Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 423
Filter
1.
Am J Physiol Gastrointest Liver Physiol ; 318(4): G613-G623, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32068440

ABSTRACT

Esophageal injury from acid exposure related to gastroesophageal reflux disease is a common problem and a risk factor for development of Barrett's esophagus and esophageal adenocarcinoma. Our previous work highlights the benefits of using porcine esophagus to study human esophageal disease because of the similarities between porcine and human esophagus. In particular, esophageal submucosal glands (ESMGs) are present in human esophagus and proximal porcine esophagus but not in rodent esophagus. Although CFTR is expressed in the ducts of ESMGs, very little is known about CFTR and alternate anion channels, including ClC-2, in the setting of acid-related esophageal injury. After finding evidence of CFTR and ClC-2 in the basal layers of the squamous epithelium, and in the ducts of the ESMGs, we developed an ex vivo porcine model of esophageal acid injury. In this model, esophageal tissue was placed in Ussing chambers to determine the effect of pretreatment with the ClC-2 agonist lubiprostone on tissue damage related to acid exposure. Pretreatment with lubiprostone significantly reduced the level of acid injury and significantly augmented the recovery of the injured tissue (P < 0.05). Evaluation of the interepithelial tight junctions showed well-defined membrane localization of occludin in lubiprostone-treated injured tissues. Pretreatment of tissues with the Na+-K+-2Cl- cotransporter inhibitor bumetanide blocked lubiprostone-induced increases in short-circuit current and inhibited the reparative effect of lubiprostone. Furthermore, inhibition of ClC-2 with ZnCl2 blocked the effects of lubiprostone. We conclude that ClC-2 contributes to esophageal protection from acid exposure, potentially offering a new therapeutic target.NEW & NOTEWORTHY This research is the first to describe the presence of anion channels ClC-2 and CFTR localized to the basal epithelia of porcine esophageal mucosa and the esophageal submucosal glands. In the setting of ex vivo acid exposure, the ClC-2 agonist lubiprostone reduced acid-related injury and enhanced recovery of the epithelial barrier. This work may ultimately provide an alternate mechanism for treating gastroesophageal reflux disease.


Subject(s)
Esophageal Mucosa/drug effects , Lubiprostone/pharmacology , 16,16-Dimethylprostaglandin E2/pharmacology , Animals , Bumetanide/pharmacology , Chloride Channel Agonists/pharmacology , Chloride Channels/genetics , Chloride Channels/metabolism , Chlorides/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Dose-Response Relationship, Drug , Female , Gene Expression Regulation/drug effects , Hydrochloric Acid/pharmacology , Male , Occludin/metabolism , Swine , Time Factors , Zinc Compounds/pharmacology
2.
Am J Physiol Gastrointest Liver Physiol ; 317(6): G862-G871, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31545918

ABSTRACT

Nonsteroidal anti-inflammatory drugs (NSAIDs) such as diclofenac (DFN) and indomethacin (INDO) are extensively used worldwide. Their main side effects are injury of the gastrointestinal tract, including erosions, ulcers, and bleeding. Since gastric epithelial cells (GEPCs) are crucial for mucosal defense and are the major target of injury, we examined the extent to which DFN- and INDO-induced GEPC injury can be reversed by nerve growth factor (NGF), 16,16 dimethyl prostaglandin E2 (dmPGE2), and 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR), the pharmacological activator of the metabolic sensor AMP kinase (AMPK). Cultured normal rat gastric mucosal epithelial (RGM1) cells were treated with PBS (control), NGF, dmPGE2, AICAR, and/or NSAID (DFN or INDO) for 1-4 h. We examined cell injury by confocal microscopy, cell death/survival using calcein AM, mitochondrial membrane potential using MitoTracker, and phosphorylation of AMPK by Western blotting. DFN and INDO treatment of RGM1 cells for 2 h decreased mitochondrial membrane potential and cell viability. NGF posttreatment (initiated 1 or 2 h after DFN or INDO) reversed the dissipation of mitochondrial membrane potential and cell injury caused by DFN and INDO and increased cell viability versus cells treated for 4 h with NSAID alone. Pretreatment with dmPGE2 and AICAR significantly protected these cells from DFN- and INDO-induced injury, whereas dmPGE2 and AICAR posttreatment (initiated 1 h after NSAID treatment) reversed cell injury and significantly increased cell viability and rescued the cells from NSAID-induced mitochondrial membrane potential reduction. DFN and INDO induce extensive mitochondrial injury and GEPC death, which can be significantly reversed by NGF, dmPGE2, and AICAR.NEW & NOTEWORTHY This study demonstrated that mitochondria are key targets of diclofenac- and indomethacin-induced injury of gastric epithelial cells and that diclofenac and indomethacin injury can be prevented and, importantly, also reversed by treatment with nerve growth factor, 16,16 dimethyl prostaglandin E2, and 5-aminoimidazole-4-carboxamide ribonucleotide.


Subject(s)
16,16-Dimethylprostaglandin E2/pharmacology , Aminoimidazole Carboxamide/analogs & derivatives , Diclofenac/adverse effects , Gastric Mucosa , Indomethacin/adverse effects , Mitochondria , Nerve Growth Factor/pharmacology , Ribonucleosides/pharmacology , Aminoimidazole Carboxamide/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Ulcer Agents/pharmacology , Cell Death/drug effects , Cell Survival/drug effects , Cells, Cultured , Gastric Mucosa/drug effects , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Rats
3.
Pulm Pharmacol Ther ; 48: 62-70, 2018 02.
Article in English | MEDLINE | ID: mdl-28951192

ABSTRACT

A feature of cough variant asthma is a heightened cough response to bronchoconstriction. The mediators of this response are unknown. This study was designed to elucidate the role of lipid mediators in bronchoconstriction-triggered cough response in an experimental animal model. We examined the influence of bronchoconstriction on cell components and mediators including prostaglandin E2 (PGE2) in bronchoalveolar lavage fluid (BALF). We studied the cough response to bronchoconstriction (CRB) by measuring the correlation between the increase in enhanced pause (Penh), an index of bronchoconstriction, and cough counts induced by methacholine (Mch) inhalation in conscious guinea pigs. We then examined the effects of intraperitoneal pretreatment with 16, 16-dimethyl-prostaglandin E2 (dm-PGE2) on CRB and cough counts. The total number of cells and cell components in the BALF were not influenced by bronchoconstriction. While levels of PGE2, prostaglandin I2, and cysteinyl leukotrienes were significantly increased, levels of prostaglandin D2, thromboxane B2, and substance P in the BALF were not. Dm-PGE2 significantly decreased the Mch-induced increase in Penh. Following bronchoconstriction by additional Mch inhalation, dm-PGE2 produced an increase in CRB and cough counts in a dose-dependent manner. Additionally, the heightened CRB following dm-PGE2 treatment was suppressed by pretreatment with PGE2 receptor (E-prostanoid EP) -1 and EP-3 antagonists in a dose-dependent manner, but not by EP-2 and EP-4 antagonists. The EP-1 antagonist also decreased cough counts. These results suggest that PGE2 acts as an exacerbating factor for bronchoconstriction-triggered cough. EP1 and EP3 may provide new therapeutic targets for cough variant asthma.


Subject(s)
16,16-Dimethylprostaglandin E2/pharmacology , Bronchoconstriction , Cough/physiopathology , Dinoprostone/metabolism , 16,16-Dimethylprostaglandin E2/administration & dosage , Animals , Bronchoalveolar Lavage Fluid , Cysteine/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Epoprostenol/metabolism , Guinea Pigs , Leukotrienes/metabolism , Male , Methacholine Chloride/administration & dosage , Receptors, Prostaglandin E/drug effects , Receptors, Prostaglandin E/metabolism
4.
Exp Hematol ; 56: 64-68, 2017 12.
Article in English | MEDLINE | ID: mdl-28822771

ABSTRACT

Monocytes/macrophages differentiating from bone marrow (BM) cells pulsed for 2 hours at 37°C with a stabilized derivative of prostaglandin E2, 16,16-dimethyl PGE2 (dmPGE2), migrated less efficiently toward a chemoattractant than monocytes/macrophages differentiated from BM cells pulsed with vehicle. To confirm that the effect on BM cells was long lasting and to replicate human BM transplantation, chimeric mice were established with donor BM cells pulsed for 2 hours with dmPGE2 before injection into marrow-ablated congenic recipient mice. After 12 weeks, when high levels (90%) of engraftment were obtained, regenerated BM-derived monocytes/macrophages differentiating in vitro or in vivo migrated inefficiently toward the chemokines colony-stimulating factor-1 (CSF-1) and chemokine (C-C motif) ligand 2 (CCL2) or thioglycollate, respectively. Our results reveal long-lasting changes to progenitor cells of monocytes/macrophages by a 2-hour dmPGE2 pulse that, in turn, limits the migration of their daughter cells to chemoattractants and inflammatory mediators.


Subject(s)
Bone Marrow Cells/metabolism , Cell Movement/drug effects , Dinoprostone/pharmacology , Macrophages/metabolism , Monocytes/metabolism , 16,16-Dimethylprostaglandin E2/pharmacology , Animals , Bone Marrow Cells/cytology , Chemokine CCL2/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/cytology , Mice , Monocytes/cytology
5.
Int J Biochem Cell Biol ; 59: 52-61, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25462156

ABSTRACT

Prostaglandin E2 (PGE2) and hypoxia-inducible factor-1α (HIF-1α) affect many mechanisms that have been shown to play a role in prostate cancer. In PGE2-treated LNCaP cells, up-regulation of HIF-1α requires the internalization of PGE2, which is in sharp contrast with the generally accepted view that PGE2 acts through EP receptors located at the cell membrane. Here we aimed to study in androgen-independent PC3 cells the role of intracellular PGE2 in several events linked to prostate cancer progression. To this end, we used bromocresol green, an inhibitor of prostaglandin uptake that blocked the immediate rise in intracellular immunoreactive PGE2 following treatment with 16,16-dimethyl-PGE2. Bromocresol green prevented the stimulatory effect of 16,16-dimethyl-PGE on cell proliferation, adhesion, migration and invasion and on HIF-1α expression and activity, the latter assessed as the HIF-dependent activation of (i) a hypoxia response element-luciferase plasmid construct, (ii) production of angiogenic factor vascular endothelial growth factor-A and (iii) in vitro angiogenesis. The basal phenotype of PC3 cells was also affected by bromocresol green, that substantially lowered expression of HIF-1α, production of vascular endothelial growth factor-A and cell proliferation. These results, and the fact that we found functional intracellular EP receptors in PC3 cells, suggest that PGE2-dependent intracrine mechanisms play a role in prostate cancer Therefore, inhibition of the prostaglandin uptake transporter might be a novel therapeutic approach for the treatment of prostate cancer.


Subject(s)
Dinoprostone/metabolism , Intracellular Space/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , 16,16-Dimethylprostaglandin E2/pharmacology , Biological Transport/drug effects , Bromcresol Green/pharmacology , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Collagen/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Neoplasm Invasiveness , Phenotype , Receptors, Prostaglandin E/metabolism
6.
Cell Stem Cell ; 14(2): 146-8, 2014 Feb 06.
Article in English | MEDLINE | ID: mdl-24506882

ABSTRACT

Translating basic research findings into therapeutic settings presents many scientific, logistic, and financial challenges for academic researchers. Here, I highlight some key insights for navigating such challenges based on recent clinical trials initiated by basic research from my lab.


Subject(s)
Clinical Trials as Topic , Translational Research, Biomedical , 16,16-Dimethylprostaglandin E2/pharmacology , Animals , Humans , Mice , Stem Cell Transplantation , United States , United States Food and Drug Administration , Zebrafish
7.
Acta Biochim Biophys Sin (Shanghai) ; 46(1): 40-7, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24296051

ABSTRACT

The effect of prostaglandin E2 (PGE2) on bone mass has been well-established in vivo. Previous studies have showed that PGE2 increases differentiation, proliferation, and regulates cell morphology through F-actin stress fiber in statically cultured osteoblasts. However, the effect of PGE2 on osteoblasts in the presence of fluid shear stress (FSS), which could better uncover the anabolic effect of PGE2 in vivo, has yet to be examined. Here, we hypothesized that PGE2 modulates F-actin stress fiber in FSS-stimulated MC3T3-E1 osteoblastic cells through protein kinase A (PKA) pathway. Furthermore, this PGE2-induced F-actin remodeling was associated with the recovery of cellular mechanosensitivity. Our data showed that treatment with 10 nM dmPGE2 for 15 min significantly suppressed the F-actin stress fiber intensity in FSS-stimulated cells in a PKA-dependent manner. In addition, dmPGE2 treatment enhanced the cells' calcium peak magnitude and the percentage of responding cells in the second FSS stimulation, though these effects were abolished and attenuated by co-treatment with phalloidin. Our results demonstrated that 10 nM dmPGE2 was able to accelerate the 'reset' process of F-actin stress fiber to its pre-stimulated level partially through PKA pathway, and thus promoted the recovery of cellular mechanosensitivity. Our finding provided a novel cellular mechanism by which PGE2 increased bone formation as shown in vivo, suggesting that PGE2 could be a potential target for treatments of bone formation-related diseases.


Subject(s)
16,16-Dimethylprostaglandin E2/pharmacology , Actins/metabolism , Cyclic AMP-Dependent Protein Kinases/physiology , Osteoblasts/metabolism , Stress Fibers/metabolism , Cells, Cultured , Models, Biological , Osteoblasts/drug effects , Osteogenesis/drug effects , Stress, Mechanical
8.
Blood ; 122(17): 3074-81, 2013 Oct 24.
Article in English | MEDLINE | ID: mdl-23996087

ABSTRACT

Umbilical cord blood (UCB) is a valuable source of hematopoietic stem cells (HSCs) for use in allogeneic transplantation. Key advantages of UCB are rapid availability and less stringent requirements for HLA matching. However, UCB contains an inherently limited HSC count, which is associated with delayed time to engraftment, high graft failure rates, and early mortality. 16,16-Dimethyl prostaglandin E2 (dmPGE2) was previously identified to be a critical regulator of HSC homeostasis, and we hypothesized that brief ex vivo modulation with dmPGE2 could improve patient outcomes by increasing the "effective dose" of HSCs. Molecular profiling approaches were used to determine the optimal ex vivo modulation conditions (temperature, time, concentration, and media) for use in the clinical setting. A phase 1 trial was performed to evaluate the safety and therapeutic potential of ex vivo modulation of a single UCB unit using dmPGE2 before reduced-intensity, double UCB transplantation. Results from this study demonstrated clear safety with durable, multilineage engraftment of dmPGE2-treated UCB units. We observed encouraging trends in efficacy, with accelerated neutrophil recovery (17.5 vs 21 days, P = .045), coupled with preferential, long-term engraftment of the dmPGE2-treated UCB unit in 10 of 12 treated participants.


Subject(s)
16,16-Dimethylprostaglandin E2/pharmacology , Blood Platelets/drug effects , Cord Blood Stem Cell Transplantation/methods , Fetal Blood/drug effects , Graft Survival/immunology , Hematologic Neoplasms/therapy , Adult , Aged , Blood Platelets/cytology , Blood Platelets/immunology , Cells, Cultured , Cryopreservation , Female , Fetal Blood/cytology , Fetal Blood/immunology , Fetal Blood/transplantation , Gene Expression Profiling , Hematologic Neoplasms/immunology , Hematologic Neoplasms/pathology , Humans , Male , Middle Aged , Transplantation Chimera , Transplantation, Homologous , Treatment Outcome
9.
PLoS One ; 7(1): e29331, 2012.
Article in English | MEDLINE | ID: mdl-22276108

ABSTRACT

BACKGROUND: Prostaglandin E(2) (PGE(2)) is an important mediator in tumor-promoting inflammation. High expression of cyclooxygenase-2 (COX-2) has been detected in the embryonic childhood tumor neuroblastoma, and treatment with COX inhibitors significantly reduces tumor growth. Here, we have investigated the significance of a high COX-2 expression in neuroblastoma by analysis of PGE(2) production, the expression pattern and localization of PGE(2) receptors and intracellular signal transduction pathways activated by PGE(2). PRINCIPAL FINDINGS: A high expression of the PGE(2) receptors, EP1, EP2, EP3 and EP4 in primary neuroblastomas, independent of biological and clinical characteristics, was detected using immunohistochemistry. In addition, mRNA and protein corresponding to each of the receptors were detected in neuroblastoma cell lines. Immunofluorescent staining revealed localization of the receptors to the cellular membrane, in the cytoplasm, and in the nuclear compartment. Neuroblastoma cells produced PGE(2) and stimulation of serum-starved neuroblastoma cells with PGE(2) increased the intracellular concentration of calcium and cyclic AMP with subsequent phosphorylation of Akt. Addition of 16,16-dimethyl PGE(2) (dmPGE(2)) increased cell viability in a time, dose- and cell line-dependent manner. Treatment of neuroblastoma cells with a COX-2 inhibitor resulted in a diminished cell growth and viability that was reversed by the addition of dmPGE(2). Similarly, PGE(2) receptor antagonists caused a decrease in neuroblastoma cell viability in a dose-dependent manner. CONCLUSIONS: These findings demonstrate that PGE(2) acts as an autocrine and/or paracrine survival factor for neuroblastoma cells. Hence, specific targeting of PGE(2) signaling provides a novel strategy for the treatment of childhood neuroblastoma through the inhibition of important mediators of tumor-promoting inflammation.


Subject(s)
16,16-Dimethylprostaglandin E2/pharmacology , Autocrine Communication/drug effects , Dinoprostone/metabolism , Neuroblastoma/metabolism , Biphenyl Compounds/pharmacology , Blotting, Western , Calcium/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclic AMP/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Humans , Immunohistochemistry , In Vitro Techniques , Phosphorylation/drug effects , Receptors, Prostaglandin E, EP1 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP1 Subtype/metabolism , Receptors, Prostaglandin E, EP2 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Receptors, Prostaglandin E, EP3 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP3 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tandem Mass Spectrometry , Thiophenes/pharmacology , Triazoles/pharmacology
10.
Br J Pharmacol ; 162(1): 193-209, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20804494

ABSTRACT

BACKGROUND AND PURPOSE: Changes in airway smooth muscle (ASM) phenotype may contribute to the pathogenesis of airway disease. Platelet-derived growth factor (PDGF) switches ASM from a contractile to a proliferative, hypo-contractile phenotype, a process requiring activation of extracellular signal-regulated kinase (ERK) and p70(S6) Kinase (p70(S6K) ). The effects of cAMP-elevating agents on these processes is unknown. Here, we investigated the effects of cAMP elevation by prostaglandin E(2) (PGE(2) ) and the activation of the cAMP effectors, protein kinase A (PKA) and exchange protein activated by cAMP (Epac) on PDGF-induced phenotype switching in bovine tracheal smooth muscle (BTSM). EXPERIMENTAL APPROACH: Effects of long-term treatment with the PGE(2) analogue 16,16-dimethyl-PGE(2) , the selective Epac activator, 8-pCPT-2'-O-Me-cAMP and the selective PKA activator, 6-Bnz-cAMP were assessed on the induction of a hypo-contractile, proliferative BTSM phenotype and on activation of ERK and p70(S6K) , both induced by PDGF. KEY RESULTS: Treatment with 16,16-dimethyl-PGE(2) inhibited PDGF-induced proliferation of BTSM cells and maintained BTSM strip contractility and contractile protein expression in the presence of PDGF. Activation of both Epac and PKA similarly prevented PDGF-induced phenotype switching and PDGF-induced activation of ERK. Interestingly, only PKA activation resulted in inhibition of PDGF-induced phosphorylation of p70(S6K) . CONCLUSIONS AND IMPLICATIONS: Our data indicate for the first time that both Epac and PKA regulated switching of ASM phenotype via differential inhibition of ERK and p70(S6K) pathways. These findings suggest that cAMP elevation may be beneficial in the treatment of long-term changes in airway disease.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/pharmacology , Muscle, Smooth/drug effects , Trachea/drug effects , 16,16-Dimethylprostaglandin E2/pharmacology , Animals , Blotting, Western , Cattle , Cell Line , Enzyme Activation , In Vitro Techniques , Muscle, Smooth/enzymology , Muscle, Smooth/physiology , Phosphorylation , Platelet-Derived Growth Factor/pharmacology , Ribosomal Protein S6 Kinases/metabolism , Trachea/enzymology , Trachea/physiology
11.
J Neurochem ; 115(2): 363-72, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20666934

ABSTRACT

Prostaglandin E2 (PGE2) is a well known pain and pro-inflammatory mediator abundantly produced in inflamed tissue. It causes pain by directly exciting nociceptive primary sensory neurons (nociceptors) and indirectly stimulating the release of pain-related peptide substance P (SP) and calcitonin gene-related peptide (CGRP). In an ex vivo culture of sensory ganglion explants, we tested the hypothesis that PGE2 could induce the synthesis of SP and CGRP in nociceptors. A stabilized PGE2 analog, 16,16-dimethyl PGE2, in a concentration- and time-dependent manner, significantly increased mRNA and peptide levels of SP and CGRP. The agonists of EP1 and EP4 receptors also significantly increased SP and CGRP levels. Moreover, 16,16-dimethyl PGE2-induced SP and CGRP were blocked by EP1 and EP4 antagonists as well as the inhibitors of both protein kinase A and protein kinase C. Nerve growth factor was partially involved in PGE2-induced SP and CGRP synthesis. Taken together, these results indicate that PGE2 contributes to the synthesis of SP and CGRP in nociceptors, an event mediated by EP1 and EP4 receptors, nerve growth factor and protein kinase A and protein kinase C signalling pathways. We thus conclude that facilitating the synthesis of pain-related peptides in nociceptors is a novel mechanism underlying the role of PGE2 in nociception and chronic pain states.


Subject(s)
16,16-Dimethylprostaglandin E2/pharmacology , Anti-Ulcer Agents/pharmacology , Calcitonin Gene-Related Peptide/metabolism , Ganglia, Spinal/drug effects , Substance P/metabolism , Animals , Calcitonin Gene-Related Peptide/genetics , Dibenz(b,f)(1,4)oxazepine-10(11H)-carboxylic acid, 8-chloro-, 2-acetylhydrazide/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay/methods , Gene Expression Regulation/drug effects , Male , Nerve Growth Factor/metabolism , Organ Culture Techniques , Prostaglandin Antagonists/pharmacology , Prostaglandins, Synthetic/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Prostaglandin E/agonists , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E/metabolism , Signal Transduction/drug effects , Substance P/genetics , Time Factors
12.
J Gastroenterol Hepatol ; 25 Suppl 1: S111-8, 2010 May.
Article in English | MEDLINE | ID: mdl-20586851

ABSTRACT

BACKGROUND AND AIMS: We examined the effect of lafutidine, a histamine H(2) receptor antagonist with a mucosal protective action mediated by capsaicin-sensitive sensory neurons (CSN), on intestinal lesions produced by loxoprofen administration in rats. METHODS: Animals were given loxoprofen (10-100 mg/kg p.o.) and killed 24 h later. Lafutidine (10 and 30 mg/kg), cimetidine (100 mg/kg) or famotidine (30 mg/kg) was given twice p.o. at 0.5 h before and 6 h after loxoprofen. Omeprazole (100 mg/kg) was given p.o. once 0.5 h before. Ampicillin (800 mg/kg) was given p.o. twice at 24 h and 0.5 h before loxoprofen, while 16,16-dimethyl prostaglandin E(2) (dmPGE(2); 0.01 mg/kg) was given i.v. twice at 5 min before and 6 h after. RESULTS: Loxoprofen dose-dependently produced hemorrhagic lesions in the small intestine, accompanied by invasion of enterobacteria and increased inducible nitric oxide synthase (iNOS) expression as well as myeloperoxidase activity in the mucosa. The ulcerogenic response to loxoprofen (60 mg/kg) was significantly prevented by lafutidine (30 mg/kg), similar to dmPGE(2) and ampicillin, and the effect of lafutidine was totally attenuated by ablation of CSN. Neither cimetidine, famotidine nor omeprazole had a significant effect against these lesions. Lafutidine alone increased mucus secretion and reverted the decreased mucus response to loxoprofen, resulting in suppression of bacterial invasion and iNOS expression. In addition, loxoprofen downregulated Muc2 expression, and this response was totally reversed by lafutidine mediated by CSN. CONCLUSION: Lafutidine protects the small intestine against loxoprofen-induced lesions, essentially mediated by the CSN, and this effect may be functionally associated with increased Muc2 expression/mucus secretion, an important factor in the suppression of bacterial invasion.


Subject(s)
Acetamides/pharmacology , Anti-Ulcer Agents/pharmacology , Histamine H2 Antagonists/pharmacology , Intestinal Mucosa/drug effects , Intestine, Small/drug effects , Peptic Ulcer/prevention & control , Phenylpropionates , Piperidines/pharmacology , Pyridines/pharmacology , 16,16-Dimethylprostaglandin E2/pharmacology , Ampicillin/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Capsaicin/pharmacology , Cimetidine/pharmacology , Disease Models, Animal , Enterobacteriaceae/pathogenicity , Famotidine/pharmacology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Intestine, Small/metabolism , Intestine, Small/microbiology , Intestine, Small/pathology , Male , Mucin-2/genetics , Nitric Oxide Synthase Type II/genetics , Omeprazole/pharmacology , Peptic Ulcer/chemically induced , Peptic Ulcer/metabolism , Peptic Ulcer/microbiology , Peptic Ulcer/pathology , Peroxidase/metabolism , Proton Pump Inhibitors/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells/drug effects
13.
J Gastroenterol Hepatol ; 25 Suppl 1: S15-22, 2010 May.
Article in English | MEDLINE | ID: mdl-20586859

ABSTRACT

BACKGROUND AND AIMS: Enteroscopic observation has clearly demonstrated that non-steroidal anti-inflammatory drugs/low-dose aspirin (usually enteric-coated) induces hemorrhagic lesions, including ulcers and bleeding, in the small intestine of patients at a high incidence. Such intestinal lesions induced by NSAIDs have been confirmed in animal experiments. With aspirin, however, it has long been believed that it is difficult to induce any damage in the intestinal mucosa of laboratory animals. Therefore, we established a new method of inducing intestinal hemorrhagic lesions in rats by injecting aspirin into the proximal duodenum. METHODS: Under ether anesthesia, aspirin (50-200 mg/body), suspended in 2% methylcellulose (with or without 0.1 N HCl), was injected into the proximal duodenum of normally fed or 20-h non-fed rats (male Sprague-Dawley, 9 weeks old). At 1 h after treatment, the animals were killed with ether and the entire small intestine was removed for histological examination. In some experiments, 1% Evans blue was injected (i.v.) into the rats 1 h after aspirin treatment to visualize the lesions. An image analyzer determined the total area of the intestinal lesions. Oral proton pump inhibitors and histamine H(2)-receptor blockers were given 1 h before aspirin injection. 16,16-dimethyl prostaglandin E(2) (dmPGE(2)) was given s.c. 30 min before aspirin injection. RESULTS: Aspirin alone clearly induced severe lesions (including bleeding and ulcers) mainly in the jejunum at 100% incidence. Total score of lesions per rat obtained by histological examination was similar to the damaged area quantified with the dye method. Dose-related induction of lesions by aspirin was confirmed both by the histological and dye methods. The irritable effect of aspirin suspended in 0.1 N HCl solution was the same as that of aspirin alone; 0.1 N HCl alone induced only minor lesions in the intestine. Both proton pump inhibitors and histamine H(2)-receptor blockers, at doses that inhibit gastric acid secretion, had no or little effect on aspirin-induced intestinal lesions. Pretreatment with dmPGE(2) (3, 10, 30 microg/kg) showed significant prevention of both aspirin- and HCl/aspirin-induced intestinal lesions. CONCLUSION: This new aspirin lesion model will be useful for screening defensive drugs against aspirin-induced intestinal lesions and to elucidate the underlying mechanism.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Aspirin/administration & dosage , Duodenal Ulcer/chemically induced , Duodenum/pathology , Jejunum/pathology , Peptic Ulcer Hemorrhage/chemically induced , 16,16-Dimethylprostaglandin E2/pharmacology , Animals , Anti-Ulcer Agents/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Duodenal Ulcer/pathology , Duodenal Ulcer/prevention & control , Duodenum/drug effects , Duodenum/ultrastructure , Histamine H2 Antagonists/pharmacology , Injections , Intestinal Mucosa/pathology , Jejunum/drug effects , Jejunum/ultrastructure , Male , Microscopy, Electron, Scanning , Peptic Ulcer Hemorrhage/pathology , Peptic Ulcer Hemorrhage/prevention & control , Proton Pump Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley
14.
J Gastroenterol Hepatol ; 25 Suppl 1: S67-74, 2010 May.
Article in English | MEDLINE | ID: mdl-20586869

ABSTRACT

BACKGROUND AND AIMS: The effects of an EP4 agonist/antagonist on the healing of lesions produced by indomethacin in the small intestine were examined in rats, especially in relation to the expression of vascular endothelial growth factor (VEGF) and angiogenesis. METHODS: Animals were given indomethacin (10 mg/kg s.c.) and killed at various time points. To impair the healing of these lesions, a small dose of indomethacin (2 mg/kg p.o.) or AE3-208 (EP4 antagonist: 3 mg/kg i.p.) was given once daily for 6 days after the ulceration was induced, with or without the co-administration of AE1-329 (EP4 agonist: 0.1 mg/kg i.p.). RESULTS: Indomethacin (10 mg/kg) caused severe damage in the small intestine, but the lesions healed rapidly decreasing to approximately one-fifth of their initial size within 7 days. The healing process was significantly impaired by indomethacin (2 mg/kg) given once daily for 6 days after the ulceration. This effect of indomethacin was mimicked by the EP4 antagonist and reversed by co-administration of the EP4 agonist. Mucosal VEGF expression was upregulated after the ulceration, reaching a peak on day 3 followed by a decrease. The changes in VEGF expression paralleled those in mucosal cyclooxygenase-2 expression, as well as prostaglandin E(2) (PGE(2)) content. Indomethacin (2 mg/kg) downregulated both VEGF expression and angiogenesis in the mucosa during the healing process, and these effects were significantly reversed by co-treatment with the EP4 agonist. CONCLUSION: The results suggest that endogenous PGE(2) promotes the healing of small intestinal lesions by stimulating angiogenesis through the upregulation of VEGF expression mediated by the activation of EP4 receptors.


Subject(s)
Dinoprostone/metabolism , Indomethacin , Intestinal Mucosa/drug effects , Intestine, Small/metabolism , Peptic Ulcer/metabolism , Receptors, Prostaglandin E/metabolism , Vascular Endothelial Growth Factor A/metabolism , Wound Healing , 16,16-Dimethylprostaglandin E2/pharmacology , Animals , Cyclooxygenase 1/genetics , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Disease Models, Animal , Gene Expression Regulation, Enzymologic , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestine, Small/drug effects , Intestine, Small/pathology , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Naphthalenes/pharmacology , Neovascularization, Physiologic/drug effects , Peptic Ulcer/chemically induced , Peptic Ulcer/drug therapy , Peptic Ulcer/pathology , Phenylbutyrates/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Prostaglandin E/agonists , Receptors, Prostaglandin E/antagonists & inhibitors , Receptors, Prostaglandin E, EP4 Subtype , Severity of Illness Index , Time Factors , Up-Regulation , Wound Healing/drug effects
15.
Pharmacology ; 84(5): 294-9, 2009.
Article in English | MEDLINE | ID: mdl-19816089

ABSTRACT

Rat gastric mucosal damage was induced by ischemia-reperfusion. The 5-lipoxygenase inhibitors MK886 and A63162, the 12-lipoxygenase inhibitor baicalein, the 15-lipoxygenase inhibitor PD146176 and the lipoxin (LX) A(4)/annexin 1 antagonist Boc1 increased mucosal damage in a dose-dependent manner. Low doses of these compounds, which have no effects on mucosal integrity, cause severe damage when combined with low doses of indomethacin, celecoxib or dexamethasone. 16,16-Dimethylprostaglandin (PG) E(2) and LXA(4) can replace each other in preventing mucosal injury induced by either cyclooxygenase or lipoxygenase inhibitors. The results suggest that not only cyclooxygenases, but also lipoxygenases have a role in limiting gastric mucosal damage during ischemia-reperfusion.


Subject(s)
Annexins/metabolism , Gastric Mucosa/drug effects , Lipoxygenase/physiology , Receptors, Formyl Peptide/physiology , Receptors, Lipoxin/physiology , Reperfusion Injury/drug therapy , 16,16-Dimethylprostaglandin E2/pharmacology , Acetamides/pharmacology , Animals , Anti-Ulcer Agents/pharmacology , Celecoxib , Cyclooxygenase Inhibitors/pharmacology , Dexamethasone/pharmacology , Drug Synergism , Flavanones/pharmacology , Fluorenes/pharmacology , Gastric Mucosa/blood supply , Gastric Mucosa/pathology , Glucocorticoids/pharmacology , Indoles/pharmacology , Indomethacin/pharmacology , Lipoxygenase Inhibitors/pharmacology , Male , Oligopeptides/pharmacology , Phenyl Ethers , Prostaglandin Antagonists/pharmacology , Pyrazoles/pharmacology , Rats , Rats, Wistar , Receptors, Lipoxin/antagonists & inhibitors , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Sulfonamides/pharmacology
16.
Proc Natl Acad Sci U S A ; 106(31): 13094-9, 2009 Aug 04.
Article in English | MEDLINE | ID: mdl-19622732

ABSTRACT

Carbonic anhydrase (CA) is strongly expressed in the duodenum and has been implicated in a variety of physiological functions including enterocyte HCO(3)(-) supply for secretion and the "sensing" of luminal acid and CO(2). Here, we report the physiological role of the intracellular CAII isoform involvement in acid-, PGE(2,) and forskolin-induced murine duodenal bicarbonate secretion (DBS) in vivo. CAII-deficient and WT littermates were studied in vivo during isoflurane anesthesia. An approximate 10-mm segment of the proximal duodenum with intact blood supply was perfused under different experimental conditions and DBS was titrated by pH immediately. Two-photon confocal microscopy using the pH-sensitive dye SNARF-1F was used to assess duodenocyte pH(i) in vivo. After correction of systemic acidosis by infusion of isotonic Na(2)CO(3), basal DBS was not significantly different in CAII-deficient mice and WT littermates. The duodenal bicarbonate secretory response to acid was almost abolished in CAII-deficient mice, but normal to forskolin- or 16,16-dimethyl PGE(2) stimulation. The complete inhibition of tissue CAs by luminal methazolamide and i.v. acetazolamide completely blocked the response to acid, but did not significantly alter the response to forskolin. While duodenocytes acidified upon luminal perfusion with acid, no significant pH(i) change occurred in CAII-deficient duodenum in vivo. The results suggest that CA II is important for duodenocyte acidification by low luminal pH and for eliciting the acid-mediated HCO(3)(-) secretory response, but is not important in the generation of the secreted HCO(3)(-) ions.


Subject(s)
Bicarbonates/metabolism , Carbonic Anhydrase II/physiology , Duodenum/metabolism , 16,16-Dimethylprostaglandin E2/pharmacology , Acetazolamide/pharmacology , Animals , Carbon Dioxide/metabolism , Carbonic Anhydrase II/antagonists & inhibitors , Colforsin/pharmacology , Hydrogen-Ion Concentration , Methazolamide/pharmacology , Mice , Mice, Inbred C57BL
17.
Am J Respir Cell Mol Biol ; 41(1): 50-8, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19059888

ABSTRACT

In this study, we evaluated the protective effect and therapeutic potential of the prostaglandin E(2) (PGE(2)) synthetic analog 16,16-dimethyl-PGE(2) (dmPGE(2)) in the animal model of pulmonary fibrosis induced by bleomycin. Mice subjected to intratracheal administration of bleomycin (1 mg/kg) received a dmPGE(2) dose of 30 microg/kg/day by continuous subcutaneous infusion. Bronchoalveolar lavage (BAL); immunohistochemical analysis for IL-1, TNF-alpha, and nitrotyrosine; measurement of fluid content in lung; myeloperoxidase activity assay; and lung histology were performed 1 week later. Lung histology and Sircol assay for collagen deposition were performed 3 weeks after treatments. Changes of body weight and survival rate were also evaluated at 1 and 3 weeks. Compared with bleomycin-treated mice, dmPGE(2) co-treated mice exhibited a reduced degree of body weight loss and mortality rate as well as of lung damage and inflammation, as shown by the significant reduction of: (1) lung infiltration by leukocytes; (2) myeloperoxidase activity; (3) IL-1, TNF-alpha, and nitrotyrosine immunostaining; (4) lung edema; and (5) histologic evidence of lung injury and collagen deposition. In a separate set of experiments, dmPGE(2) treatment was started 3 days after bleomycin administration, and the evaluation of lung damage and inflammation was assessed 4 days later. Importantly, delayed administration of dmPGE(2) also was able to protect from inflammation and lung injury induced by bleomycin. These results, indicating that dmPGE(2) is able to prevent and to reduce bleomycin-induced lung injury through its regulatory and anti-inflammatory properties, encourage further research to find new options for the treatment of pulmonary fibrosis.


Subject(s)
16,16-Dimethylprostaglandin E2/pharmacology , Lung Injury/prevention & control , Lung/drug effects , Protective Agents/pharmacology , Pulmonary Fibrosis/prevention & control , 16,16-Dimethylprostaglandin E2/administration & dosage , Animals , Bleomycin , Body Weight/drug effects , Bronchoalveolar Lavage Fluid/cytology , Collagen/metabolism , Disease Models, Animal , Infusions, Subcutaneous , Interleukin-1beta/metabolism , Lung/immunology , Lung/pathology , Lung Injury/chemically induced , Lung Injury/immunology , Lung Injury/pathology , Male , Mice , Peroxidase/metabolism , Pneumonia/chemically induced , Pneumonia/prevention & control , Protective Agents/administration & dosage , Pulmonary Edema/chemically induced , Pulmonary Edema/prevention & control , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/pathology , Time Factors , Tumor Necrosis Factor-alpha/metabolism , Tyrosine/analogs & derivatives , Tyrosine/metabolism
18.
Pharmacology ; 81(3): 236-40, 2008.
Article in English | MEDLINE | ID: mdl-18212515

ABSTRACT

The majority of prostaglandins (PGs) are known to induce intestinal fluid secretion (enteropooling). In contrast, PGD(2) has been demonstrated to inhibit fluid secretion induced by other PGs. This study was aimed to investigate, by the use of selective agonists/antagonists, which type of PGD(2) receptor mediates this inhibitory effect. The DP1 agonist BW245C dose-dependently inhibited the enteropooling effect of 16,16-dimethyl-PGE(2). This inhibition was counteracted by the DP1 antagonist BWA868C. In contrast, the CRTH2 receptor does not seem to be involved in the anti-enteropooling effect of PGD(2), since the selective agonists 13,14-dihydro-15-keto-PGD(2) and 15(R)-15-methyl-PGD(2) were without effect. Therefore, our results suggest that the inhibitory effect of PGD(2) in the small intestine is mediated via activation of the DP1 receptor.


Subject(s)
Intestinal Secretions/drug effects , Prostaglandin D2/pharmacology , Receptors, Immunologic/metabolism , Receptors, Prostaglandin/metabolism , 16,16-Dimethylprostaglandin E2/pharmacology , Animals , Female , Hydantoins/administration & dosage , Hydantoins/pharmacology , Intestine, Small/drug effects , Intestine, Small/metabolism , Prostaglandin D2/analogs & derivatives , Rats , Rats, Sprague-Dawley , Receptors, Immunologic/agonists , Receptors, Immunologic/antagonists & inhibitors , Receptors, Prostaglandin/agonists , Receptors, Prostaglandin/antagonists & inhibitors
19.
Am J Physiol Cell Physiol ; 294(1): C223-32, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17959725

ABSTRACT

Cyclooxygenase-1 (Cox-1) contributes to gastric defense of healthy tissue, but the role in the protection of the gastric epithelium after minor, acute damage has been difficult to study in vivo. Using 710-nm two-photon light absorption to create microscopic gastric damage in anesthetized mice with the gastric mucosal surface surgically exposed and perfused on the microscope stage, the acute response of surface cells to injury could be monitored using in vivo microscopy within seconds after injury. Using exogenous (Cl-NERF) and endogenous fluorophores, extracellular pH and cell death were monitored in real time during the entire damage and repair cycle. Two-photon damage was initiated by scanning approximately 200 microm(2) of gastric surface cells with high laser intensity, causing rapid bleaching of NAD(P)H fluorescence in optically targeted cells. In both Cox-1(+/-) and Cox-1(-/-) mice, a similar initial damage area expanded to include bystander epithelial cells over the next 2-5 min, with larger maximal damage noted in Cox-1(-/-) mice. The maximal damage size seen in Cox-1(-/-) mice could be reduced by exogenous dimethyl-PGE(2). All damaged cells exfoliated, and the underlying epithelium was coincidently repaired over a time interval that was briefer in Cox-1(+/-) (12 +/- 2 min, n = 12) than in Cox-1(-/-) (24 +/- 4 min, n = 14) mice. Directly after damage, pH increased transiently in the juxtamucosal layer (maximal at 3-6 min). A smaller peak pH change was noted in Cox-1(-/-) mice (DeltapH = 0.3 +/- 0.04) than in Cox-1(+/-) mice (DeltapH = 0.6 +/- 0.2). Recovery to normal surface pH took longer in Cox-1(-/-) mice (27 +/- 5 min) than in Cox-1(+/-) mice (12 +/- 1 min). In conclusion, constitutive loss of Cox-1 leaves the gastric mucosa more prone to damage and slowed repair of microlesions.


Subject(s)
Cyclooxygenase 1/metabolism , Gastric Mucosa/enzymology , Lasers , Membrane Proteins/metabolism , NADP/radiation effects , Wound Healing , 16,16-Dimethylprostaglandin E2/pharmacology , Animals , Cell Death , Cyclooxygenase 1/deficiency , Cyclooxygenase 1/genetics , Gastric Mucosa/drug effects , Gastric Mucosa/pathology , Gastric Mucosa/radiation effects , Hydrogen-Ion Concentration , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Knockout , Microscopy, Confocal , Microscopy, Fluorescence, Multiphoton , Microscopy, Video , Models, Animal , Time Factors , Wound Healing/drug effects
20.
Eur J Immunol ; 37(6): 1562-74, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17474154

ABSTRACT

During an acute blood-stage malaria infection, T cell responses to malaria and other bystander antigens are inhibited. Plasmodium infection induces strong cytokine responses that facilitate parasite clearance but may interfere with T cell functions, as some of the soluble immune mediators induced are also general inhibitors of T cell responses. Using a malaria mouse model, we have analyzed the cytokines produced by dendritic cells in response to P. yoelii infection that have potential T cell inhibitory activity. We found that during acute infection DC migrate to the spleen and secrete TGF-beta, prostaglandin E2 (PGE2) and IL-10. We have analyzed the role of these general T cell inhibitors in a particular T cell response of evident importance in malaria infections: the CD8+ T cells generated against the liver-stage of the disease. During blood-stage infection, inhibition of the activity of TGF-beta and PGE2 restores the CD8+ T cell responses generated by sporozoites, increasing protection against re-infection. Our findings suggest that the strong cytokine response induced by blood-stage P. yoelii infection affects host T cell responses, inhibiting protective CD8+ T cells against the liver-stage of the disease.


Subject(s)
Dinoprostone/metabolism , Malaria/immunology , Plasmodium yoelii/immunology , T-Lymphocytes/immunology , Transforming Growth Factor beta/metabolism , 16,16-Dimethylprostaglandin E2/pharmacology , Adoptive Transfer , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antigens, Protozoan/immunology , CD11c Antigen/analysis , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/transplantation , Dinoprostone/physiology , Erythrocytes/immunology , Erythrocytes/parasitology , Interleukin-10/metabolism , Malaria/metabolism , Mice , Mice, Inbred BALB C , Peptide Fragments/pharmacology , Receptors, CCR7 , Receptors, Chemokine/metabolism , Receptors, Transforming Growth Factor beta , Spleen/cytology , Spleen/immunology , Spleen/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Transforming Growth Factor beta/physiology , Vaccination
SELECTION OF CITATIONS
SEARCH DETAIL
...