Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
Biochim Biophys Acta Mol Basis Dis ; 1870(2): 166966, 2024 02.
Article in English | MEDLINE | ID: mdl-37995775

ABSTRACT

BACKGROUND AND AIMS: The stiffening of the extracellular matrix, and changes in its cellular and molecular composition, have been reported in the pathogenesis of fibrosis. We analyze the mechanisms that perpetuate ileal fibrosis in surgical resections of complicated Crohn's disease patients. METHODS: Ileal resections were obtained from affected and non-affected tissue of stenotic or penetrating Crohn's disease behavior. Ilea from non-IBD patients were used as control tissue. All samples underwent RNA sequencing. Human small intestinal fibroblasts were treated for 48 h with IL-1ß, TFGß1, PDGFB or TNF-α. Resistance to apoptosis was analysed by RT-PCR, western blot and immunohistochemistry in ileal tissue and by RT-PCR and FACS in cultured cells. RESULTS: Growth factor-driven signaling pathways and increased RAS GTPase activity were up-regulated in affected ilea in which we found expression of both the antiapoptotic molecule MCL1 and the transcription factor ETS1 in submucosal fibroblasts, and a senescence-associated secretory phenotype. In cultured intestinal fibroblasts, PDGFB induced an ETS1-mediated resistance to apoptosis that was associated with the induction of both of TGFB1 and IL1B, a cytokine that replicated the expression of SASP detected in ileal tissue. ETS1 drove fibroblast polarization between inflammatory and fibrogenic phenotypes in IL1ß-treated cells. CONCLUSIONS: Our data show resistance to apoptosis in complicated ileal CD, and demonstrate that PDGFB induce an ETS1-mediated resistance to apoptosis associated with an inflammatory and fibrogenic pattern of expression in intestinal fibroblasts. Results point to PDGFRB, IL1R1 or MCL1 as potential targets against ileal fibrosis.


Subject(s)
Crohn Disease , Humans , Crohn Disease/complications , Crohn Disease/genetics , Crohn Disease/metabolism , Proto-Oncogene Proteins c-sis , Myeloid Cell Leukemia Sequence 1 Protein , Apoptosis , Fibrosis
2.
J Crohns Colitis ; 10(5): 582-92, 2016 May.
Article in English | MEDLINE | ID: mdl-26802079

ABSTRACT

BACKGROUND: The Notch signalling pathway plays an essential role in mucosal regeneration, which constitutes a key goal of Crohn's disease (CD) treatment. Macrophages coordinate tissue repair and several phenotypes have been reported which differ in the expression of surface proteins, cytokines and hypoxia-inducible factors (HIFs). We analysed the role of HIFs in the expression of Notch ligands in macrophages and the relevance of this pathway in mucosal regeneration. METHODS: Human monocytes and U937-derived macrophages were polarized towards the M1 and M2 phenotypes and the expression levels of HIF-1α, HIF-2α, Jagged 1 (Jag1) and delta-like 4 (Dll4) were evaluated. The effects of macrophages on the expression of hairy and enhancer of split-1 (HES1, the main target of Notch signalling) and intestinal alkaline phosphatase (IAP, enterocyte marker) in epithelial cells in co-culture were also analysed. Phenotype macrophage markers and Notch signalling were evaluated in the mucosa of CD patients. RESULTS: M1 macrophages were associated with HIF-1-dependent induction of Jag1 and Dll4, which increased HES1 protein levels and IAP activity in co-cultured epithelial cells. In the mucosa of CD patients a high percentage of M1 macrophages expressed both HIF-1α and Jag1 while M2 macrophages mainly expressed HIF-2α and we detected a good correlation between the ratio of M1/M2 macrophages and both HES1 and IAP protein levels. CONCLUSION: M1, but not M2, macrophages are associated with HIF-1-dependent induction of Notch ligands and activation of epithelial Notch signalling pathway. In the mucosa of chronic CD patients, the prevalence of M2 macrophages is associated with diminution of Notch signalling and impaired enterocyte differentiation.


Subject(s)
Colon/metabolism , Crohn Disease/metabolism , Intestinal Mucosa/metabolism , Macrophages/metabolism , Receptors, Notch/metabolism , Adolescent , Adult , Biomarkers/metabolism , Caco-2 Cells , Case-Control Studies , Coculture Techniques , Colon/pathology , Crohn Disease/pathology , Cytokines/metabolism , Epithelial Cells/metabolism , Female , HT29 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Intestinal Mucosa/pathology , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Signal Transduction , Young Adult
3.
Mucosal Immunol ; 9(4): 986-98, 2016 07.
Article in English | MEDLINE | ID: mdl-26601901

ABSTRACT

The complete repair of the mucosa constitutes a key goal in inflammatory bowel disease (IBD) treatment. The Wnt signaling pathway mediates mucosal repair and M2 macrophages that coordinate efficient healing have been related to Wnt ligand expression. Signal transducer and activator of transcription 6 (STAT6) mediates M2 polarization in vitro and we hypothesize that a STAT6-dependent macrophage phenotype mediates mucosal repair in acute murine colitis by activating the Wnt signaling pathway. Our results reveal an impaired mucosal expression of M2 macrophage-associated genes and delayed wound healing in STAT6(-/-) mice treated with 2,4,6-trinitrobenzenesulfonic acid (TNBS). These mice also exhibited decreased mucosal expression of Wnt2b, Wnt7b, and Wnt10a, diminished protein levels of nuclear ß-catenin that is mainly located in crypts adjacent to damage, and reduced mRNA expression of two Wnt/ß-catenin target molecules Lgr5 and c-Myc when compared with wild-type (WT) mice. Murine peritoneal macrophages treated with interleukin-4 (IL-4) and polarized toward an M2a phenotype overexpressed Wnt2b, Wnt7b, and Wnt10a in a STAT6-dependent manner. Administration of a Wnt agonist as well as transfer of properly polarized M2a macrophages to STAT6(-/-) mice activated the Wnt signaling pathway in the damaged mucosa and accelerated wound healing. Our results demonstrate that a STAT6-dependent macrophage phenotype promotes mucosal repair in TNBS-treated mice through activation of the Wnt signaling pathway.


Subject(s)
Colitis/immunology , Inflammatory Bowel Diseases/immunology , Intestinal Mucosa/immunology , Macrophages, Peritoneal/immunology , STAT6 Transcription Factor/metabolism , Animals , Cell Differentiation , Cells, Cultured , Colitis/chemically induced , Humans , Intestinal Mucosa/pathology , Macrophages, Peritoneal/transplantation , Mice , Mice, Inbred BALB C , Mice, Knockout , Phenotype , STAT6 Transcription Factor/genetics , Signal Transduction , Trinitrobenzenesulfonic Acid , Wnt Proteins/metabolism , Wound Healing
4.
Mucosal Immunol ; 7(4): 929-38, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24301659

ABSTRACT

A defective induction of epithelial autophagy may have a role in the pathogenesis of inflammatory bowel diseases. This process is regulated mainly by extracellular factors such as nutrients and growth factors and is highly induced by diverse situations of stress. We hypothesized that epithelial autophagy is regulated by the immune response that in turn is modulated by local hypoxia and inflammatory signals present in the inflamed mucosa. Our results reveal that HIF-1α and Wnt1 were co-localized with CD68 in cells of the mucosa of IBD patients. We have observed increased protein levels of ß-catenin, phosphorylated mTOR, and p62 and decreased expression of LC3II in colonic epithelial crypts from damaged mucosa in which ß-catenin positively correlated with phosphorylated mTOR and negatively correlated with autophagic protein markers. In cultured macrophages, HIF-1 mediated the increase in Wnt1 expression induced by hypoxia, which enhanced protein levels of ß-catenin, activated mTOR, and decreased autophagy in epithelial cells in co-culture. Our results demonstrate a HIF-1-dependent induction of Wnt1 in hypoxic macrophages that undermines autophagy in epithelial cells and suggest a role for Wnt signaling and mTOR pathways in the impaired epithelial autophagy observed in the mucosa of IBD patients.


Subject(s)
Autophagy , Epithelial Cells/metabolism , Macrophages/immunology , Macrophages/metabolism , Wnt1 Protein/metabolism , Adolescent , Adult , Cell Hypoxia , Female , Gene Expression Regulation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Middle Aged , TOR Serine-Threonine Kinases/metabolism , Wnt Signaling Pathway , Wnt1 Protein/genetics , Young Adult
5.
Br J Pharmacol ; 159(8): 1636-45, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20233223

ABSTRACT

BACKGROUND AND PURPOSE: Nitric oxide (NO) modulates expression of hypoxia inducible factor-1 (HIF-1), a transcription factor regulating function of myeloid cells. Here, we have assessed the role played by NO, formed by inducible NOS (iNOS), in the inflammation induced by aspirin in the gut, by modulating HIF-1 activity. EXPERIMENTAL APPROACH: The role of iNOS-derived NO on leucocyte-endothelial interactions induced by aspirin was evaluated by intravital microscopy in mesenteric venules of rats pretreated with selective iNOS inhibitors, 1400W or l-N6-(1-iminoethyl)-lysine. NO was localized by fluorescence microscopy, using DAF-FM. iNOS, HIF-1alpha and CD36 were localized by immunohistochemistry. KEY RESULTS: Leucocyte-endothelial interactions increased at 6 h and returned to normal levels 24 h after aspirin administration. Numbers of migrated leucocytes were similar between 6 and 24 h after aspirin. iNOS expression and iNOS-derived NO synthesis were observed in leucocytes of the mesentery of aspirin-treated rats. Blockade of iNOS activity in aspirin-treated rats: (i) did not modify leucocyte infiltration at 6 h, but reduced the number of polymorphonuclear leucocyte and increased that of macrophages at 24 h; (ii) increased HIF-1alpha immunostaining in macrophages of the mesentery; and (iii) prevented the decrease in CD36 immunostaining induced by aspirin in these cells. CONCLUSIONS AND IMPLICATIONS: NO, associated with acute gut inflammation induced by aspirin, diminished HIF-1alpha stabilization in macrophages. Early inhibition of iNOS-derived NO synthesis, by increasing the activity of HIF-1 in these cells, may accelerate the clearance of leucocytes.


Subject(s)
Aspirin/toxicity , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation/chemically induced , Mesentery/drug effects , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide/physiology , Animals , Enzyme Inhibitors/pharmacology , Immunohistochemistry , Male , Mesentery/pathology , Microscopy, Fluorescence , Nitric Oxide Synthase Type II/antagonists & inhibitors , Rats , Rats, Sprague-Dawley
6.
Br J Pharmacol ; 156(2): 262-72, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19076725

ABSTRACT

BACKGROUND AND PURPOSE: Mucosal microcirculation is compromised during gastric damage induced by non-steroidal anti-inflammatory drugs, such as aspirin. Consequently, oxygen supply to epithelial cells is decreased. The trefoil factor (TFF) peptides are involved in mechanisms of defence and repair in the gastrointestinal tract but their regulation at sites of gastric injury is unknown. EXPERIMENTAL APPROACH: Hypoxia and expression of TFF genes and peptides were measured in the damaged stomach of aspirin-treated rats. In a human gastric cell line (AGS cells), the effects of hypoxia and of hypoxia inducible factor (HIF)-1 (through transient transfection of HIF-1alpha siRNA or over-expression of HIF-1alpha) on TFF gene expression were evaluated. KEY RESULTS: Hypoxyprobe immunostaining, up-regulation of TFF2 (1.9-fold) and TFF3 (1.8-fold) and a non-significant increase of TFF1 (1.5-fold) mRNA were observed in the damaged stomach of aspirin-treated rats, compared with control animals. Hypoxia (3% O(2), 16 h) induced mRNA for TFF1 (5.8-fold), TTF2 (9.1-fold) and TFF3 (9.3-fold) in AGS cells, an effect mediated by HIF-1, as transient transfection of HIF-1alpha siRNA reduced the effects of hypoxia. Over-expression of HIF-1alpha by transfection in non-hypoxic epithelial cells produced a similar pattern of TFF induction to that observed with hypoxia and transactivated a TFF1 reporter construct. CONCLUSIONS AND IMPLICATIONS: Hypoxia inducible factor-1 mediated the induction of TFF gene expression by hypoxia in gastric epithelial cells. Low oxygen levels and up-regulation of TFF gene expression in the damaged stomach of aspirin-treated rats suggest that hypoxia induced expression of TFF genes at sites of gastric injury.


Subject(s)
Epithelial Cells/drug effects , Gastric Mucosa/drug effects , Hypoxia-Inducible Factor 1/physiology , Peptides/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/toxicity , Aspirin/toxicity , Cell Hypoxia , Cell Line , Epithelial Cells/metabolism , Gastric Mucosa/cytology , Gastric Mucosa/metabolism , Humans , Hypoxia-Inducible Factor 1/genetics , Male , Peptides/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Trefoil Factor-2 , Up-Regulation
7.
Curr Pharm Des ; 12(34): 4525-35, 2006.
Article in English | MEDLINE | ID: mdl-17168758

ABSTRACT

After bacterial infection, the host reacts by signalling to the central nervous system where a cascade of physiologic, neuroendocrine and behavioural processes is orchestrated, collectively termed the acute phase response. Endotoxemia following Gram-negative bacterial infection induces a wide array of effects, including fever, loss of appetite and changes in gastrointestinal function that attempt to eliminate the challenge and restore homeostasis. Systemic administration of low doses of endotoxin (5-40 microg/kg) to rats is associated with changes in gastrointestinal motor function, inhibition of gastric acid secretion and increase in the gastric mucosal resistance to damage. These changes are rapid in onset (observed within one hour), not related to vascular dysfunction, and appear to be mediated by mechanisms that involve the peripheral and the central nervous system. Nitric oxide (NO) plays a central role in the physiology of the gastrointestinal tract and its response to illness. Accumulated evidence supports an increase of NO synthesis in the brainstem, as well as in the gastric myenteric plexus thirty minutes after endotoxin administration. Such a synthesis is due to constitutive nitric oxide synthase (NOS) and occurs before the induction of NOS takes place. In this review we provide experimental evidence supporting the hypothesis that activation of a physiologic mechanism, mediated by the autonomic and the central nervous systems as well as constitutive NOS isoforms, is involved in acute changes of gastrointestinal function during early endotoxemia.


Subject(s)
Endotoxemia/physiopathology , Gastrointestinal Tract/innervation , Nitrergic Neurons/metabolism , Nitric Oxide/metabolism , Animals , Central Nervous System/metabolism , Endotoxemia/metabolism , Gastric Acid/metabolism , Gastric Mucosa/metabolism , Gastrointestinal Motility , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/physiopathology , Humans , Myenteric Plexus/metabolism , Nitric Oxide Synthase/metabolism , Rats , Regional Blood Flow , Vagus Nerve/metabolism
9.
Br J Pharmacol ; 134(2): 325-32, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11564650

ABSTRACT

1. This study analyses the neural pathway involved in the modulation of gastric motor function by stress. 2. Systemic administration of low doses of endotoxin (40 microg kg(-1), i.v.) prevents the increase in gastric tone induced by 2-deoxy-D-glucose (200 mg kg(-1), i.v., 2-DG) in urethane-anaesthetized rats. 3. Functional inhibition of afferent neurones by systemic administration of capsaicin (20+30+50 mg kg(-1), i.m.) in adult rats prevented the inhibitory effects of endotoxin. 4. Pre-treatment with the nitric oxide synthase (NOS) inhibitor, N(G)-nitro-L-arginine methyl ester (L-NAME), both i.v. (10 mg kg(-1)) and i.c. (200 microg rat(-1)), prevented the inhibitory effects of endotoxin on gastric tone induced by 2-DG. 5. Immunohistochemical studies show Fos expression in the dorsal vagal complex (DVC) of the brainstem of 2-DG-treated animals. Peripheral administration of endotoxin (40 microg kg(-1), i.p.) increased the number of Fos-immunoreactive cells induced by 2-DG, both in the nucleus tractus solitarii (NTS) and in the dorsal motor nucleus (DMN) of the DVC. Pre-treatment with L-NAME prevented the increase in Fos expression induced by endotoxin in both nuclei. 6. Endotoxin (40 microg kg(-1), i.p.) increased Ca(2+)-dependent nitric oxide synthase (cNOS) activity in the brainstem. Addition of 7-nitroindazole (600 microM, 7-NI) to the assay significantly inhibited the increase in cNOS activity caused by endotoxin. No change in NOS activity of any isoform was observed in the stomach of animals treated with endotoxin. 7. The present study suggests that inhibition of gastric motor function by low doses of endotoxin involves activation of capsaicin-sensitive afferent neurones and neuronal NOS in the brainstem.


Subject(s)
Brain/enzymology , Endotoxins/pharmacology , Gastrointestinal Motility/drug effects , Nitric Oxide Synthase/metabolism , Animals , Brain/drug effects , Brain/metabolism , Brain Stem/drug effects , Brain Stem/enzymology , Brain Stem/metabolism , Capsaicin/pharmacology , Deoxyglucose/pharmacology , Enzyme Inhibitors/pharmacology , Male , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Pressure , Proto-Oncogene Proteins c-fos/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Stomach/drug effects , Stomach/physiology
10.
Naunyn Schmiedebergs Arch Pharmacol ; 363(3): 276-80, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11284441

ABSTRACT

The effects of endotoxin on gastric emptying of a solid nutrient meal and the neural mechanisms involved in such a response were investigated in conscious rats. The intraperitoneal (i.p.) administration of E. coli endotoxin (40 microg/kg) significantly reduced the 4-h rate of gastric emptying of a standard solid nutrient meal. Ablation of primary afferent neurons by systemic administration of high doses of capsaicin (20+30+50 mg/kg s.c.) to adult rats did not modify the rate of gastric emptying in control animals but prevented the delay in gastric transit induced by endotoxin. Local application of capsaicin to the vagus nerve rather than application of capsaicin to the celiac ganglion significantly repressed endotoxin-induced delay in gastric emptying. Neither treatment modified the rate of gastric emptying in vehicle-treated animals. Blockade of CGRP receptors (CGRP 8-37, 100 microg/kg i.v.) did not alter gastric emptying in control animals but significantly prevented endotoxin-induced inhibition of gastric emptying. In contrast, a tachykinin receptor antagonist ([D-Pro2, D-Trp7.9]-substance P, 2 mg/kg i.p.) significantly reduced the rate of gastric emptying in control animals and did not modify the inhibitory effects of endotoxin. Adrenergic blockade with phentolamine (3 mg/kg i.p.) +/- propranolol (5 mg/kg i.p.) or muscarinic antagonism with atropine (0.1 mg/kg i.p.) failed to reverse the delay in gastric emptying induced by endotoxin. These observations indicate that endotoxin-induced delay in gastric emptying of a solid nutrient meal is mediated by capsaicin-sensitive afferent neurons.


Subject(s)
Afferent Pathways/drug effects , Capsaicin/pharmacology , Gastric Emptying/drug effects , Lipopolysaccharides/pharmacology , Adrenergic alpha-Antagonists/pharmacology , Animals , Atropine/pharmacology , Calcitonin Gene-Related Peptide/pharmacology , Calcitonin Gene-Related Peptide Receptor Antagonists , Drug Interactions , Gastric Emptying/physiology , Lipopolysaccharides/antagonists & inhibitors , Male , Muscarinic Antagonists/pharmacology , Neurons, Afferent/drug effects , Neurons, Afferent/physiology , Neurons, Efferent/drug effects , Neurons, Efferent/physiology , Peptide Fragments/pharmacology , Phentolamine/pharmacology , Rats , Rats, Sprague-Dawley
11.
Curr Pharm Des ; 7(1): 31-48, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11172700

ABSTRACT

Nitric oxide is a ubiquitous molecule involved in a variety of biological processes. The specific action of NO depends on its enzymatic sources namely neuronal nitric oxide synthase (nNOS), endothelial NOS (eNOS) and inducible NOS (iNOS) and all three isoforms have been localized in the gastrointestinal tract. Constitutive synthesis of NO by nNOS or eNOS isoforms is involved in the maintaining of the gastrointestinal mucosal integrity through modulation of gastric mucosal blood flow, epithelial secretion and barrier function. However, large amounts of NO synthesized from the inducible isoform have been implicated in tissue injury in the gut during inflammatory reactions. In this review we provide an overview of the dual role of nitric oxide in modulating gastrointestinal mucosal defense and injury. In addition, we highlight the therapeutic potential of NO modulation.


Subject(s)
Anti-Ulcer Agents/therapeutic use , Gastroenteritis/drug therapy , Gastroenteritis/metabolism , Gastrointestinal Agents/therapeutic use , Nitric Oxide/physiology , Peptic Ulcer/drug therapy , Peptic Ulcer/metabolism , Animals , Enzyme Inhibitors/therapeutic use , Gastroenteritis/pathology , Humans , Nitric Oxide Donors/therapeutic use , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Peptic Ulcer/pathology
12.
Br J Pharmacol ; 130(6): 1283-8, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10903967

ABSTRACT

1. This study examines the role of a central pathway involving glutamate receptors, nitric oxide (NO) and cyclic GMP in the acute inhibitory effects of low doses of peripheral endotoxin on pentagastrin-stimulated acid production. 2. Vagotomy or intracisternal (i.c.) microinjections of the NO-inhibitor, N(G)-nitro-L-arginine methyl esther (L-NAME; 200 microg rat(-1)) restored acid secretory responses in endotoxin (10 microg kg(-1), i.v.)-treated rats. 3. The acid-inhibitory effect of i.v. endotoxin (10 microg kg(-1), i.v.) was prevented by prior i.c. administration of the NMDA receptor antagonists, dizocilpine maleate (MK-801; 10 nmol rat(-1)) and D-2-amino-5-phosphono-valeric acid (AP-5; 20 nmol rat(-1)), or the AMPA/kainate antagonist 6,7-dinitroquinoxaline-2,3-dione (DNQX; 10 nmol rat(-1)). However, the competitive metabotropic glutamate receptor antagonist (+)-alpha-methyl-4-carboxyphenylglycine (MCPG; 20 - 1000 nmol rat(-1)) did not antagonize the effects of endotoxin. 4. I.c. administration of L-glutamate (0.1 nmol rat(-1)) inhibited pentagastrin-stimulated gastric acid secretion. Coadministration with L-NAME (200 microg rat(-1)) prevented the inhibition of gastric acid secretion by the aminoacid. 5. I.c. administration of 1H-[1,2, 4]Oxazodiolo[4,3-a]quinoxalin-1-one (ODQ; 100 nmol rat(-1)), a soluble guanylyl cyclase (sGC) blocker, reversed the hyposecretory effect of endotoxin. 6. I.c. administration of the cyclic GMP analogue 8-Bromoguanosine-3,5-cyclic monophosphate (8-Br-cGMP; 100 - 300 nmol rat(-1)) reduced gastric acid production in a dose-dependent manner. 7. We conclude that central NMDA and AMPA/kainate receptors are involved in the acid inhibitory effect of peripherally administered endotoxin. This central pathway involves synthesis of NO, which acts on the enzyme sGC.


Subject(s)
Endotoxins/pharmacology , Gastric Acid/metabolism , Guanylate Cyclase/physiology , Nitric Oxide/physiology , Receptors, Glutamate/physiology , Animals , Benzoates/pharmacology , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Dizocilpine Maleate/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Gastric Mucosa/metabolism , Glycine/analogs & derivatives , Glycine/pharmacology , Guanylate Cyclase/antagonists & inhibitors , Male , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/antagonists & inhibitors , Nitric Oxide Synthase/antagonists & inhibitors , Oxadiazoles/pharmacology , Pentagastrin/pharmacology , Quinoxalines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Glutamate/drug effects , Solubility , Stomach/drug effects , Vagotomy
13.
Neuroreport ; 10(15): 3217-21, 1999 Oct 19.
Article in English | MEDLINE | ID: mdl-10574563

ABSTRACT

The effects of leptin injected intracerebroventricularly (i.c.v.) or i.p. on food intake and gastric emptying of a solid nutrient meal were studied in fasted Long-Evan rats. Leptin (3 microg, i.c.v.) reduced the 5 h cumulative food intake by 39% and gastric transit by 50% while i.p. leptin (300 microg) resulted in a 35% decrease in food intake and no change in gastric transit after 5 h. Lower i.p. doses of leptin (30 or 3 microg) did not alter food intake. These results show that central, unlike peripheral, injection of leptin inhibits gastric transit of an ingested meal; such a central action of leptin may contribute to the greater potency of i.c.v. than i.p. leptin to suppress food intake.


Subject(s)
Eating/drug effects , Gastric Emptying/drug effects , Leptin/administration & dosage , Animals , Eating/physiology , Gastric Emptying/physiology , Injections, Intraventricular , Leptin/physiology , Rats , Rats, Long-Evans , Time Factors
14.
Br J Pharmacol ; 127(7): 1603-10, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10455316

ABSTRACT

1. Central administration of bombesin inhibits gastric acid production independently of the centrally or peripherally-acting stimuli employed. This study evaluates the role and location of the cerebral nitric oxide (NO) implicated in the inhibitory effect of central bombesin on in vivo rat gastric acid secretion, as induced by distension with 15 cm H2O, insulin (0.75 u.i. kg-1 i.p.) TRH (1.2 microg kg-1, i.c.) or pentagastrin (100 microg kg-1, i.p.). 2. The acid-inhibitory effect of i.c. bombesin (40 ng kg-1) was prevented by prior administration of L-NAME (80 microg kg-1) in the dorsal motor nucleus of the vagus (DMN). This dose of L-NAME when administered into the nucleus of the tractus solitarious (NTS) did not influence the effects of bombesin. Administration of L-arginine (400 microg kg-1) into the DMN restored the acid-inhibitory effect of i.c. bombesin in animals treated with L-NAME. 3. Microinjection of bombesin (12 ng kg-1) into the paraventricular nucleus of the hypothalamus (PvN) inhibits acid secretion stimulated by pentagastrin. This inhibitory effect was prevented by a previous injection of L-NAME (80 microg kg-1) into the DMN. 4. The release of NO in the DMN following i.c. administration of bombesin was confirmed by in vivo electrochemical detection. 5. Administration by microdialysis in the DMN of the NO-donor SNAP (25 mM in 1.5 microl min-1) into the DMN inhibits pentagastrin-stimulated gastric acid secretion. 6. The present study suggests that nNOS-containing neurons in the DMN have an inhibitory role in the control of gastric acid responses.


Subject(s)
Bombesin/pharmacology , Gastric Acid/metabolism , Nitric Oxide/biosynthesis , Vagus Nerve/metabolism , Animals , Bombesin/administration & dosage , Enzyme Inhibitors/pharmacology , Male , Microdialysis , Microinjections , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type I , Paraventricular Hypothalamic Nucleus , Penicillamine/analogs & derivatives , Penicillamine/pharmacology , Pentagastrin/antagonists & inhibitors , Pentagastrin/pharmacology , Rats , Rats, Sprague-Dawley , S-Nitroso-N-Acetylpenicillamine , Solitary Nucleus/cytology , Solitary Nucleus/drug effects , Thyrotropin-Releasing Hormone/pharmacology , Vagus Nerve/drug effects
15.
Naunyn Schmiedebergs Arch Pharmacol ; 360(6): 676-82, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10619185

ABSTRACT

The gastric acid hyposecretory state associated with endotoxemia is mediated by a nervous reflex involving the central nervous system. The aim of the present study was to analyse the central effects of different peptides on distension-stimulated gastric acid secretion and the endogenous role of such peptides on the hyposecretory effects of endotoxin. The effect of an intracisternal (i.c.) administration of oxytocin, vasopressin, corticotropin releasing factor (CRF), bombesin, somatostatin and the opioid receptor agonist BW443C or an intravenous (i.v.) injection of a small dose of endotoxin on distension-stimulated gastric acid secretion was studied in the continuously perfused stomach of anaesthetised rats. In some animals, specific receptor antagonists for oxytocin (Compound VI [d(CH2)5, Tyr(Me)2, Thr4, Tyr-NH2(9)]-OVT, 0.01-1 microg/rat), vasopressin (des-Gly9-[beta-Mercapto-beta,beta-cyclopentamethylene-propiony l1, O-Et-Tyr2, Val4, Arg8]-VP, 20 microg/rat), CRF (alpha-helical CRF [9-41], 50 microg/rat) or bombesin (D-Phe12-Bombesin, 20 microg/rat) were administered i.c. before endotoxin. Distension-stimulated acid secretion was significantly inhibited by central oxytocin (0.2, 2 or 4 nmol/rat, 45+/-16%, 69+/-10% and 79+/-5% reduction, respectively), CRF (0.5, 1 or 2 nmol/rat, 52.2+/-15.6%, 74.3+/-9.1% and 93.2+/-1.6% reduction, respectively) and bombesin (2 nmol/rat, 79.1+/-5.8% reduction). The hyposecretory effect induced by endotoxin (5 microg/kg, 60.2+/-2.3% reduction) was reversed in a dose-dependent manner by pretreatment with the oxytocin receptor antagonist (0.01, 0.1 and 1 microg/rat, 65.2+/-14.4%, 88.0+/-22.5% and 112.4+/-25.2% of control response, respectively) while the vasopressin (20 microg/rat), CRF (50 microg/rat) or bombesin (20 microg/rat) receptor antagonists had no effect. The present results support a role for the endogenous release and action in the central nervous system of oxytocin in the inhibitory effect of endotoxin on gastric acid secretion.


Subject(s)
Endotoxemia/metabolism , Gastric Acid/metabolism , Gastric Dilatation/metabolism , Gastric Mucosa/metabolism , Oxytocin/metabolism , Adrenergic Agents/pharmacology , Animals , Antidiuretic Hormone Receptor Antagonists , Bombesin/administration & dosage , Corticotropin-Releasing Hormone/administration & dosage , Dose-Response Relationship, Drug , Endotoxemia/chemically induced , Endotoxins , Female , Gastric Dilatation/chemically induced , Gastric Mucosa/drug effects , Gastric Mucosa/physiology , Hormones/administration & dosage , Injections, Intraventricular , Male , Narcotic Antagonists/administration & dosage , Oligopeptides/administration & dosage , Oxytocin/administration & dosage , Rats , Rats, Wistar , Receptors, Bombesin/antagonists & inhibitors , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Receptors, Oxytocin/antagonists & inhibitors , Somatostatin/administration & dosage , Vasoconstrictor Agents/administration & dosage , Vasopressins/administration & dosage
16.
J Pharm Pharmacol ; 50(10): 1095-100, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9821654

ABSTRACT

Gastric mucosal damage induced by haemorrhagic shock in the anaesthetized rat has been evaluated by studying changes in capillary-to-lumen clearance of fluorescein isothiocyanate (FITC)-labelled dextran. Haemorrhagic shock (20 min ischaemia + 20 min reperfusion) induced a significant increase in blood-to-lumen permeability to FITC-dextran of different molecular weight (10,000, 40,000 and 70,000) without modifying the macroscopic integrity of the gastric mucosa. The increase in vascular permeability was dependent on the time of administration of the tracer and was correlated with an elevation of the protein content of the gastric lumen. Intravenous administration of the secretagogue pentagastrin (20 or 50 micrograms kg-1 h-1) did not significantly modify the vascular permeability to dextran in control animals or in animals subjected to haemorrhagic shock. When the intraluminal pH was reduced by intragastric administration of acidic saline solution, only pH 1, which itself induced the appearance of macroscopic mucosal lesions, significantly increased vascular permeability to dextran, both in control animals and in animals subjected to haemorrhagic shock. These findings suggest that stress induced by haemorrhagic shock increases vascular gastric permeability to dextran, by an acid-independent mechanism, without affecting the macroscopic integrity of the gastric mucosa.


Subject(s)
Capillary Permeability , Dextrans/pharmacokinetics , Fluorescein-5-isothiocyanate/analogs & derivatives , Gastric Mucosa/metabolism , Shock, Hemorrhagic/metabolism , Animals , Fluorescein-5-isothiocyanate/pharmacokinetics , Hydrogen-Ion Concentration , Rats , Rats, Wistar , Shock, Hemorrhagic/pathology
17.
Brain Res ; 791(1-2): 157-66, 1998 Apr 27.
Article in English | MEDLINE | ID: mdl-9593872

ABSTRACT

We previously reported a synergistic interaction between leptin and cholecystokinin (CCK) to reduce food intake through CCK-A receptors in lean mice fasted for 24 h. To identify the activated neuronal pathways, we investigated changes in Fos expression in brain nuclei 2 h after single or combined intraperitoneal (i.p.) injections of leptin (120 microg/kg) and sulfated CCK-8 (3.5 microg/kg) in male lean mice (C57BL/6) fasted for 24 h using immunohistochemistry for Fos, the protein product of the early gene, c-fos. Leptin did not increase Fos expression in the brain compared with vehicle-treated mice. CCK increased the numbers of Fos-positive neurons in the nucleus of the solitary tract (NTS)/area postrema (AP), central nucleus of the amygdala (CeA) and, to a smaller extent, in the paraventricular nucleus of the hypothalamus (PVN) (5.2-, 2.3- and 0. 3-fold respectively). Injections of leptin-CCK further enhanced Fos expression by 40% in the PVN compared with that induced by CCK alone, but not in the other nuclei. Devazepide (a CCK-A receptor antagonist, 1 mg/kg, i.p.) prevented the increase in Fos expression induced by leptin-CCK in the PVN and by CCK alone in the PVN, CeA and NTS/AP. These results indicate that in fasted mice, i.p. injection of CCK increases Fos expression in specific brain nuclei through CCK-A receptors while leptin alone had no effect. Leptin in conjunction with CCK selectively enhanced Fos expression in the PVN. The PVN may be an important site mediating the synergistic effect of leptin-CCK to regulate food intake.


Subject(s)
Adipose Tissue , Brain/drug effects , Cholecystokinin/pharmacology , Nerve Tissue Proteins/biosynthesis , Proteins/pharmacology , Proto-Oncogene Proteins c-fos/biosynthesis , Animals , Arcuate Nucleus of Hypothalamus/drug effects , Brain/metabolism , Fasting/metabolism , Immunohistochemistry , Injections, Intraperitoneal , Leptin , Limbic System/drug effects , Male , Mice , Mice, Inbred C57BL , Recombinant Proteins/pharmacology , Rhombencephalon/drug effects
18.
J Pharm Pharmacol ; 49(10): 988-90, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9364408

ABSTRACT

Because of the evidence that endogenous nitric oxide (NO) plays an essential role in the physiological regulation of gastrointestinal motility we have investigated, by use of the NO synthase inhibitor, NG-nitro-L-arginine methyl ester (L-NAME), the role of endogenous NO in the acute endotoxin-induced changes of gastrointestinal transit. Pre-treatment with E. coli endotoxin (100 micrograms kg-1, i.v.) induced a significant increase in the gastrointestinal transit of a charcoal suspension in anaesthetized rats. Previous administration of the NO synthase inhibitor, L-NAME (10 mg kg-1, i.v.) significantly prevented the effects of endotoxin. L-arginine (200 mg kg-1, i.v.) and the substance P antagonist [D-Pro2, D-Trp7,9]-substance P (SPA), significantly reversed the effects of L-NAME on gastrointestinal transit in rats treated with endotoxin. Pre-treatment with dexamethasone (5 mg kg-1, s.c., twice), an inhibitor of the expression of inducible NO synthase, did not affect the increase in the gastrointestinal transit through constitutive NO synthesis. The results suggest that constitutive nitric oxide is involved in the increase of gastrointestinal transit induced by endotoxin and that the reduction in transit induced by L-NAME in endotoxin-treated rats is mediated by endogenous tachykinins.


Subject(s)
Endotoxins/pharmacology , Gastrointestinal Transit/drug effects , Nitric Oxide/physiology , Tachykinins/physiology , Animals , Anti-Inflammatory Agents/pharmacology , Dexamethasone/pharmacology , Enzyme Inhibitors/pharmacology , Male , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Rats , Rats, Wistar , Substance P/pharmacology
19.
Proc Natl Acad Sci U S A ; 94(19): 10455-60, 1997 Sep 16.
Article in English | MEDLINE | ID: mdl-9294232

ABSTRACT

Leptin is a circulating protein involved in the long-term regulation of food intake and body weight. Cholecystokinin (CCK) is released postprandially and elicits satiety signals. We investigated the interaction between leptin and CCK-8 in the short-term regulation of food intake induced by 24-hr fasting in lean mice. Leptin, injected intraperitoneally (i.p.) at low doses (4-120 microg/kg), which did not influence feeding behavior for the first 3 hr postinjection, decreased food intake dose dependently by 47-83% during the first hour when coinjected with a subthreshold dose of CCK. Such an interaction was not observed between leptin and bombesin. The food-reducing effect of leptin injected with CCK was not associated with alterations in gastric emptying or locomotor behavior. Leptin-CCK action was blocked by systemic capsaicin at a dose inducing functional ablation of sensory afferent fibers and by devazepide, a CCK-A receptor antagonist but not by the CCK-B receptor antagonist, L-365,260. The decrease in food intake which occurs 5 hr after i.p. injection of leptin alone was also blunted by devazepide. Coinjection of leptin and CCK enhanced the number of Fos-positive cells in the hypothalamic paraventricular nucleus by 60%, whereas leptin or CCK alone did not modify Fos expression. These results indicate the existence of a functional synergistic interaction between leptin and CCK leading to early suppression of food intake which involves CCK-A receptors and capsaicin-sensitive afferent fibers.


Subject(s)
Cholecystokinin/pharmacology , Feeding Behavior/drug effects , Proteins/pharmacology , Animals , Benzodiazepinones/pharmacology , Bombesin/pharmacology , Capsaicin/pharmacology , Devazepide , Drug Synergism , Gastric Emptying/drug effects , Immunohistochemistry , Leptin , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Stereotyped Behavior/drug effects
20.
Eur J Pharmacol ; 326(2-3): 211-22, 1997 May 20.
Article in English | MEDLINE | ID: mdl-9196274

ABSTRACT

We investigated the effect of Mn2+ on the mechanical responses evoked by high K+ (60 mM) or low Na+ (25 mM) solutions, oxytocin and neurokinin A in the oestrogen-primed rat uterus. In a Ca2+-free, Mn2+ (0.54 mM)-containing solution, high K+ or low Na+ solutions produced contractions of smaller amplitude than those observed in a normal Ca2+ (0.54 mM) solution, which were abolished by nifedipine (1 microM). Oxytocin (1 microM) and neurokinin A (1 microM, in the presence of phosphoramidon 1 microM) evoked nifedipine-insensitive contractile responses similar to (oxytocin) or smaller (neurokinin A) in amplitude than those observed in Ca2+ (0.54 mM)-containing solution. In strips loaded with Ca2+ (2.16 mM) for 10 min and then exposed to a Ca2+- and Mn2+-free, EGTA (3 mM)-containing medium for 4 min, both oxytocin and neurokinin A induced transient contraction followed by a small sustained response. The transient component of the response was abolished by cyclopiazonic acid (10 microM). When preparations were loaded with Mn2+ (2.16 mM) for 10 min, only the small, tonic contraction was observed. In Ca2+-containing solution, Mn2+ (0.01-10 mM) inhibited in a concentration-dependent manner the rhythmic contractions developed either spontaneously or by electrical stimulation as well as high K+- and neurokinin A-induced contractions. Mn2+ also abolished the rhythmic, but not the tonic component of the response to oxytocin, and the preparation remained maximally contracted. These data suggest that in the oestrogen-primed rat uterus, Mn2+ acts as an antagonist of Ca2+ influx through L-type voltage-operated Ca2+ channels. In addition, Mn2+ enters the cell mainly through nifedipine-insensitive receptor-operated channels and, to a lesser degree, through L-type Ca2+ channels to produce contraction by directly activating the contractile machinery.


Subject(s)
Manganese/pharmacology , Neurokinin A/pharmacology , Oxytocin/pharmacology , Potassium/pharmacology , Sodium/pharmacology , Uterine Contraction/drug effects , Animals , Calcium/pharmacology , Drug Evaluation, Preclinical , Electric Stimulation , Estrogens/pharmacology , Female , In Vitro Techniques , Inositol Phosphates/biosynthesis , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL