Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 302
Filter
1.
Sci Adv ; 6(1): eaay2432, 2020 01.
Article in English | MEDLINE | ID: mdl-31911947

ABSTRACT

The mechanism by which the cytosolic protein Zap70 physically interacts with and phosphorylates its substrate, the transmembrane protein LAT, upon T cell receptor (TCR) stimulation remains largely obscure. In this study, we found that the pharmacological inhibition of formins, a major class of actin nucleators, suppressed LAT phosphorylation by Zap70, despite TCR stimulation-dependent phosphorylation of Zap70 remaining intact. High-resolution imaging and three-dimensional image reconstruction revealed that localization of phosphorylated Zap70 to the immune synapse (IS) and subsequent LAT phosphorylation are critically dependent on formin-mediated actin polymerization. Using knockout mice, we identify mDia1 and mDia3, which are highly expressed in T cells and which localize to the IS upon TCR activation, as the critical formins mediating this process. Our findings therefore describe previously unsuspected roles for mDia1 and mDia3 in the spatiotemporal control of Zap70-dependent LAT phosphorylation at the IS through regulation of filamentous actin, and underscore their physiological importance in TCR signaling.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Formins/immunology , Membrane Proteins/genetics , ZAP-70 Protein-Tyrosine Kinase/genetics , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/immunology , Actins/antagonists & inhibitors , Actins/chemistry , Actins/genetics , Adaptor Proteins, Signal Transducing/immunology , Animals , Formins/genetics , Formins/pharmacology , Gene Expression Regulation/drug effects , Humans , Immune System/drug effects , Immune System/metabolism , Jurkat Cells/immunology , Membrane Proteins/immunology , Mice , Mice, Knockout , Phosphorylation/drug effects , Polymerization/drug effects , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Signal Transduction/drug effects
2.
Scand J Med Sci Sports ; 28(3): 826-833, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28759126

ABSTRACT

We previously demonstrated that nerve growth factor (NGF) and glial cell line-derived neurotrophic factor (GDNF) were upregulated after lengthening contractions (LC) in exercised muscle through B2 bradykinin receptor activation and cyclooxygenase (COX)-2 upregulation, respectively, and that these trophic factors sensitized nociceptors resulting in mechanical hyperalgesia (delayed-onset muscle soreness, DOMS). Here, we examined the prostaglandin receptor subtype involved in DOMS. The mechanical withdrawal threshold of the exercised muscle was measured before and after LC in rats administered prostaglandin E2 receptor (EP) antagonists before LC, or in wild-type (WT), EP2 knockout (EP2-/- ), and IP knockout (IP-/- ) mice. The change in expression of NGF, GDNF, or COX-2 mRNA was examined using real-time RT-PCR in the muscle in EP2-/- and WT mice. None of the antagonists to EP1, EP3, and EP4 receptors (ONO-8713, ONO-AE5-599, and ONO-AE3-208, respectively) induced a significant difference in DOMS compared with controls in rats. WT and IP-/- mice developed mechanical hyperalgesia after LC, but EP2-/- mice did not. Upregulation of NGF, GDNF, and COX-2 mRNA was observed after LC in WT mice but not in EP2-/- mice. Injecting an EP2 agonist (ONO-AE1-259-01) into the mouse muscle increased expression of COX-2 mRNA. These results suggest that EP2 contributes to generating mechanical hyperalgesia through positive feedback upregulation of COX-2 expression in muscle after LC.


Subject(s)
Hyperalgesia/physiopathology , Muscle Contraction , Myalgia/physiopathology , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Animals , Cyclooxygenase 2/metabolism , Dinoprostone/analogs & derivatives , Dinoprostone/pharmacology , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Nerve Growth Factor/metabolism , Physical Conditioning, Animal , Rats , Rats, Sprague-Dawley , Receptors, Prostaglandin E, EP2 Subtype/agonists , Receptors, Prostaglandin E, EP2 Subtype/antagonists & inhibitors
3.
Mol Psychiatry ; 23(8): 1717-1730, 2018 08.
Article in English | MEDLINE | ID: mdl-28924188

ABSTRACT

Dopamine in prefrontal cortices is implicated in cognitive and emotional functions, and the dysfunction of prefrontal dopamine has been associated with cognitive and emotional deficits in mental illnesses. These findings have led to clinical trials of dopamine-targeting drugs and brain imaging of dopamine receptors in patients with mental illnesses. Rodent studies have suggested that dopaminergic pathway projecting to the medial prefrontal cortex (mPFC) suppresses stress susceptibility. Although various types of mPFC neurons express several dopamine receptor subtypes, previous studies neither isolated a role of dopamine receptor subtype nor identified the site of its action in mPFC. Using social defeat stress (SDS) in mice, here we identified a role of dopamine D1 receptor subtype in mPFC excitatory neurons in suppressing stress susceptibility. Repeated social defeat stress (R-SDS) reduces the expression of D1 receptor subtype in mPFC of mice susceptible to R-SDS. Knockdown of D1 receptor subtype in whole neuronal populations or excitatory neurons in mPFC facilitates the induction of social avoidance by SDS. Single social defeat stress (S-SDS) induces D1 receptor-mediated extracellular signal-regulated kinase phosphorylation and c-Fos expression in mPFC neurons. Whereas R-SDS reduces dendritic lengths of mPFC layer II/III pyramidal neurons, S-SDS increases arborization and spines of apical dendrites of these neurons in a D1 receptor-dependent manner. Collectively, our findings show that D1 receptor subtype and related signaling in mPFC excitatory neurons mediate acute stress-induced dendritic growth of these neurons and contribute to suppression of stress susceptibility. Therefore, we propose that D1 receptor-mediated dendritic growth in mPFC excitatory neurons suppresses stress susceptibility.


Subject(s)
Dendrites/metabolism , Prefrontal Cortex/metabolism , Receptors, Dopamine D1/metabolism , Resilience, Psychological , Stress, Psychological/metabolism , Animals , Avoidance Learning/physiology , Cell Enlargement , Dendrites/pathology , Disease Models, Animal , Disease Susceptibility/metabolism , Dominance-Subordination , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Knockdown Techniques , Male , Mice, Inbred C57BL , Mice, Inbred ICR , Prefrontal Cortex/pathology , Proto-Oncogene Proteins c-fos/metabolism , Pyramidal Cells/metabolism , Pyramidal Cells/pathology , Receptors, Dopamine D1/genetics , Stress, Psychological/pathology
4.
Pharmacol Rev ; 63(3): 471-538, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21752876

ABSTRACT

It is now more than 15 years since the molecular structures of the major prostanoid receptors were elucidated. Since then, substantial progress has been achieved with respect to distribution and function, signal transduction mechanisms, and the design of agonists and antagonists (http://www.iuphar-db.org/DATABASE/FamilyIntroductionForward?familyId=58). This review systematically details these advances. More recent developments in prostanoid receptor research are included. The DP(2) receptor, also termed CRTH2, has little structural resemblance to DP(1) and other receptors described in the original prostanoid receptor classification. DP(2) receptors are more closely related to chemoattractant receptors. Prostanoid receptors have also been found to heterodimerize with other prostanoid receptor subtypes and nonprostanoids. This may extend signal transduction pathways and create new ligand recognition sites: prostacyclin/thromboxane A(2) heterodimeric receptors for 8-epi-prostaglandin E(2), wild-type/alternative (alt4) heterodimers for the prostaglandin FP receptor for bimatoprost and the prostamides. It is anticipated that the 15 years of research progress described herein will lead to novel therapeutic entities.


Subject(s)
Receptors, Prostaglandin/classification , Receptors, Thromboxane/classification , Animals , Humans , International Agencies , Molecular Targeted Therapy , Prostaglandin Antagonists/therapeutic use , Prostaglandins/agonists , Prostaglandins/metabolism , Protein Isoforms/chemistry , Protein Isoforms/classification , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, Prostaglandin/chemistry , Receptors, Prostaglandin/genetics , Receptors, Prostaglandin/metabolism , Receptors, Thromboxane/chemistry , Receptors, Thromboxane/genetics , Receptors, Thromboxane/metabolism , Second Messenger Systems/drug effects , Terminology as Topic , Thromboxanes/agonists , Thromboxanes/antagonists & inhibitors , Thromboxanes/metabolism
5.
Br J Pharmacol ; 163(6): 1237-49, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21426319

ABSTRACT

BACKGROUND AND PURPOSE: Cerebral aneurysm is a frequent cerebrovascular event and a major cause of fatal subarachnoid haemorrhage, but there is no medical treatment for this condition. Haemodynamic stress and, recently, chronic inflammation have been proposed as major causes of cerebral aneurysm. Nevertheless, links between haemodynamic stress and chronic inflammation remain ill-defined, and to clarify such links, we evaluated the effects of prostaglandin E(2) (PGE(2) ), a mediator of inflammation, on the formation of cerebral aneurysms. EXPERIMENTAL APPROACH: Expression of COX and prostaglandin E synthase (PGES) and PGE receptors were examined in human and rodent cerebral aneurysm. The incidence, size and inflammation of cerebral aneurysms were evaluated in rats treated with COX-2 inhibitors and mice lacking each prostaglandin receptor. Effects of shear stress and PGE receptor signalling on expression of pro-inflammatory molecules were studied in primary cultures of human endothelial cells (ECs). KEY RESULTS: COX-2, microsomal PGES-1 and prostaglandin E receptor 2 (EP(2) ) were induced in ECs in the walls of cerebral aneurysms. Shear stress applied to primary ECs induced COX-2 and EP(2) . Inhibition or loss of COX-2 or EP(2) in vivo attenuated each other's expression, suppressed nuclear factor κB (NF-κB)-mediated chronic inflammation and reduced incidence of cerebral aneurysm. EP(2) stimulation in primary ECs induced NF-κB activation and expression of the chemokine (C-C motif) ligand 2, essential for cerebral aneurysm. CONCLUSIONS AND IMPLICATIONS: These results suggest that shear stress activated PGE(2) -EP(2) pathway in ECs and amplified chronic inflammation via NF-κB. We propose EP(2) as a therapeutic target in cerebral aneurysm.


Subject(s)
Dinoprostone/metabolism , Intracranial Aneurysm/metabolism , NF-kappa B/metabolism , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Animals , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Dinoprostone/genetics , Humans , Inflammation/genetics , Inflammation/metabolism , Intracranial Aneurysm/drug therapy , Mice , Mice, Knockout , NF-kappa B/genetics , Prostaglandin-Endoperoxide Synthases/genetics , Prostaglandin-Endoperoxide Synthases/metabolism , Rats , Receptors, Prostaglandin E, EP2 Subtype/genetics , Signal Transduction
6.
Clin Exp Immunol ; 163(2): 260-9, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21166666

ABSTRACT

Otitis media is one of the most common and intractable ear diseases, and is the major cause of hearing loss, especially in children. Multiple factors affect the onset or development of otitis media. Prostaglandin D2 is the major prostanoid involved in infection and allergy. However, the role of prostaglandin D2 and prostaglandin D2 receptors on the pathogenesis of otitis media remains to be determined. Recent studies show that D prostanoid receptor (DP) and chemoattractant receptor-homologous molecule expressed on T helper type 2 (Th2) cells (CRTH2) are major prostaglandin D2 receptors. In this study, homozygous DP single gene-deficient (DP⁻(/)⁻) mice, CRTH2 single gene-deficient (CRTH2⁻(/)⁻) mice and DP/CRTH2 double gene-deficient (DP⁻(/)⁻ CRTH2⁻(/)⁻) mice were used to investigate the role of prostaglandin D2 and its receptors in otitis media. We demonstrate that prostaglandin D2 is induced by lipopolysaccharide (LPS), a major component of Gram-negative bacteria, and that transtympanic injection of prostaglandin D2 up-regulates macrophage inflammatory protein 2 (MIP-2), interleukin (IL)-1ß and IL-6 in the middle ear. We also show that middle ear inflammatory reactions, including infiltration of inflammatory cells and expression of MIP-2, IL-1ß and IL-6 induced by LPS, are reduced significantly in DP⁻(/)⁻ mice and DP⁻(/)⁻ CRTH2⁻(/)⁻ mice. CRTH2⁻(/)⁻ mice display inflammatory reactions similar to wild-type mice. These findings indicate that prostaglandin D2 may play significant roles in LPS-induced experimental otitis media via DP.


Subject(s)
Cytokines/immunology , Lipopolysaccharides/immunology , Otitis Media/immunology , Receptors, Immunologic/immunology , Receptors, Prostaglandin/immunology , Animals , Chemokine CXCL2/immunology , Disease Models, Animal , Female , Interleukin-1beta/immunology , Interleukin-6/immunology , Mice , Mice, Inbred BALB C , Prostaglandin D2/immunology , Receptors, Immunologic/genetics , Receptors, Prostaglandin/genetics , Th2 Cells/immunology
7.
Neuroscience ; 170(1): 317-23, 2010 Sep 29.
Article in English | MEDLINE | ID: mdl-20621166

ABSTRACT

We and others have identified that inhibition of cyclooxygenase might not be the optimal approach to limiting brain damage after stroke. Now we are investigating the unique properties of the various prostaglandin receptors to determine whether blocking those that mediate toxicity or stimulating those that reduce toxicity will improve neurological outcomes. Here, we determined the respective contribution of the prostaglandin I(2) (PGI(2)) receptor in transient middle cerebral artery (MCA) occlusion (tMCAO) and permanent MCAO (pMCAO) preclinical stroke models by using male wildtype (WT) and IP receptor knockout (IP(-/-)) C57Bl/6 mice. In addition, we investigated the putative preventive and therapeutic effects of the IP receptor agonist beraprost. The infarct volumes and neurological deficit scores (NDS) were significantly greater in IP(-/-) than in WT mice after both tMCAO and pMCAO. Interestingly, beraprost pretreatment (50 or 100 microg/kg p.o.) 30 min before tMCAO and post-treatment (100 microg/kg p.o.) at 2 or 4.5 h of reperfusion significantly reduced the neurological deficit score and infarct volume in WT mice. Post-treatment with beraprost (100 microg/kg p.o.) 4.5 h after pMCAO also significantly decreased neurological deficits and infarct volume in WT mice. Together, these novel findings suggest for the first time that PGI(2) IP receptor activation can attenuate anatomical and functional damage following ischemic stroke.


Subject(s)
Brain Ischemia/metabolism , Brain Ischemia/prevention & control , Epoprostenol/analogs & derivatives , Neuroprotective Agents/metabolism , Receptors, Epoprostenol/agonists , Receptors, Epoprostenol/physiology , Animals , Cohort Studies , Epoprostenol/metabolism , Epoprostenol/pharmacology , Epoprostenol/therapeutic use , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use
9.
Biomed Pharmacother ; 64(2): 93-100, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20015609

ABSTRACT

Chronic inflammation, which is characterized by the proliferation of granulation tissues, is known to be regulated by angiogenesis. Recent results suggest that bone marrow-derived (BM-derived) hematopoietic cells regulate angiogenesis in vivo. We previously reported that the angiogenesis occurring during chronic inflammation is enhanced in response to the endogenous prostaglandins (PGs) derived from an inducible cyclooxygenase-2 (COX-2). In the present study, we examined the role of BM-derived cells expressing an E-type PG receptor subtype, EP3, in sponge-induced angiogenesis. The replacement of wild-type (WT) BM with BM cells (BMCs) from green fluorescent protein (GFP) transgenic mice revealed that the formation of granulation tissue around the sponge implants developed via the recruitment of BMCs. This recruitment was enhanced by topical injections of vascular endothelial growth factor (VEGF)-A, and a VEGF-dependent increase in the recruitment of BMCs was inhibited by a COX-2 inhibitor, celecoxib. FACS analysis of the granulation tissues after treatment with collagenase revealed that the Mac-1-positive macrophage fraction was enhanced by topical injections of VEGF-A, and that this increased recruitment of Mac-1-positive BMCs was inhibited by celecoxib. Selective knockdown of EP3 performed by BM transplantation with BMCs isolated from EP3 knockout (EP3) mice reduced sponge-induced angiogenesis, as estimated by mean vascular number and CD31 expression in the granulation tissues. This reduction in angiogenesis in EP3(-/-) BM chimeric mice was accompanied by reductions in the recruitment of BMCs, especially of Mac-1-positive cells and Gr-1-positive cells. These results indicate that the recruited bone marrow cells that express the EP3 receptor have a significant role in enhancing angiogenesis during chronic proliferative inflammation.


Subject(s)
Bone Marrow Cells/metabolism , Bone Marrow Transplantation/methods , Inflammation/metabolism , Neovascularization, Pathologic/metabolism , Receptors, Prostaglandin E/genetics , Animals , Celecoxib , Cyclooxygenase 2/drug effects , Cyclooxygenase 2 Inhibitors/pharmacology , Disease Models, Animal , Granulation Tissue/drug effects , Granulation Tissue/pathology , Inflammation/pathology , Inflammation/surgery , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Pathologic/pathology , Neutrophil Infiltration/drug effects , Neutrophil Infiltration/genetics , Pyrazoles/pharmacology , Receptors, Prostaglandin E, EP3 Subtype , Sulfonamides/pharmacology
10.
Neuroscience ; 156(4): 1111-7, 2008 Oct 28.
Article in English | MEDLINE | ID: mdl-18790018

ABSTRACT

Transient global cerebral ischemia causes delayed neuronal death in the hippocampal CA1 region. It also induces an increase in cyclooxygenase 2 (COX-2), which generates several metabolites of arachidonic acid, known as prostanoids, including prostacyclin (PGI(2)). To determine the role of the PGI(2) receptor (IP) in post-ischemic delayed cell death, wild-type and IP knockout (IP(-/-)) C57Bl/6 mice were subjected to 12-min bilateral common carotid artery occlusion or sham surgery, followed by 7 days of reperfusion. In the sham-operated mice, no statistical difference in CA1 hippocampal neuronal density was observed between the wild-type (2836+/-18/mm(2)) and IP(-/-) (2793+/-43/mm(2)) mice. Interestingly, in animals subjected to ischemia, surviving neuronal density in wild-type mice decreased to 50.5+/-7.9% and that of IP(-/-) mice decreased to 23.0+/-4.5% of their respective sham-operated controls (P<0.05). The results establish a role for the IP receptor in protecting pyramidal hippocampal neurons after this global ischemic model and suggest that IP receptor agonists could be developed to prevent delayed pyramidal neuronal cell death.


Subject(s)
Carotid Artery Diseases/genetics , Carotid Artery Diseases/pathology , Hippocampus/pathology , Pyramidal Cells/pathology , Pyramidal Cells/physiopathology , Receptors, Epoprostenol/deficiency , Animals , Blood Pressure/genetics , Body Temperature/genetics , Brain Ischemia/etiology , Brain Ischemia/genetics , Brain Ischemia/pathology , Carotid Artery Diseases/complications , Cell Death/physiology , Cerebrovascular Circulation/genetics , Cerebrovascular Circulation/physiology , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Reperfusion
11.
Exp Eye Res ; 84(1): 39-43, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17052707

ABSTRACT

The purpose of the study was to investigate the role of EP1 receptors in intraocular inflammation and to determine possible interplay between EP1, EP2 and EP4 receptors. The eyes of separate groups of EP1 receptor knockout and wild type mice were: 1) treated topically with prostaglandin E2 (PGE2) or the EP2 receptor selective agonist, butaprost; 2) given intravitreal injection of LPS; or 3) paracentesis performed. Another group of knockout mice were pretreated topically with an EP4 receptor selective antagonist prior to paracentesis or LPS treatment. Results demonstrated a significant increase (50% or more) in the protein levels of aqueous humor of the EP1 knockout mice in response to PGE2, paracentesis or LPS. The leukocyte infiltration in the aqueous humor of the knockout mice was 47% higher when compared with that in the wild type controls in response to LPS injection. No significant change was observed in the protein levels in response to butaprost. Pretreating the knockout mice with an EP4 receptor antagonist prior to paracentesis and LPS treatment substantially reduced the aqueous humor protein levels. Also, the leukocyte count in the aqueous humor of the knockout mice in response to LPS was reduced 4 fold after pretreatment with EP4 receptor antagonist when compared with the findings in knockout mice receiving LPS only. We concluded that EP1 receptor has no modulatory effect on EP2 receptors but there is definitely cross-talk between EP1 and EP4 receptors.


Subject(s)
Blood-Aqueous Barrier/physiology , Inflammation/physiopathology , Receptors, Prostaglandin E/physiology , Alprostadil/analogs & derivatives , Alprostadil/pharmacology , Animals , Aqueous Humor/cytology , Aqueous Humor/metabolism , Blood-Aqueous Barrier/drug effects , Dinoprostone/pharmacology , Eye Proteins/metabolism , Inflammation/chemically induced , Inflammation/metabolism , Leukocyte Count , Lipopolysaccharides , Mice , Mice, Inbred C57BL , Mice, Knockout , Paracentesis , Receptors, Prostaglandin E/agonists , Receptors, Prostaglandin E/antagonists & inhibitors , Receptors, Prostaglandin E, EP1 Subtype , Receptors, Prostaglandin E, EP2 Subtype , Receptors, Prostaglandin E, EP4 Subtype
12.
Gut ; 53(8): 1151-8, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15247185

ABSTRACT

BACKGROUND AND AIMS: Involvement of prostaglandin E(2) (PGE(2)) receptors EP(1), EP(2), and EP(4) in the formation of aberrant crypt foci (ACF) and/or intestinal polyps has been suggested. In contrast, EP(3) appears to have no influence on the early stages of colon carcinogenesis. In the present study, we examined expression of PGE(2) receptor subtypes EP(1), EP(2), EP(3), and EP(4) in normal colon mucosa and colon cancers, and assessed the contribution of EP(3) to colon cancer development. METHODS: mRNA expression of PGE(2) receptor subtypes EP(1), EP(2), EP(3), and EP(4) in normal colon mucosa and colon cancers in azoxymethane (AOM) treated mice and rats, and in humans, were examined by reverse transcription-polymerase chain reaction (RT-PCR), quantitative real time RT-PCR, and immunohistochemical analyses. Evaluation of the role of EP(3) was performed by intraperitoneal injection of AOM, using EP(3) receptor knockout mice. Effects of EP(3) receptor activation on cell growth of human colon cancer cell lines were examined using ONO-AE-248, an EP(3) selective agonist. Moreover, EP(3) expression in colon cancer cell lines was analysed with or without 5-aza-2'-deoxycytidine (5-aza-dC) treatment. RESULTS: Expression levels of EP(1) and EP(2) mRNA were increased in cancer tissues. EP(4) mRNA was constantly expressed in normal mucosa and cancers. In contrast, expression of EP(3) mRNA was markedly decreased in colon cancer tissues, being 5% in mice, 9% in rats, and 28% in humans compared with normal colon mucosa, analysed by quantitative real time RT-PCR. Immunohistochemical staining demonstrated the rat EP(3) receptor protein to be expressed in epithelial cells of normal mucosa and some parts of small carcinomas but hardly detectable in large carcinomas of the colon. Colon cancer development induced by AOM in EP(3) receptor knockout mice was enhanced compared with wild-type mice, with a higher incidence of colon tumours (78% v 57%) and mean number of tumours per mouse (2.17 (0.51) v 0.75 (0.15); p<0.05). Expression of EP(3) mRNA was detected in only one of 11 human colon cancer cell lines tested. Treatment with 5 microM of an EP(3) selective agonist, ONO-AE-248, resulted in a 30% decrease in viable cell numbers in the HCA-7 human colon cancer cell line in which EP(3) was expressed. Treatment with 5-aza-dC restored EP(3) expression in CACO-2, CW-2, and DLD-1 cells but not in WiDr cells, suggesting involvement of hypermethylation in the downregulation of EP(3) to some extent. CONCLUSION: The PGE(2) receptor subtype EP(3) plays an important role in suppression of cell growth and its downregulation enhances colon carcinogenesis at a later stage. Hypermethylation of the EP(3) receptor gene could occur and may contribute towards downregulating EP(3) expression to some extent in colon cancers.


Subject(s)
Colonic Neoplasms/metabolism , Receptors, Prostaglandin E/metabolism , Animals , Caco-2 Cells , Cell Line, Tumor , Colon/metabolism , Dinoprostone/analogs & derivatives , Dinoprostone/pharmacology , Down-Regulation , Humans , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/analysis , RNA, Neoplasm/analysis , Rats , Receptors, Prostaglandin E/agonists , Receptors, Prostaglandin E, EP3 Subtype , Receptors, Prostaglandin E, EP4 Subtype
13.
Gut ; 52(9): 1242-9, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12912853

ABSTRACT

BACKGROUND: We previously reported that endogenous prostaglandin I(2), generated by a mild irritant, sensitised calcitonin gene related peptide (CGRP) containing sensory nerves and facilitated the release of CGRP and gastric mucosal protection against ethanol. Administration of capsaicin also inhibited ethanol induced gastric mucosal injury through immediate release of CGRP from primary sensory neurones, which is termed the neural emergency system. In the present study, we tested whether endogenous prostaglandin I(2) also modulates the cytoprotective action of capsaicin using prostaglandin I receptor knockout mice (IP(-/-)). METHODS: The stomachs of IP(-/-) or their wild-type counterparts (IP(+/+)), anaesthetised with urethane (1.225 g/kg), were doubly cannulated from the oesophageal and duodenal sides, and the gastric mucosa was perfused (1 ml/min) with physiological saline. Perfusate was changed to 50% ethanol alone, or 50% ethanol containing capsaicin (16 approximately 1600 micro M). The injured area was estimated at the end of each perfusion experiment. In some animals, CGRP-(8-37), a CGRP antagonist (0.3 mg/kg), or indomethacin (1 mg/kg) was intravenously injected before perfusion of 50% ethanol containing capsaicin. RESULTS: Capsaicin inhibited the injured area in a dose dependent manner. Fifty per cent ethanol containing capsaicin (480 micro M) immediately increased intragastric levels of CGRP although 50% ethanol alone did not. The protective action of capsaicin (480 micro M) against ethanol was completely abolished by intravenous injection of CGRP-(8-37). Indomethacin also inhibited the protective action of capsaicin, and this was accompanied by reduced levels of intragastric CGRP. Intragastric levels of prostaglandin E(2) were not increased by capsaicin treatment but those of 6-keto-prostaglandin F(1alpha), a metabolite of prostaglandin I(2), were markedly increased. No protective action of capsaicin was observed in IP(-/-) which lacked the ability to increase intragastric CGRP levels in response to ethanol containing capsaicin. The CGRP content of the stomach from untreated IP(-/-) did not differ from those in IP(+/+). Capsaicin (160 micro M) together with intragastric perfusion of beraprost sodium (PGI(2) analogue, 2.5 micro g/ml) showed enhanced protection against ethanol induced injury. This enhanced protection was completely blocked by intravenous injection of CGRP-(8-37). CONCLUSIONS: The present results suggest that endogenous prostaglandin I(2) enhances the protective action of the capsaicin mediated neural emergency system against ethanol induced gastric mucosal injury through enhancement of CGRP release.


Subject(s)
Calcitonin Gene-Related Peptide/metabolism , Capsaicin/therapeutic use , Epoprostenol/physiology , Ethanol/adverse effects , Gastric Mucosa/drug effects , 6-Ketoprostaglandin F1 alpha/metabolism , Animals , Calcitonin Gene-Related Peptide/analysis , Capsaicin/antagonists & inhibitors , Cyclooxygenase Inhibitors/administration & dosage , Dose-Response Relationship, Drug , Ethanol/administration & dosage , Gastric Mucosa/metabolism , Indomethacin/administration & dosage , Mice , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction/methods
14.
Circulation ; 104(18): 2210-5, 2001 Oct 30.
Article in English | MEDLINE | ID: mdl-11684633

ABSTRACT

BACKGROUND: Prostaglandin (PG) I(2) and thromboxane (TX) A(2), the most common prostanoids in the cardiovascular system, are produced abundantly during cardiac ischemia/reperfusion (I/R); their roles in I/R injury, however, remain undetermined. We intended to clarify these roles of PGI(2) and TXA(2) using mice lacking the PGI(2) receptor, IP(-/-) mice, or the TXA(2) receptor, TP(-/-) mice. METHODS AND RESULTS: The left anterior descending coronary artery was occluded for 1 hour and then reperfused for 24 hours. The size of myocardial infarct in IP(-/-) mice was significantly larger than that in wild-type mice, although the size of the area at risk was similar between the 2 groups of mice. In contrast, there was no such difference between TP(-/-) and wild-type mice. To further determine whether PGI(2) and TXA(2) act directly on the cardiac tissue or indirectly through their action on blood constituents, we perfused excised heart according to the Langendorff technique. The isolated heart was then subjected to global ischemia followed by reperfusion. In IP(-/-) mice, developed tension and coronary flow rate during reperfusion were significantly lower and release of creatine kinase was significantly higher than those in wild-type mice. There were no such differences, however, between TP(-/-) and wild-type mice. CONCLUSIONS: PGI(2), which was produced endogenously during cardiac I/R, exerts a protective effect on cardiomyocytes independent of its effects on platelets and neutrophils. In contrast, TXA(2) has little role in the cardiac I/R injury.


Subject(s)
Epoprostenol/metabolism , Myocardial Reperfusion Injury/metabolism , Receptors, Prostaglandin/deficiency , Receptors, Thromboxane/deficiency , Thromboxane A2/metabolism , Adenosine Triphosphate/metabolism , Animals , Blood Flow Velocity/drug effects , Blood Pressure/drug effects , Coronary Circulation , Creatine Kinase/metabolism , Cytoprotection/drug effects , Disease Models, Animal , Electrocardiography , Epoprostenol/pharmacology , Heart/drug effects , Heart/physiopathology , Heart Rate/drug effects , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Ischemia/metabolism , Myocardial Ischemia/pathology , Myocardial Reperfusion Injury/pathology , Myocardium/metabolism , Myocardium/pathology , Receptors, Epoprostenol , Receptors, Prostaglandin/genetics , Receptors, Thromboxane/genetics , Thromboxane A2/pharmacology
15.
J Cell Sci ; 114(Pt 18): 3273-84, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11591816

ABSTRACT

Citron-kinase (Citron-K) is a Rho effector working in cytokinesis. It is enriched in cleavage furrow, but how Rho mobilizes Citron-K remains unknown. Using anti-Citron antibody and a Citron-K Green Fluorescence Protein (GFP)-fusion, we monitored its localization in cell cycle. We have found: (1) Citron-K is present as aggregates in interphase cells, disperses throughout the cytoplasm in prometaphase, translocates to cell cortex in anaphase and accumulates in cleavage furrow in telophase; (2) Rho colocalizes with Citron-K in the cortex of ana- to telophase cells and the two proteins are concentrated in the cleavage furrow and to the midbody; (3) inactivation of Rho by C3 exoenzyme does not affect the dispersion of Citron-K in prometaphase, but prevented its transfer to the cell cortex, and Citron-K stays in association with the midzone spindles of C3 exoenzyme-treated cells. To clarify further the mechanism of the Rho-mediated transfer and concentration of Citron-K in cleavage furrow, we expressed active Val14RhoA in interphase cells expressing GFP-Citron-K. Val14RhoA expression transferred Citron-K to the ventral cortex of interphase cells, where it formed band-like structures in a complex with Rho. This structure was localized at the same plane as actin stress fibers, and they exclude each other. Disruption of F-actin abolished the band and dispersed the Citron-K-Rho-containing patches throughout the cell cortex. Similarly, in dividing cells, a structure composed of Rho and Citron-K in cleavage furrow excludes cortical actin cytoskeleton, and disruption of F-actin disperses Citron-K throughout the cell cortex. These results suggest that Citron-K is a novel type of a passenger protein, which is dispersed to the cytoplasm in prometaphase and associated with midzone spindles by a Rho-independent signal. Rho is then activated, binds to Citron-K and translocates it to cell cortex, where the complex is then concentrated in the cleavage furrow by the action of actin cytoskeleton beneath the equator of dividing cells.


Subject(s)
Actin Cytoskeleton/metabolism , Acute-Phase Proteins/metabolism , Botulinum Toxins , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Spindle Apparatus/chemistry , ADP Ribose Transferases/pharmacology , Anaphase/physiology , Cell Cycle/physiology , Cell Division/physiology , Cytoplasm/chemistry , Green Fluorescent Proteins , HeLa Cells/metabolism , Humans , Interphase/physiology , Intracellular Signaling Peptides and Proteins , Luminescent Proteins/chemistry , Luminescent Proteins/genetics , Macromolecular Substances , Metaphase/physiology , Protein Serine-Threonine Kinases/drug effects , Protein Transport/physiology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Subcellular Fractions/chemistry
16.
Mol Endocrinol ; 15(11): 2037-49, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11682632

ABSTRACT

The essential role of CCAAT/enhancer binding proteins (C/EBPs) beta and delta for adipocyte differentiation has been clearly established. In preadipocytes, their expression is up-regulated by the activation of leukemia inhibitory factor receptor (LIF-R) and prostacyclin receptor (IP-R) via the extracellular signal-regulated kinase (ERK) pathway and cAMP production, respectively. However, the molecular mechanisms by which LIF and prostacyclin-induced signals are propagated to the nucleus and the transcription factors mediating ERK and cAMP-induced C/EBP gene expression were unknown. Here we report that both pathways share cAMP responsive element binding protein/activation transcription factor 1 (CREB/ATF-1) as common downstream effectors. LIF-R and IP-R activation induced binding of CREB and/or ATF-1 to C/EBP promoters and CREB-dependent transcription. Expression of dominant negative forms of CREB dramatically reduced the LIF- and prostacyclin-stimulated C/EBP beta and C/EBP delta expression. Upon stimulation of the IP-R, the ERK pathway was activated in a PKA-dependent manner. ERK activation by the PKA pathway was not required for CREB/ATF-1 phosphorylation but rather was necessary for CREB-dependent up-regulation of C/EBPs expression. Our findings suggest that ERK activation is required for CREB transcriptional activity, possibly by recruitment of a coactivator.


Subject(s)
Adipocytes/physiology , CCAAT-Enhancer-Binding Protein-beta/metabolism , CCAAT-Enhancer-Binding Proteins/metabolism , DNA-Binding Proteins , Mitogen-Activated Protein Kinases/metabolism , Transcription Factors/metabolism , Activating Transcription Factor 1 , Animals , CCAAT-Enhancer-Binding Protein-beta/drug effects , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-delta , CCAAT-Enhancer-Binding Proteins/drug effects , CCAAT-Enhancer-Binding Proteins/genetics , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation , Epoprostenol/pharmacology , Gene Expression Regulation/drug effects , Leukemia Inhibitory Factor Receptor alpha Subunit , Mice , Mice, Inbred Strains , Mice, Mutant Strains , Mitogen-Activated Protein Kinases/genetics , Phosphorylation , Receptors, Cytokine/metabolism , Receptors, Epoprostenol , Receptors, OSM-LIF , Receptors, Prostaglandin/agonists , Transcription Factors/genetics , Transcription Factors/immunology , Transfection
17.
Biochem Biophys Res Commun ; 287(4): 962-8, 2001 Oct 05.
Article in English | MEDLINE | ID: mdl-11573959

ABSTRACT

Diabetes mellitus accelerating atherosclerosis was associated with the enhanced glycoxidative modification of lipoproteins. LOX-1, the endothelial oxidized LDL receptor might be involved in the pathogenesis of diabetic atherosclerosis. In this study, we examined the vascular expression of LOX-1 in streptozotocin-induced diabetic rats. We found that LOX-1 was significantly increased in diabetic rat aorta compared with nondiabetic control. Immunohistochemistry revealed that the most distinctive staining of LOX-1 was in the endothelial cells, especially in the bifurcations of artery branches from aorta. In cultured aortic endothelial cells, diabetic rat serum and advanced glycation endproducts-BSA induced LOX-1 expression, while control rat serum along with high glucose did not. Applying a competitive inhibition assay, we found that LOX-1 ligand activity was accumulated in the diabetic rat serum, mainly in VLDL/LDL fractions. In addition, VLDL/LDL prominently increased LOX-1 among all the lipoprotein fractions of diabetic rat serum. In conclusion, diabetes markedly upregulated LOX-1 expression in the aortic endothelial cells. The enhanced glycoxidative modification of lipoproteins may contribute to the underlying mechanisms.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Endothelium, Vascular/metabolism , Glycation End Products, Advanced/metabolism , Receptors, LDL/metabolism , Animals , Aorta/cytology , Aorta/metabolism , Arteries/cytology , Arteries/metabolism , Cells, Cultured , Ligands , Male , Rats , Rats, Sprague-Dawley , Receptors, LDL/genetics , Receptors, Oxidized LDL , Scavenger Receptors, Class E
18.
Circulation ; 104(10): 1176-80, 2001 Sep 04.
Article in English | MEDLINE | ID: mdl-11535576

ABSTRACT

BACKGROUND: Among the prostanoids, thromboxane (TX) A(2) is a potent stimulator of platelets, whereas prostaglandin (PG) I(2) inhibits their activation. The roles of PGE(2) in the regulation of platelet function have not been established, however, and the contribution of PGE(2) in hemostasis and thromboembolism is poorly understood. The present study was intended to clarify these roles of PGE(2) by using mice lacking the PGE(2) receptor subtype 3 (EP(3)(-/-) mice). METHODS AND RESULTS: Expression of mRNAs for EP(3) in murine platelets was confirmed by quantitative reverse transcription-polymerase chain reaction. PGE(2) and AE-248, a selective EP(3) agonist, showed concentration-dependent potentiation of platelet aggregation induced by U46619, a TXA(2) receptor agonist, although PGE(2) alone could not induce aggregation. PGE(2) and AE-248 increased cytosolic calcium ion concentration ([Ca(2+)](i)), and AE-248 inhibited the forskolin-induced increase in cytosolic cAMP concentration ([cAMP](i)), suggesting G(i) coupling of EP(3). The potentiating effects of PGE(2) and AE-248 on platelet aggregation along with their effects on [Ca(2+)](i) and [cAMP](i) were absent in EP(3)(-/-) mice. In vivo, the bleeding time was significantly prolonged in EP(3)(-/-) mice. Moreover, when mice were challenged intravenously with arachidonic acid, mortality and thrombus formation in the lung were significantly reduced in EP(3)(-/-) mice. CONCLUSIONS: - PGE(2) potentiated platelet aggregation induced by U46619 via EP(3) by increasing [Ca(2+)](i), decreasing [cAMP](i), or both. This potentiating action of PGE(2) via EP(3) is essential in mediating both physiological and pathological effects of PGE(2) in vivo.


Subject(s)
Hemorrhage/physiopathology , Receptors, Prostaglandin E/physiology , Thromboembolism/prevention & control , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Animals , Blood Platelets/metabolism , Dinoprostone/analogs & derivatives , Dinoprostone/pharmacology , Disease Susceptibility , Dose-Response Relationship, Drug , Drug Synergism , Female , Gene Expression , Male , Mice , Mice, Mutant Strains , Platelet Aggregation/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Prostaglandin E/agonists , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E, EP3 Subtype , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Thromboembolism/genetics
19.
J Biol Chem ; 276(45): 42527-33, 2001 Nov 09.
Article in English | MEDLINE | ID: mdl-11546796

ABSTRACT

Expression of the constitutively active mutant of Galpha(11) (Galpha(11)QL) induces the formation of vinculin-containing focal adhesion-like structures in HeLa cells. This was found to be inhibited by Y-27632, a specific inhibitor of Rho-associated kinases (ROCK), but not by co-expression with a dominant negative mutant of RhoA, suggesting Rho-independent activation of ROCK by Galpha(11)QL. Investigation of trypan blue exclusion and immunocytochemistry with an antibody against cleaved caspase revealed the cellular phenotype of Galpha(11)QL-expressing cells to be identical to that displayed by cells undergoing apoptosis, and the caspase inhibitor zVAD-fmk blocked all morphological changes induced by Galpha(11)QL. Transfection of Galpha(11)QL induced cleavage of ROCK-I, and this proteolysis was also prevented by zVAD-fmk. ROCK-I C-terminally truncated at its authentic caspase sites also induced the formation of vinculin-containing focal adhesion-like structures. In addition, cleavage of ROCK-I was observed when cells overexpressing m1 muscarinic acetylcholine receptors were stimulated with carbachol. These results suggest that Galpha(11) induces proteolytic activation of ROCK-I by caspase and thereby regulates the actin cytoskeleton during apoptosis.


Subject(s)
Caspases/physiology , Heterotrimeric GTP-Binding Proteins/physiology , Protein Serine-Threonine Kinases/metabolism , Calcium/metabolism , Enzyme Activation , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins , Protein Kinase C/physiology , rho-Associated Kinases
20.
Nat Med ; 7(9): 1048-51, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11533709

ABSTRACT

Arachidonic acid is metabolized to prostaglandin H(2) (PGH(2)) by cyclooxygenase (COX). COX-2, the inducible COX isozyme, has a key role in intestinal polyposis. Among the metabolites of PGH(2), PGE(2) is implicated in tumorigenesis because its level is markedly elevated in tissues of intestinal adenoma and colon cancer. Here we show that homozygous deletion of the gene encoding a cell-surface receptor of PGE(2), EP2, causes decreases in number and size of intestinal polyps in Apc(Delta 716) mice (a mouse model for human familial adenomatous polyposis). This effect is similar to that of COX-2 gene disruption. We also show that COX-2 expression is boosted by PGE(2) through the EP2 receptor via a positive feedback loop. Homozygous gene knockout for other PGE(2) receptors, EP1 or EP3, did not affect intestinal polyp formation in Apc(Delta 716) mice. We conclude that EP2 is the major receptor mediating the PGE2 signal generated by COX-2 upregulation in intestinal polyposis, and that increased cellular cAMP stimulates expression of more COX-2 and vascular endothelial growth factor in the polyp stroma.


Subject(s)
Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/physiopathology , Isoenzymes/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Receptors, Prostaglandin E/genetics , Adenoma/genetics , Adenoma/pathology , Adenomatous Polyposis Coli/pathology , Animals , Cyclooxygenase 2 , Dinoprostone/metabolism , Endothelial Growth Factors/genetics , Homozygote , Isoenzymes/genetics , Lymphokines/genetics , Mice , Mice, Knockout , Prostaglandin-Endoperoxide Synthases/genetics , Receptors, Prostaglandin/genetics , Receptors, Prostaglandin/metabolism , Receptors, Prostaglandin E/metabolism , Receptors, Prostaglandin E, EP1 Subtype , Receptors, Prostaglandin E, EP2 Subtype , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...