Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 432
Filter
1.
IEEE Trans Biomed Eng ; 57(10): 2535-48, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20550980

ABSTRACT

Repetitive closure of the upper airway characterizes obstructive sleep apnea. It disrupts sleep causing excessive daytime drowsiness and is linked to hypertension and cardiovascular disease. Previous studies simulating the underlying fluid mechanics are based upon geometries, time-averaged over the respiratory cycle, obtained usually via MRI or CT scans. Here, we generate an anatomically correct geometry from data captured in vivo by an endoscopic optical technique. This allows quantitative real-time imaging of the internal cross section with minimal invasiveness. The steady inhalation flow field is computed using a k-ω shear-stress transport (SST) turbulence model. Simulations reveal flow mechanisms that produce low-pressure regions on the sidewalls of the pharynx and on the soft palate within the pharyngeal section of minimum area. Soft-palate displacement and side-wall deformations further reduce the pressures in these regions, thus creating forces that would tend to narrow the airway. These phenomena suggest a mechanism for airway closure in the lateral direction as clinically observed. Correlations between pressure and airway deformation indicate that quantitative prediction of the low-pressure regions for an individual are possible. The present predictions warrant and can guide clinical investigation to confirm the phenomenology and its quantification, while the overall approach represents an advancement toward patient-specific modeling.


Subject(s)
Image Processing, Computer-Assisted/methods , Models, Biological , Pharynx/anatomy & histology , Sleep Apnea Syndromes/etiology , Endoscopy/instrumentation , Humans , Palate, Soft/anatomy & histology , Pharynx/physiopathology , Sleep Apnea Syndromes/physiopathology , Tomography, Optical Coherence/instrumentation , Tomography, Optical Coherence/methods
2.
Aliment Pharmacol Ther ; 17(9): 1109-14, 2003 May 01.
Article in English | MEDLINE | ID: mdl-12752347

ABSTRACT

BACKGROUND: Peptone meal-stimulated gastric acid output is considered to be a reliable means to evaluate drug-mediated inhibition of stimulated gastric acid output, an important measure of the efficacy of the agents--such as proton pump inhibitors--used to treat acid-related disorders. AIM: To compare the initial and overall inhibitory effects on peptone meal-stimulated gastric acid secretion of rabeprazole and omeprazole, 20 mg, in Helicobacter pylori-negative subjects on the first and eighth days of treatment. METHODS: Healthy volunteers (n = 27) were randomized in a single-centre, double-blind, double-dummy, 2 x 2 cross-over study. Subjects received an oral dose of rabeprazole or omeprazole, 20 mg once daily, for 8 days. After a 2-4-week washout period, subjects were crossed over to receive the other medication for 8 days. Peptone meal-stimulated gastric acid secretion was measured at hours 11 and 23 at baseline and on days 1 and 8 of treatment. RESULTS: On days 1 and 8, rabeprazole demonstrated a significantly greater inhibition of peptone meal-stimulated gastric acid secretion compared with omeprazole at all time points (P < 0.03). Median values of steady-state inhibition on day 1 were statistically significant at hour 23 (rabeprazole 100% vs. omeprazole 74%, P < 0.02). CONCLUSIONS: Rabeprazole, 20 mg, demonstrated superior control of peptone meal-stimulated gastric acid secretion compared with omeprazole, 20 mg, after the first dose and after the eighth daily dose. Rabeprazole achieved a more rapid onset of acid inhibition and a greater steady-state reduction in peptone meal-stimulated gastric acid secretion.


Subject(s)
Anti-Ulcer Agents/pharmacology , Benzimidazoles/pharmacology , Gastric Acid/metabolism , Omeprazole/pharmacology , 2-Pyridinylmethylsulfinylbenzimidazoles , Adult , Cross-Over Studies , Dose-Response Relationship, Drug , Female , Gastric Acidity Determination , Humans , Male , Postprandial Period/physiology , Rabeprazole , Treatment Outcome
3.
Neuroscience ; 111(3): 443-532, 2002.
Article in English | MEDLINE | ID: mdl-12031341

ABSTRACT

The distribution of neuropeptide Y (NPY) Y1 receptor-like immunoreactivity (Y1R-LI) has been studied in detail in the CNS of rat using a rabbit polyclonal antibody against the C-terminal 13 amino acids of the rat receptor protein. The indirect immunofluorescence technique with tyramide signal amplification has been employed. For specificity and comparative reasons Y1 knock-out mice and wild-type controls were analyzed. The distribution of Y1R mRNA was also studied using in situ hybridization. A limited comparison between Y1R-LI and NPY-LI was carried out.A widespread and abundant distribution of Y1R-LI, predominantly in processes but also in cell bodies, was observed. In fact, Y1R-LI was found in most regions of the CNS with a similar distribution pattern between rat and wild-type mouse. This staining was specific in the sense that it was absent in adjacent sections following preadsorption of the antibody with 10(-5) M of the antigenic peptide, and that it could not be observed in sections of the Y1 KO mouse. In contrast, the staining obtained with an N-terminally directed Y1R antiserum did not disappear, strongly suggesting unspecificity. In brief, very high levels of Y1R-LI were seen in the islands of Calleja, the anterior olfactory nucleus, the molecular layer of the dentate gyrus, parts of the habenula, the interpeduncular nucleus, the mammillary body, the spinal nucleus of the trigeminal, caudal part, the paratrigeminal nucleus, and superficial layers of the dorsal horn. High levels were found in most cortical areas, many thalamic nuclei, some subnuclei of the amygdaloid complex, the hypothalamus and the nucleus of the stria terminalis, the nucleus of the solitary tract, the parabrachial nucleus, and the inferior olive. Moderate levels of Y1R-LI were detected in the cornu Ammonis and the subicular complex, many septal, some thalamic and many brainstem regions. Y1R staining of processes, often fiber and/or dot-like, and occasional cell bodies was also seen in tracts, such as the lateral lemniscus, the rubrospinal tract and the spinal tract of the trigeminal. There was in general a good overlap between Y1R-LI and NPY-LI, but some exceptions were found. Thus, some areas had NPY innervation but apparently lacked Y1Rs, whereas in other regions Y1R-LI, but no or only few NPY-positive nerve endings could be detected. Our results demonstrate that NPY signalling through the Y1R is common in the rat (and mouse) CNS. Mostly the Y1R is postsynaptic but there are also presynaptic Y1Rs. Mostly there is a good match between NPY-releasing nerve endings and Y1Rs, but 'volume transmission' may be 'needed' in some regions. Finally, the importance of using proper control experiments for immunohistochemical studies on seven-transmembrane receptors is stressed.


Subject(s)
Central Nervous System/anatomy & histology , Central Nervous System/metabolism , Receptors, Neuropeptide Y/biosynthesis , Animals , Antibody Specificity , Central Nervous System/chemistry , Central Nervous System/cytology , Densitometry , Fluorescent Antibody Technique , Immunohistochemistry , In Situ Hybridization , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuropeptide Y/analysis , Neuropeptide Y/biosynthesis , Organ Specificity , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Receptors, Neuropeptide Y/analysis , Receptors, Neuropeptide Y/genetics , Tissue Distribution
4.
Transgenic Res ; 10(4): 329-41, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11592712

ABSTRACT

The goal of this study was to engineer gastrin-producing G cells of the gastric antrum to produce insulin. A pGas-Ins chimeric gene in which the gastrin promoter drives expression of the human insulin gene was constructed and was validated by transient transfection of GH4 and AGS cells. RT-PCR analysis and sequencing revealed three forms of differentially spliced insulin mRNA in GH4 cells transiently transfected by pGas-Ins. Gas-Ins transgenic mice were generated utilizing this chimeric gene. Northern blot analysis, in situ hybridization, and immunohistochemistry demonstrated expression of the human insulin gene specifically in antral G cells. Northern blot analysis demonstrated that the shortest of the insulin mRNA three forms is predominantly expressed in stomach tissue. RT-PCR analysis also showed expression of the transgene in colon, pancreas, and brain tissues that was undetectable by northern analysis. We conclude that gastrin promoter can be used for targeting expression of human insulin to antral G cells and that antral G cells can express human insulin. Further refining of the chimeric gene design is required to enhance expression.


Subject(s)
Gastrin-Secreting Cells/metabolism , Insulin/genetics , Animals , Blotting, Northern , Cells, Cultured , Chimera , DNA Primers/chemistry , Female , Gastrins/biosynthesis , Gene Expression , Humans , Immunoenzyme Techniques , In Situ Hybridization , Mice , Mice, Transgenic , Polymerase Chain Reaction , Pyloric Antrum/cytology , Pyloric Antrum/metabolism , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
5.
J Biol Chem ; 276(52): 48847-53, 2001 Dec 28.
Article in English | MEDLINE | ID: mdl-11677249

ABSTRACT

The structure of a small-molecule, non-peptide chemotactic factor has been determined from activity purified to apparent homogeneity from Helicobacter pylori supernatants. H. pylori was grown in brucella broth media until one liter of solution had 0.9 absorbance units. The culture was centrifuged, and the bacteria re-suspended in physiological saline and incubated at 37 degrees C for 4 h. A monocyte migration bioassay revealed the presence of a single active chemotactic factor in the supernatant from this incubation. The chemotactic factor was concentrated by solid phase chromatography and purified by reverse phase high pressure liquid chromatography. The factor was shown to be indistinguishable from diethyl phthalate (DEP) on the basis of multiple criteria including nuclear magnetic resonance spectroscopy, electron impact mass spectroscopy, UV visible absorption spectrometry, GC and high pressure liquid chromatography retention times, and chemotactic activity toward monocytes. Control experiments with incubated culture media without detectable bacteria did not yield detectable DEP, suggesting it is bacterially derived. It is not known if the bacteria produce diethyl phthalate de novo or if it is a metabolic product of a precursor molecule present in culture media. DEP produced by H. pylori in addition to DEP present in man-made products may contribute to the high levels of DEP metabolites observed in human urine. DEP represents a new class of chemotactic factor.


Subject(s)
Chemotactic Factors/metabolism , Chemotaxis, Leukocyte , Helicobacter pylori/metabolism , Monocytes/physiology , Phthalic Acids/metabolism , Animals , Cell Fractionation , Chemotactic Factors/chemistry , Chemotactic Factors/isolation & purification , Chemotactic Factors/pharmacology , Chromatography, High Pressure Liquid , Helicobacter pylori/chemistry , Humans , Magnetic Resonance Spectroscopy , Mass Spectrometry , Molecular Structure , Monocytes/drug effects , Phthalic Acids/chemistry , Phthalic Acids/isolation & purification , Phthalic Acids/pharmacology
6.
Neurogastroenterol Motil ; 13(3): 229-36, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11437985

ABSTRACT

This study aimed to characterize the role of the neuropeptide calcitonin gene-related peptide (CGRP) in the development of mechanically induced visceral hyperalgesia. Tonic colorectal distension (CRD) was performed in fasted, conscious male Sprague-Dawley rats. The visceromotor reflex associated with noxious CRD was determined as the number of contractions during each of two consecutive tonic distensions (10 min at 60 mmHg), which were separated by a series of phasic distensions (repeated 15-s distensions to 80 mmHg at 30-s intervals). The effect of the CGRP receptor antagonist h-CGRP8-37 given intrathecally (i.t.) (0.03-3 nmol rat-1) or intravenously (i.v.) (20 microg kg-1 bodyweight [bw]) on the visceromotor response was evaluated. The dose for i.v. administration was chosen based on previous results from similar studies. In addition, the effect of a CGRP monoclonal antibody (6 mg kg-1 bw) given intravenously was evaluated. Compared to the baseline response, a significant increase in the number of abdominal contractions was observed during the second tonic distension. The i.t. application of h-CGRP8-37 dose-dependently reduced the numbers of abdominal contractions both during the first and the second tonic distension period, with a maximum effect observed at a peptide concentration of 3 nmol. Intravenous administration of h-CGRP8-37 or of the CGRP antiserum produced a small reduction of the visceromotor response induced by the second tonic distension and had no effect on colonic compliance. The development of mechanically induced colorectal hyperalgesia by repeated tonic distension involves the spinal release of CGRP, while peripheral release of CGRP plays only a minor role.


Subject(s)
Calcitonin Gene-Related Peptide/pharmacology , Colon/physiology , Hyperalgesia/physiopathology , Miotics/pharmacology , Peptide Fragments/pharmacology , Spinal Cord/physiology , Visceral Afferents/metabolism , Acute Disease , Animals , Antibodies, Monoclonal/pharmacology , Calcitonin Gene-Related Peptide/immunology , Calcitonin Gene-Related Peptide Receptor Antagonists , Catheterization , Colon/innervation , Gastrointestinal Motility/drug effects , Gastrointestinal Motility/physiology , Injections, Intravenous , Injections, Spinal , Male , Muscle, Smooth/innervation , Muscle, Smooth/physiology , Peptide Fragments/immunology , Rats , Rats, Sprague-Dawley , Receptors, Calcitonin Gene-Related Peptide/metabolism
7.
Hybridoma ; 20(2): 109-15, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11394529

ABSTRACT

A monoclonal antibody (MAb) to galanin was prepared by cell fusion of myeloma Fox-NY and spleen cells from Robertsonian mice immunized with rat galanin. Hybridomas producing high-affinity antibodies were cloned in pristine-primed Balb/c mice. The antibody was purified by affinity chromatography and concentrated to 12 mg IgG/mL by dialysis. Immunoreactivity of the antibody was screened by radioimmunoassay. Ascites fluid contained approximately 10 mg/mL IgG that belong to the subclass of IgG2a as determined by enzyme-linked immunoadsorbent assay (ELISA). The titer of this IgG2a antibody entitled #G65G was 1:10,000 and the ID50 for rat galanin was 1000 fmol/mL as determined by liquid phase radioimmunoassay. Immunohistochemistry showed that this galanin MAb stains densed, beaded processes distributed to the enteric plexuses, where they appear to encircle neuronal cell bodies, to the muscle layer, where they are particularly abundant in the circular muscle layer and in the deep muscular layer, and to the mucosa. In vivo capacity of immunoneutralization by this antibody was tested in male Sprague-Dawley rats fasted for 24 h and anesthetized with urethane. Systemic injection of protein A purified galanin antibody (6 mg/rat) decreased by 70% of the inhibitory effect of intravenous galanin (2 nmol/kg/h i.v.) on gastric acid secretion induced by intracisternal TRH analog. These results show that galanin antibody #G65G is useful for in vivo immunoneutralization of galanin effects and is a valuable tool for immunohistochemical localization of galanin in gastrointestinal tissues.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Galanin/immunology , Animals , Antibodies, Monoclonal/pharmacology , Antibody Specificity , Galanin/administration & dosage , Galanin/antagonists & inhibitors , Gastric Acid/metabolism , Hybridomas , Immunoglobulin G , Immunoglobulin Isotypes , Immunohistochemistry , Intestines/chemistry , Male , Neurons/chemistry , Neutralization Tests , Rats , Rats, Sprague-Dawley
8.
Exp Cell Res ; 267(1): 37-44, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11412036

ABSTRACT

G-protein-coupled receptors (GPCRs) are membrane proteins that exhibit a decreased mobile fraction compared to a freely mobile plasma membrane protein. Recently, interest has focused on proteins other than heterotrimeric G-proteins that interact with GPCRs as scaffolding structures that affect receptor signal transduction. In order to investigate the physical state of receptors before and after agonist, we used fluorescence recovery after photobleaching of the bombesin/gastrin-releasing peptide (GRP) receptor fused to the intrinsically fluorescent green fluorescent protein (GFP-GRP receptor) expressed in KNRK cells to measure both the fraction of mobile receptors and their diffusion rate before and after agonist stimulation. In live cells at 37 degrees C, addition of GRP (100 nM) caused a rapid decrease in GFP-GRP receptor mobile fraction from 0.8 +/- 0.1 to 0.49 +/- 0.05, which was independent of endocytosis. Concurrently, the remaining mobile GFP-GRPreceptors showed an increase in the diffusion rate with the half-time of fluorescent recovery, tau(1/2) = 46 +/- 7 s for untreated cells, decreasing to tau(1/2) = 30 +/- 6 s for cells treated with GRP. Prior treatment with the Src-specific inhibitor PP-2 (10 microM) blocked GFP-GRP receptor immobilization while treatment with the inactive analog PP-3 (10 microM) did not affect receptor immobilization. These data suggest that agonist-bound GPCR have increased plasma membrane diffusion rates but an increased affinity for immobilization into a multiprotein complex that is mediated by Src activity.


Subject(s)
Cell Membrane/metabolism , Receptors, Bombesin/metabolism , Diffusion , Endocytosis , Green Fluorescent Proteins , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Membrane Fluidity , Oncogene Protein pp60(v-src)/antagonists & inhibitors , Protein Transport , Receptors, Bombesin/agonists , Receptors, Bombesin/genetics , Recombinant Fusion Proteins/agonists , Recombinant Fusion Proteins/metabolism , src-Family Kinases/antagonists & inhibitors
9.
Annu Rev Physiol ; 63: 49-76, 2001.
Article in English | MEDLINE | ID: mdl-11181948

ABSTRACT

Gastrin, produced by G cells in the gastric antrum, has been identified as the circulating hormone responsible for stimulation of acid secretion from the parietal cell. Gastrin also acts as a potent cell-growth factor that has been implicated in a variety of normal and abnormal biological processes including maintenance of the gastric mucosa, proliferation of enterochromaffin-like cells, and neoplastic transformation. Here, we review the models used to study the effects of gastrin on cell proliferation in vivo and in vitro with respect to mechanisms by which this hormone might influence normal and cancerous cell growth. Specifically, human and animal models of hypergastrinemia and hypogastrinemia have been described in vivo, and several cells that express cholecystokinin (CCK)B/gastrin receptors have been used for analysis of intracellular signaling pathways initiated by biologically active amidated gastrins. The binding of gastrin or CCK to their common cognate receptor triggers the activation of multiple signal transduction pathways that relay the mitogenic signal to the nucleus and promote cell proliferation. A rapid increase in the synthesis of lipid-derived second messengers with subsequent activation of protein phosphorylation cascades, including mitogen-activated protein kinase, is an important early response to these signaling peptides. Gastrin and CCK also induce rapid Rho-dependent actin remodeling and coordinate tyrosine phosphorylation of cellular proteins including the non-receptor tyrosine kinases p125fak and Src and the adaptor proteins p130cas and paxillin. This article reviews recent advances in defining the role of gastrin and CCK in the control of cell proliferation in normal and cancer cells and in dissecting the signal transduction pathways that mediate the proliferative responses induced by these hormonal GI peptides in a variety of normal and cancer cell model systems.


Subject(s)
Cholecystokinin/physiology , Gastrins/physiology , Neoplasms/physiopathology , Signal Transduction/physiology , Animals , Humans
10.
J Immunol ; 166(3): 2063-70, 2001 Feb 01.
Article in English | MEDLINE | ID: mdl-11160257

ABSTRACT

The development of allergic asthma is influenced by both genetic and environmental factors. Epidemiologic data often show no clear relationship between the levels of allergen and clinical symptoms. Recent data suggest that bacterial LPS may be a risk factor related to asthma severity. Airborne LPS is typically present at levels that are insufficient to activate alveolar macrophages in the absence of the accessory molecule LPS binding protein (LBP). LBP levels are markedly elevated in bronchoalveolar lavage fluids obtained from asthmatic subjects compared with those in normal controls. We hypothesized that LBP present in the lung could augment the pulmonary inflammation and airway reactivity associated with allergic asthma by sensitizing alveolar macrophages to LPS or other bacterial products and triggering them to release proinflammatory mediators. We compared wild-type (WT) and LBP-deficient mice using a defined Ag immunization and aerosol challenge model of allergic asthma. Immunized LBP-deficient mice did not develop substantial Ag-induced airway reactivity, whereas WT mice developed marked bronchoconstriction following aerosol Ag sensitization and challenge with methacholine. Similarly, production of NO synthase 2 protein and the NO catabolite peroxynitrite was dramatically higher in the lungs of WT mice following challenge compared with that in LBP-deficient mice. Thus, NO production appears to correlate with airway reactivity. In contrast, both mice developed similar pulmonary inflammatory cell infiltrates and elevated mucin production. Thus, LBP appears to participate in the development of Ag-induced airway reactivity and peroxynitrite production, but does not seem to be required for the development of pulmonary inflammation.


Subject(s)
Acute-Phase Proteins , Adjuvants, Immunologic/physiology , Allergens/immunology , Asthma/immunology , Bronchial Hyperreactivity/etiology , Carrier Proteins/physiology , Lipopolysaccharides/metabolism , Membrane Glycoproteins , Adjuvants, Immunologic/deficiency , Adjuvants, Immunologic/genetics , Aerosols , Allergens/administration & dosage , Animals , Asthma/genetics , Asthma/metabolism , Asthma/pathology , Bronchial Hyperreactivity/genetics , Bronchial Hyperreactivity/metabolism , Bronchial Hyperreactivity/pathology , Carrier Proteins/genetics , Disease Models, Animal , Immunoglobulin E/biosynthesis , Immunoglobulin G/biosynthesis , Inflammation/etiology , Inflammation/pathology , Injections, Intraperitoneal , Lung/enzymology , Lung/immunology , Lung/metabolism , Lung/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Nitrates/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Ovalbumin/administration & dosage , Ovalbumin/immunology
11.
J Comp Neurol ; 429(2): 183-91, 2001 Jan 08.
Article in English | MEDLINE | ID: mdl-11116213

ABSTRACT

Using immunohistochemistry in combination with confocal laser scanning microscopy, we studied the ontogeny of neuropeptide Y-Y1 receptor (Y1-R) expression in the trigeminal system of the rat. The study was limited to the nerve fibers innervating the mystacial pad and the trigeminal ganglia. In the trigeminal ganglia, Y1-R-immunoreactive (IR) neurons were first observed at E16.5. At this same stage some nerve fibers in the trigeminal ganglia also exhibited Y1-R-like immunoreactivity (LI). Strongly Y1-R-IR nerve fibers innervating the follicles of the mystacial vibrissae were first observed at E18. After double labeling, the Y1-R-LI was found to be colocalized with the neuronal marker protein gene product 9.5. At P1 only weak labeling for the Y1-R was found around the vibrissae follicles, whereas the neurons in the trigeminal ganglia were intensely labeled. The same was true for the adult rat, but at this stage no Y1-R labeling at all was observed in nerve fibers around the vibrissal follicles. These results strongly support an axonal localization of the Y1-R at this developmental stage. The transient expression of the Y1-R during prenatal mystacial pad development suggests a role for the Y1-R in the functional development of the vibrissae.


Subject(s)
Axons/metabolism , Receptors, Neuropeptide Y/metabolism , Trigeminal Nerve/growth & development , Trigeminal Nerve/metabolism , Vibrissae/growth & development , Vibrissae/innervation , Animals , Animals, Newborn , Hair Follicle/metabolism , Immunohistochemistry , Male , Microscopy, Confocal , Rats , Rats, Sprague-Dawley , Thiolester Hydrolases/metabolism , Trigeminal Nerve/embryology , Trigeminal Nerve/ultrastructure , Ubiquitin Thiolesterase , Vibrissae/embryology
12.
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
13.
Am J Physiol Cell Physiol ; 279(6): C2019-27, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11078719

ABSTRACT

The role of actin organization in occupancy-induced receptor internalization remains poorly defined. Here we report that treatment of mouse Swiss 3T3 cells with latrunculin A, a potent inhibitor of actin polymerization (including cortical actin), inhibited the internalization of the endogenous bombesin/gastrin-releasing peptide (GRP) receptor, as judged by uptake of (125)I-labeled GRP or fluorescent Cy3-labeled bombesin. In contrast, cells pretreated with cytochalasin D showed minimal inhibition of bombesin/GRP receptor internalization. Similarly, pretreatment of Swiss 3T3 cells with the potent Rho-kinase inhibitor HA-1077, at concentrations (10-20 microM) that abrogated bombesin-mediated stress fiber formation, did not significantly alter receptor-mediated internalization of (125)I-GRP. These results indicate that bombesin/GRP receptor internalization depends on latrunculin A-sensitive cortical actin rather than on rapidly turning over actin stress fibers that are disrupted by either cytochalasin D or HA-1077. The rates and total levels of internalization of the endogenously expressed endothelin A receptor and epidermal growth factor receptor were also markedly reduced by latrunculin A in Swiss 3T3 cells. The potency of latrunculin A for inhibiting G protein-coupled receptor endocytosis was comparable to that for reducing internalization of the epidermal growth factor tyrosine kinase receptor. We conclude that cortical actin structures, disrupted by latrunculin A, are necessary for occupancy-induced receptor internalization in animal cells.


Subject(s)
1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , Actins/metabolism , ErbB Receptors/metabolism , Receptors, Bombesin/metabolism , Receptors, Endothelin/metabolism , Signal Transduction/physiology , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , 3T3 Cells , Animals , Bombesin/pharmacokinetics , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cytochalasin D/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Epidermal Growth Factor/pharmacokinetics , GTP-Binding Proteins/metabolism , Gastrin-Releasing Peptide/pharmacokinetics , Iodine Radioisotopes , Kinetics , Mice , Nucleic Acid Synthesis Inhibitors/pharmacology , Phosphorylation , Signal Transduction/drug effects , Thiazoles/pharmacology , Thiazolidines
14.
Biochem Biophys Res Commun ; 276(2): 406-10, 2000 Sep 24.
Article in English | MEDLINE | ID: mdl-11027488

ABSTRACT

Cyclooxygenase-2 (Cox-2) gene expression which is rapidly induced by cytokines, growth factors and tumor promoters, is important for inflammation, angiogenesis, and is markedly enhanced in various cancer cells. Many of these factors initiate signaling through Ras- and Rho-family small GTPases. Here, we investigated the ability of Ras, Rac, Rho, and Cdc42Hs to differentially regulate transcription from the murine COX-2 promoter in NIH 3T3 cells. Over-expression of constitutively active mutants of Ras, Rac, Rho, but not Cdc42Hs induced transcription from the COX-2 promoter. Transactivation by Rac and Rho required cis-acting elements located between -80 and -40 of the COX-2 promoter whereas deletion of this region enhanced transactivation by Ras. A CRE/ATF element located at -56 was critical for Ras- and Rac-induced transactivation of the COX-2 promoter, but was not required for transactivation by Rho. This demonstrates Rho-dependent transactivation of the COX-2 promoter through novel trans-acting elements and suggests that, in NIH 3T3 cells, signaling by small GTPases that result in COX-2 expression is not through a sequential pathway from Cdc42 to Rac to Rho, but rather through independent, parallel signaling pathways.


Subject(s)
Gene Expression Regulation , Isoenzymes/genetics , Monomeric GTP-Binding Proteins/physiology , Promoter Regions, Genetic/physiology , Prostaglandin-Endoperoxide Synthases/genetics , rho GTP-Binding Proteins/physiology , 3T3 Cells , Animals , Cyclooxygenase 2 , Mice , Repressor Proteins/physiology , Signal Transduction , Transcription, Genetic , Transcriptional Activation , cdc42 GTP-Binding Protein/physiology , rac GTP-Binding Proteins/physiology , ras Proteins/physiology
15.
Am J Physiol Gastrointest Liver Physiol ; 279(1): G126-31, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10898754

ABSTRACT

We synthesized PYY-(1-36) (nonselective between Y(1) and Y(2) receptor subtype agonists), [Pro(34)]PYY (selective for Y(1)), and PYY-(3-36) (selective for Y(2)) to determine whether solution conformation plays a role in receptor subtype selectivity. The three peptides exhibited the expected specificities in displacing labeled PYY-(1-36) from cells transfected with Y(1) receptors (dissociation constants = 0.42, 0.21, and 1,050 nM, respectively) and from cells transfected with Y(2) receptors (dissociation constants = 0.03, 710, and 0.11 nM, respectively) for PYY-(1-36), [Pro(34)]PYY, and PYY-(3-36). Sedimentation equilibrium analyses revealed that the three PYY analogs were 80-90% monomer at the concentrations used for the subsequent circular dichroism (CD) and (1)H-nuclear magnetic resonance (NMR) studies. CD analysis measured helicities for PYY-(1-36), [Pro(34)]PYY, and PYY-(3-36) of 42%, 31%, and 24%, suggesting distinct differences in secondary structure. The backbone (1)H-NMR resonances of the three peptides further substantiated marked conformational differences. These patterns support the hypothesis that Y(1) and Y(2) receptor subtype binding affinities depend on the secondary and tertiary solution state structures of PYY and its analogs.


Subject(s)
Peptide YY/chemistry , Receptors, Neuropeptide Y/chemistry , Amino Acid Sequence , Animals , Binding Sites , Circular Dichroism , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Peptide Fragments , Protein Conformation , Swine , Ultracentrifugation
16.
Am J Physiol Gastrointest Liver Physiol ; 279(1): G148-56, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10898757

ABSTRACT

In rodents, cholecystokinin (CCK) induces pancreatic enzyme secretion and pancreas growth through its CCK(A) receptors. It is unknown whether occupation of the CCK(B) receptors present in pig and human pancreas can cause the same effects. This study evaluates CCK(B) receptor expression in rat, mouse, pig, and fetal human pancreata using Northern blot, Western blot, and immunofluorescence techniques. The reported 2.7-kb CCK(B) receptor mRNA transcript in the rat brain and gastric fundus is absent in pancreas; the message was, however, detected by RT-PCR and by a CCK(B) receptor antibody as an 80-kDa protein present uniquely in islet delta-cells. Proteins of 50 and 80 kDa appear in mouse pancreas, and proteins of 50 and 115 kDa appear in pig and human pancreas, respectively, all localized in islet delta-cells. Gastrin mRNAs are strongly present in fetal rat pancreas, and the hormone is localized in islets; both are repressed 10 days after birth. In conclusion, the CCK(B) receptors are present in pancreas of four species with exclusive location in islet delta-cells. In such a location, they could be indirectly involved in the control of enzyme secretion.


Subject(s)
Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Receptors, Cholecystokinin/metabolism , Somatostatin/metabolism , Animals , Blotting, Northern , Blotting, Western , Cell Division/physiology , Cholecystokinin/metabolism , Female , Fluorescent Antibody Technique, Indirect , Gastrins/metabolism , Gene Expression/physiology , Humans , Islets of Langerhans/chemistry , Male , Mice , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Receptor, Cholecystokinin B , Receptors, Cholecystokinin/analysis , Receptors, Cholecystokinin/genetics , Swine
17.
Aliment Pharmacol Ther ; 14(6): 651-68, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10848649

ABSTRACT

This review examines the evidence for the development of adverse effects due to prolonged gastric acid suppression with proton pump inhibitors. Potential areas of concern regarding long-term proton pump inhibitor use have included: carcinoid formation; development of gastric adenocarcinoma (especially in patients with Helicobacter pylori infection); bacterial overgrowth; enteric infections; and malabsorption of fat, minerals, and vitamins. Prolonged proton pump inhibitor use may lead to enterochromaffin-like cell hyperplasia, but has not been demonstrated to increase the risk of carcinoid formation. Long-term proton pump inhibitor treatment has not been documented to hasten the development or the progression of atrophic gastritis to intestinal metaplasia and gastric cancer, although long-term studies are required to allow definitive conclusions. At present, we do not recommend that patients be tested routinely for H. pylori infection when using proton pump inhibitors for prolonged periods. Gastric bacterial overgrowth does increase with acid suppression, but important clinical sequelae, such a higher rate of gastric adenocarcinoma, have not been seen. The risk of enteric infection may increase with acid suppression, although this does not seem to be a common clinical problem with prolonged proton pump inhibitor use. The absorption of fats and minerals does not appear to be significantly impaired with chronic acid suppression. However, vitamin B12 concentration may be decreased when gastric acid is markedly suppressed for prolonged periods (e.g. Zolllinger-Ellison syndrome), and vitamin B12 levels should probably be assessed in patients taking high-dose proton pump inhibitors for many years. Thus, current evidence suggests that prolonged gastric acid suppression with proton pump inhibitors rarely, if ever, produces adverse events. Nevertheless, continued follow-up of patients taking proton pump inhibitors for extended periods will provide greater experience regarding the potential gastrointestinal adverse effects of long-term acid suppression.


Subject(s)
Adenocarcinoma/chemically induced , Anti-Ulcer Agents/adverse effects , Carcinoid Tumor/chemically induced , Gastric Acid/metabolism , Proton Pump Inhibitors , Stomach Neoplasms/chemically induced , Anti-Ulcer Agents/administration & dosage , Helicobacter Infections/complications , Humans , Malabsorption Syndromes/chemically induced , Risk Factors , Stomach Diseases/chemically induced , Stomach Ulcer/drug therapy
18.
Aliment Pharmacol Ther ; 14(6): 701-8, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10848652

ABSTRACT

AIM: To compare acid inhibiting activity and duration of action of different doses of rabeprazole, a substituted benzimidazole characterized as a highly potent and irreversible H+, K+-ATPase inhibitor, administered for 7 days to subjects infected with Helicobacter pylori. METHODS: A total of 38 subjects (mean age 39.3 years) were enrolled in a single-centre, double-blind, randomized, crossover study. All subjects were confirmed positive for H. pylori by 14C urea breath test and ELISA serologies. Subjects were divided into two groups of 19 to receive two doses of rabeprazole, either 5 and 20 mg or 10 and 40 mg, and placebo, given in random order daily in the morning for 7 days. Peptone-stimulated acid, pH, and gastrin measurements were made for 24 h after the 1st dose and for 48 h after the 7th dose. RESULTS: Peptone-stimulated acid secretion rates were decreased from 12.5 to 6.7, 4.0, 1.5, and 0.26 h after initial 5, 10, 20, and 40 mg doses, respectively; to 7.3, 4.3, 2.1, and 1.2 mmol/h 23 h after the initial dose; and to 2.4, 2.6, 0.6, and 0.8 mmol/h 23 h after the 7th dose. After 48 h, stimulated acid secretion had recovered less than 40% for all treatment groups compared to placebo. Median intragastric pH also increased from 2.0 with placebo to 4.9, 6.2, 6.6 and 6.9 during the 24-h period after the 7th dose of 5, 10, 20, and 40 mg. The 20 mg dose of rabeprazole produced equivalent acid inhibition to the 40 mg dose with less increase in plasma gastrin. CONCLUSION: Rabeprazole in doses from 5 to 40 mg was a highly effective inhibitor of gastric acid secretion in subjects infected with H. pylori. The inhibition was rapid, dose-related, and long-acting, with less than 50% recovery of acid by 48 h after the 7th dose. The optimal acid inhibitory dose in these subjects appeared to be 20 mg daily, however 5 mg and 10 mg doses produced potent inhibition of gastric acid secretion.


Subject(s)
Anti-Ulcer Agents/pharmacology , Benzimidazoles/pharmacology , Gastric Acid/metabolism , Helicobacter Infections/complications , 2-Pyridinylmethylsulfinylbenzimidazoles , Administration, Oral , Adolescent , Adult , Aged , Anti-Ulcer Agents/administration & dosage , Anti-Ulcer Agents/therapeutic use , Benzimidazoles/administration & dosage , Benzimidazoles/therapeutic use , Cross-Over Studies , Dose-Response Relationship, Drug , Double-Blind Method , Female , Gastric Acidity Determination , Helicobacter pylori/pathogenicity , Humans , Male , Middle Aged , Omeprazole/analogs & derivatives , Peptones/pharmacology , Rabeprazole
19.
Am J Gastroenterol ; 95(4): 943-6, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10763942

ABSTRACT

OBJECTIVE: Pepsinogen 1 (PG1) is a proenzyme precursor to pepsin, a protease secreted by the gastric chief cell. PG1 levels correlate with maximal gastric acid output. In 1979, Rotter et al. reported two pedigrees in which elevated PG1 levels and duodenal ulcers were prevalent. They proposed autosomal dominant inheritance of elevated PG1 and suggested that it was a risk factor for duodenal ulcer disease. In 1982, Helicobacter pylori (Hp) was discovered and was shown to be an important factor in peptic ulcer disease. Hp infection is also associated with increased PGI levels. We tested serum from one of the original pedigrees for Hp antibodies to determine whether Hp infection could explain the ulcers and elevated PG1 levels. METHODS: ELISA tests were performed using the urease fraction of a crushed Hp extract. Banked serum from one of the original families was thawed and tested. RESULTS: Of the subjects, 90% (nine of 10) with elevated PG1 were seropositive for Hp, compared to only 31% (17 of 55) of those with normal PG1 levels (p < 0.001). The mean PG1 level was higher in the seropositive (94.1+/-13.3 ng/ml) than the seronegative subjects (54.8+/-3.6, p < 0.05). Three of the four subjects with ulcers were Hp-seropositive. The prevalence of Hp-seropositivity and elevated PG1 declined in parallel in each successive generation. When neither parent was seropositive, children were seronegative. CONCLUSIONS: The etiology of elevated PG1 levels in this pedigree is more likely due to Helicobacter pylori infection than to a genetic predisposition.


Subject(s)
Duodenal Ulcer/genetics , Genetic Predisposition to Disease/genetics , Helicobacter Infections/genetics , Helicobacter pylori , Pepsin A/blood , Adult , Chromosome Aberrations/genetics , Chromosome Disorders , Duodenal Ulcer/diagnosis , Enzyme-Linked Immunosorbent Assay , Female , Genes, Dominant/genetics , Helicobacter Infections/diagnosis , Humans , Male , Middle Aged , Risk Factors
20.
Gastroenterology ; 118(4): 696-704, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10734021

ABSTRACT

BACKGROUND & AIMS: Parenteral control of gastric acid hypersecretion in conditions such as Zollinger-Ellison syndrome (ZES) or idiopathic gastric acid hypersecretion is necessary perioperatively or when oral medications cannot be taken for other reasons (e.g., during chemotherapy, acute upper gastrointestinal bleeding, or in intensive care unit settings). METHODS: We evaluated the efficacy and safety of 15-minute infusions of the proton pump inhibitor pantoprazole (80-120 mg every 8-12 hours) in controlling acid output for up to 7 days. Effective control was defined as acid output >10 milliequivalents per hour (mEq/h) (<5 mEq/h in patients with prior acid-reducing surgery) for 24 hours. RESULTS: The 21 patients enrolled had a mean age of 51.9 years (range, 29-75) and a mean disease duration of 8.1 years (range, <0.5-21); 13 were male, 7 had multiple endocrine neoplasia syndrome type I, 4 had undergone acid-reducing surgery, 2 had received chemotherapy, and 13 had undergone gastrinoma resections without cure. Basal acid output (mean +/- SD) was 40.2 +/- 27.9 mEq/h (range, 11.2-117.9). In all patients, acid output was controlled within the first hour (mean onset of effective control, 41 minutes) after an initial 80-mg intravenous pantoprazole dose. Pantoprazole, 80 mg every 12 hours, was effective in 17 of 21 patients (81%) for up to 7 days. Four patients required upward dose titration, 2 required 120 mg pantoprazole every 12 hours, and 2 required 80 mg every 8 hours. At study end, acid output remained controlled for 6 hours beyond the next expected dose in 71% of patients (n = 15); mean acid output increased to 4.0 mEq/h (range, 0-9.7). No serious or unexpected adverse events were observed. CONCLUSIONS: Intravenous pantoprazole, 160-240 mg/day administered in divided doses by 15-minute infusion, rapidly and effectively controlled acid output within 1 hour and maintained control for up to 7 days in all ZES patients.


Subject(s)
Anti-Ulcer Agents/therapeutic use , Benzimidazoles/administration & dosage , Gastric Acid/metabolism , Sulfoxides/administration & dosage , Zollinger-Ellison Syndrome/drug therapy , Zollinger-Ellison Syndrome/metabolism , 2-Pyridinylmethylsulfinylbenzimidazoles , Adult , Aged , Benzimidazoles/adverse effects , Benzimidazoles/therapeutic use , Female , Humans , Injections, Intravenous , Male , Middle Aged , Omeprazole/analogs & derivatives , Pantoprazole , Sulfoxides/adverse effects , Sulfoxides/therapeutic use , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...