype. Reverse-transcription polymerase chain reaction of ECL cell RNA showed that the receptor was the nontruncated Y1 isoform. The inhibitory action of PYY and related peptides on gastrin-stimulated histamine release and calcium signaling was eliminated by pretreatment with 200 ng/mL pertussis toxin. Additive but not synergistic inhibitory effects of PYY and somatostatin on gastrin-stimulated histamine release were observed. CONCLUSIONS: Activation of a Y1 inhibitory receptor subtype present on the gastric ECL cell that inhibits gastrin-induced ECL cell histamine release and Ca2+ entry by activation of a Gi or G(o) class of protein may account for inhibition of gastric acid secretion by PYY released from the small intestine.


Subject(s)
Gastrins/antagonists & inhibitors , Gastrointestinal Hormones/pharmacology , Histamine Release/drug effects , Peptides/pharmacology , Animals , Calcium/metabolism , Cells, Cultured , Gastrins/pharmacology , Peptide YY , Pertussis Toxin , Rats , Receptors, Gastrointestinal Hormone/drug effects , Somatostatin/pharmacology , Virulence Factors, Bordetella/pharmacology
15.
Am J Physiol ; 271(5 Pt 1): G799-804, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8944693

ABSTRACT

To investigate the physiological role of endogenous gastrin-releasing peptide (GRP) in regulating the release of gastrin, we evaluated the response of intragastric pH, gastrin, and GRP after omeprazole treatment in rats. A significant elevation of the plasma level of GRP (P < 0.01) and a significant reduction of the antral content of GRP (P < 0.05) were observed after the administration of 100 mg/kg omeprazole. The antral content of GRP was significantly decreased 12 h after omeprazole administration and thereafter gradually returned to control levels. Peak values for intragastric pH and plasma GRP were observed 3 h after omeprazole administration and before the peak of serum gastrin. The bombesin antagonist [D-Phe6]-bombesin-(6,13)-methyl ester significantly inhibited gastrin release after omeprazole treatment (P < 0.05). These observations indicate that omeprazole-induced inhibition of acid secretion stimulates the release of GRP and suggest that the secretion of GRP induced by omeprazole may stimulate the secretion of gastrin, at least in the early phase.


Subject(s)
Gastric Mucosa/physiology , Gastrins/metabolism , Omeprazole/pharmacology , Peptides/physiology , Animals , Anti-Ulcer Agents/pharmacology , Gastric Fundus , Gastric Mucosa/drug effects , Gastrin-Releasing Peptide , Gastrins/biosynthesis , Gastrins/blood , Gastrointestinal Hormones/physiology , Humans , Hydrogen-Ion Concentration , Male , Peptides/blood , Peptides/pharmacology , Pyloric Antrum , RNA, Messenger/biosynthesis , Radioimmunoassay , Rats , Rats, Wistar , Time Factors , Transcription, Genetic
16.
Eur J Biochem ; 230(2): 439-46, 1995 Jun 01.
Article in English | MEDLINE | ID: mdl-7607213

ABSTRACT

Avian gastrin is a gastric acid secretagogue, but is structurally more closely related to mammalian cholecystokinin, which is functionally distinct from gastrin. This apparently anomalous structure/activity relationship raises important issues for understanding the evolution of regulatory peptides and the mechanisms that control their expression. To clarify the possible mechanisms, we have determined the sequence of a 6.5-kb BamHI genomic DNA fragment that includes the entire avian gastrin transcriptional unit. The complete cDNA sequence, determined by anchored PCR, encoded a precursor of 105 amino acids. Conserved primary amino acid structures were limited to a decapeptide determining biological activity, and essential sites for post-translational processing. Significantly, however, the genomic regulatory regions, and introns, were unlike those of any previously reported gastrin/cholecystokinin gene. The avian gastrin gene contained no recognizable TATA-box motif, a feature unique to this gene family, but had a G+C-rich region immediately upstream of the transcription initiation site, and a Z-DNA purine-pyrimidine repeat sequence. Moreover, physiological regulation of the avian gastrin gene differed markedly from that observed in mammals, in that the important local paracrine downregulation by antral somatostatin was absent; instead, evidence for a hormonal regulation was demonstrated. The data indicate that genomic regulatory elements within regulatory peptide families such as the gastrin/cholecystokinin family, and mechanisms of physiological control, are not conserved during evolution, even though biologically important amino acid sequence information is retained.


Subject(s)
Gastrins/genetics , Gene Expression Regulation/physiology , TATA Box , Amino Acid Sequence , Animals , Base Sequence , Chickens , DNA, Complementary , Molecular Sequence Data , Sequence Homology, Amino Acid
17.
Gastroenterology ; 107(4): 1067-74, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7523214

ABSTRACT

BACKGROUND/AIMS: Gastric enterochromaffinlike (ECL) cells play an important role in peripheral regulation of acid secretion. This study investigated the somatostatin receptor subtype on ECL cells. METHODS: ECL cells were isolated from rat fundic mucosa to a purity of 90%-95% by combining enzymatic digestion, elutriation, density gradient centrifugation, and culture. RESULTS: Polymerase chain reaction performed with templates from an ECL cell complementary DNA library and primers specific to each of the five known somatostatin receptor subtypes showed that the somatostatin receptor type 2 was significantly enriched in ECL complementary DNA. Single cell videoimaging of highly purified ECL cells in culture showed that only the somatostatin receptor type 2 selective agonist, DC 32-87, inhibited the gastrin-induced calcium signal at 10(-11) mol/L. The type 3 and type 4 selective agonists, DC 25-12 and DC 32-92, and also somatostatin 14 required 100-1000 times higher concentrations (10(-8) mol/L). The somatostatin receptor type 2 analogue also inhibited gastrin-stimulated histamine release with a 50% inhibitory concentration (IC50) value of 2 x 10(-12) mol/L, whereas somatostatin 14 and the type 3 and 4 analogues showed IC50 values of 1 to 5 x 10(-9) mol/L. CONCLUSIONS: The predominant somatostatin receptors on rat gastric ECL cells are of the somatostatin receptor 2 subtype; they inhibit histamine secretion by interfering with the gastrin-induced calcium signal.


Subject(s)
Enterochromaffin Cells/metabolism , Receptors, Somatostatin/metabolism , Amino Acid Sequence , Animals , Base Sequence , Calcium/metabolism , DNA, Complementary , Enterochromaffin Cells/physiology , Gene Library , Histamine Release/drug effects , Molecular Probes/genetics , Molecular Sequence Data , Polymerase Chain Reaction , Rats , Receptors, Somatostatin/classification , Somatostatin/agonists , Somatostatin/analogs & derivatives , Somatostatin/genetics
18.
Neuroscience ; 61(3): 655-63, 1994 Aug.
Article in English | MEDLINE | ID: mdl-7969936

ABSTRACT

The stimulation of thyrotropin release by cold is associated with an increase in thyrotropin-releasing hormone gene expression in the paraventricular nucleus of the hypothalamus. Cold exposure also stimulates autonomic outflow to viscera. There is evidence that caudal raphe nuclei are involved in autonomic regulation through thyrotropin-releasing hormone projections to the dorsal vagal complex and spinal cord. To determine whether cold modulates thyrotropin-releasing hormone gene expression in the caudal raphe nuclei, the effect of cold exposure on thyrotropin-releasing hormone messenger RNA levels in the rat lower brainstem was examined by quantitative Northern blot analysis and thyrotropin-releasing hormone messenger RNA was localized by in situ hybridization. The gastric responses to cold exposure were also assessed in sham or vagotomized rats with pylorus ligation. Thyrotropin-releasing hormone messenger RNA signal was detected in the RNA extracted from the medulla and hypothalamus but not from the amygdala, periaqueductal gray or cerebellum. Cold exposure (4 degrees C) for 1 or 3 h increased thyrotropin-releasing hormone messenger RNA levels in the medulla by 77 +/- 37 and 142 +/- 39% respectively. In situ hybridization histochemistry showed that the increase in silver grain density occurred exclusively in the raphe pallidus and raphe obscurus. Exposure to cold stress for 2 h stimulated gastric acid secretion and resulted in gastric lesion formation in sham but not vagotomized rats. There are established thyrotropin-releasing hormone projections from the raphe pallidus and obscurus to the dorsal vagal complex.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Cold Temperature , Gene Expression/physiology , Raphe Nuclei/metabolism , Raphe Nuclei/physiopathology , Stomach Ulcer/physiopathology , Thyrotropin-Releasing Hormone/biosynthesis , Vagus Nerve/physiopathology , Animals , Base Sequence , Blotting, Northern , Gastric Acid/metabolism , Gastric Mucosa/drug effects , Gastric Mucosa/physiology , In Situ Hybridization , Male , Molecular Sequence Data , Pylorus/physiology , Rats , Rats, Sprague-Dawley , Vagotomy
20.
Am J Physiol ; 266(3 Pt 1): G417-24, 1994 Mar.
Article in English | MEDLINE | ID: mdl-7909409

ABSTRACT

This study demonstrates the effectiveness of parenteral and oral anti-somatostatin monoclonal antibody to stimulate gastrin cell activity in suckling rats. Intraperitoneal anti-somatostatin monoclonal antibody increased serum gastrin concentration (> 2-fold), and orally administered antibodies retained their neutralizing capabilities as demonstrated by a 30% induction of gastrin synthesis. Absorption of luminal monoclonal antibody was time dependent and saturable as demonstrated by measurement of serum murine immunoglobulin G and the subsequent effects on serum gastrin and antral gastrin mRNA concentrations. Enhanced gastrin synthesis after oral administration required whole antibody in that enterally delivered F(ab')2 fragments were not absorbed and did not increase serum gastrin concentrations. In addition, pretreatment with excess Fc fragments decreased monoclonal antibody absorption and eliminated the serum gastrin response to oral monoclonal antibody. These results demonstrate that orally administered murine anti-somatostatin monoclonal antibody was rapidly and efficiently absorbed via an Fc-dependent mechanism and retained immunoneutralizing activity against endogenous somatostatin, neutralizing its inhibition of gastrin cell activity. These results indicate that orally administered monoclonal antibodies could potentially be used to determine the role of other peptides and growth factors as potential physiological regulators during the suckling period of postnatal development.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Gastrins/metabolism , Somatostatin/immunology , Administration, Oral , Animals , Animals, Suckling , Antibodies, Monoclonal/pharmacology , Base Sequence , Biological Transport , Dose-Response Relationship, Drug , Gastrins/genetics , Immunoglobulin Fc Fragments/pharmacology , Immunoglobulin G/metabolism , Infusions, Parenteral , Intestinal Absorption/drug effects , Intestines , Molecular Sequence Data , Oligonucleotide Probes/genetics , Pyloric Antrum/metabolism , RNA, Messenger/metabolism , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...