Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.340
Filter
1.
J Med Chem ; 66(14): 9313-9324, 2023 07 27.
Article in English | MEDLINE | ID: mdl-37458373

ABSTRACT

Cyclooxygenase-1 and -2 (COX1 and COX2) derived endogenous ligand prostaglandin-E2 (PGE2) triggers several physiological and pathological conditions. It mediates signaling through four G-protein coupled receptors, EP1, EP2, EP3, and EP4. Among these, EP2 is expressed throughout the body including the brain and uterus. The functional role of EP2 has been extensively studied using EP2 gene knockout mice, cellular models, and selective small molecule agonists and antagonists for this receptor. The efficacy data from in vitro and in vivo animal models indicate that EP2 receptor is a major proinflammatory mediator with deleterious functions in a variety of diseases suggesting a path forward for EP2 inhibitors as the next generation of selective anti-inflammatory and antiproliferative agents. Interestingly in certain diseases, EP2 action is beneficial; therefore, EP2 agonists seem to be clinically useful. Here, we highlight the strengths, weaknesses, opportunities, and potential threats (SWOT analysis) for targeting EP2 receptor for therapeutic development for a variety of unmet clinical needs.


Subject(s)
Dinoprostone , Receptors, Prostaglandin E , Animals , Mice , Receptors, Prostaglandin E/agonists , Receptors, Prostaglandin E/genetics , Dinoprostone/pharmacology , Dinoprostone/physiology , Cyclooxygenase 2 , Drug Discovery , Receptors, Prostaglandin E, EP2 Subtype , Receptors, Prostaglandin E, EP4 Subtype
2.
Mediators Inflamm ; 2021: 9087816, 2021.
Article in English | MEDLINE | ID: mdl-34867083

ABSTRACT

Prostaglandin E2 (PGE2) is a lipid mediator derived from the fatty acid arachidonic acid. As an essential inflammatory factor, PGE2 has a critical impact on immune regulation through the prostanoid E (EP) receptor pathway. T cells, including CD4+ and CD8+ T cell subsets, play crucial roles in the adaptive immune response. Previous studies have shown that PGE2 is involved in regulating CD4+ T cell differentiation and inflammatory cytokine production via the EP receptor pathway, thereby affecting the development of diseases mediated by CD4+ T cells. In this review, we summarize the signaling pathway of PGE2 and describe the relationship between PGE2 and T cell differentiation. Hence, this review may provide important evidence for immune therapies and may even promote the development of biomedicines.


Subject(s)
Dinoprostone/physiology , T-Lymphocytes/cytology , Cell Differentiation , Humans , Receptors, Prostaglandin E/physiology , Signal Transduction/physiology
3.
J BUON ; 26(4): 1219-1225, 2021.
Article in English | MEDLINE | ID: mdl-34564973

ABSTRACT

PURPOSE: To explore the effects of atorvastatin (ATST) on the proliferation and apoptosis of colon cancer cells through the cyclooxygenase-2 (COX-2)/prostaglandin E2 (PGE2)/ß-catenin pathway. METHODS: HCT116 cells were cultured and transfected, and they were treated with ATST at different concentrations for different time. The association between the expressions of COX-2 and PGE2 and the survival time of patients with colon cancer was analyzed via Kaplan-Meier survival analysis. Then the protein expressions of COX-2, ß-catenin and apoptosis-related molecules in HCT116 cells were determined using Western blotting, and the proliferation of HCT116 cells was detected via cell counting kit-8 (CCK-8) assay. RESULTS: There was a significant difference in the survival rate between HCT116 cells treated with 30 µM ATST and those treated with 0 µM ATST. The survival time was obviously longer in patients with low expressions of COX-2 and PGE2 than that those with high expressions of COX-2 and PGE2. Low expressions of COX-2 and PGE2 in colon cancer tissues indicate a longer survival time. Moreover, a positive correlation was found between HCT116 cell density and COX-2 level, HCT116 cell density and PGE2 level, and COX-2 and PGE2 levels. ATST could down-regulate COX-2 and ß-catenin, and knocking down COX-2 could lower ß-catenin. After treatment with ATST and ATST + anti-COX-2, the activity of cleaved caspase-9, caspase-3 and PARP was remarkably enhanced, suggesting that ATST and ATST + anti-COX-2 can promote apoptosis of HCT116 cells. It was found that ATST and ATST + anti-COX-2 could also inhibit the proliferation of HCT116 cells.


Subject(s)
Apoptosis/drug effects , Atorvastatin/pharmacology , Cell Proliferation/drug effects , Colonic Neoplasms/pathology , Cyclooxygenase 2/drug effects , Cyclooxygenase 2/physiology , Dinoprostone/physiology , beta Catenin/drug effects , beta Catenin/physiology , HCT116 Cells , Humans , Signal Transduction , Tumor Cells, Cultured
4.
Theranostics ; 11(18): 8836-8854, 2021.
Article in English | MEDLINE | ID: mdl-34522214

ABSTRACT

Tissue regeneration following injury from disease or medical treatment still represents a challenge in regeneration medicine. Prostaglandin E2 (PGE2), which involves diverse physiological processes via E-type prostanoid (EP) receptor family, favors the regeneration of various organ systems following injury for its capabilities such as activation of endogenous stem cells, immune regulation, and angiogenesis. Understanding how PGE2 modulates tissue regeneration and then exploring how to elevate the regenerative efficiency of PGE2 will provide key insights into the tissue repair and regeneration processes by PGE2. In this review, we summarized the application of PGE2 to guide the regeneration of different tissues, including skin, heart, liver, kidney, intestine, bone, skeletal muscle, and hematopoietic stem cell regeneration. Moreover, we introduced PGE2-based therapeutic strategies to accelerate the recovery of impaired tissue or organs, including 15-hydroxyprostaglandin dehydrogenase (15-PGDH) inhibitors boosting endogenous PGE2 levels and biomaterial scaffolds to control PGE2 release.


Subject(s)
Dinoprostone/physiology , Regeneration/physiology , Wound Healing/physiology , Animals , Dinoprostone/metabolism , Humans , Signal Transduction/drug effects
5.
PLoS One ; 16(8): e0255335, 2021.
Article in English | MEDLINE | ID: mdl-34347801

ABSTRACT

The SARS-CoV-2 coronavirus has led to a pandemic with millions of people affected. The present study finds that risk-factors for severe COVID-19 disease courses, i.e. male sex, older age and sedentary life style are associated with higher prostaglandin E2 (PGE2) serum levels in blood samples from unaffected subjects. In COVID-19 patients, PGE2 blood levels are markedly elevated and correlate positively with disease severity. SARS-CoV-2 induces PGE2 generation and secretion in infected lung epithelial cells by upregulating cyclo-oxygenase (COX)-2 and reducing the PG-degrading enzyme 15-hydroxyprostaglandin-dehydrogenase. Also living human precision cut lung slices (PCLS) infected with SARS-CoV-2 display upregulated COX-2. Regular exercise in aged individuals lowers PGE2 serum levels, which leads to increased Paired-Box-Protein-Pax-5 (PAX5) expression, a master regulator of B-cell survival, proliferation and differentiation also towards long lived memory B-cells, in human pre-B-cell lines. Moreover, PGE2 levels in serum of COVID-19 patients lowers the expression of PAX5 in human pre-B-cell lines. The PGE2 inhibitor Taxifolin reduces SARS-CoV-2-induced PGE2 production. In conclusion, SARS-CoV-2, male sex, old age, and sedentary life style increase PGE2 levels, which may reduce the early anti-viral defense as well as the development of immunity promoting severe disease courses and multiple infections. Regular exercise and Taxifolin treatment may reduce these risks and prevent severe disease courses.


Subject(s)
COVID-19/pathology , Dinoprostone/blood , Immunity , Adolescent , Adult , Animals , COVID-19/blood , COVID-19/immunology , Case-Control Studies , Cells, Cultured , Chlorocebus aethiops , Dinoprostone/pharmacology , Dinoprostone/physiology , Disease Progression , Female , Humans , Immunity/drug effects , Immunity/physiology , Male , Middle Aged , SARS-CoV-2/drug effects , SARS-CoV-2/physiology , Vero Cells , Young Adult
7.
Sci Rep ; 11(1): 4044, 2021 02 17.
Article in English | MEDLINE | ID: mdl-33597646

ABSTRACT

The trabecular meshwork (TM) constitutes the main pathway for aqueous humor drainage and is exposed to complex intraocular pressure fluctuations. The mechanism of homeostasis in which TM senses changes in intraocular pressure and leads to normal levels of outflow resistance is not yet well understood. Previous reports have shown that Piezo1, a mechanically-activated cation channel, is expressed in TM and isolated TM cells. Therefore, we tested hypothesis that Piezo1 may function in response to membrane tension and stretch in TM. In human trabecular meshwork (hTM) cells, PIEZO1 was showed to be abundantly expressed, and Piezo1 agonist Yoda1 and mechanical stretch caused a Piezo1-dependent Ca2+ influx and release of arachidonic acid and PGE2. Treatment with Yoda1 or PGE2 significantly inhibited hTM cell contraction. These results suggest that mechanical stretch stimuli in TM activates Piezo1 and subsequently regulates TM cell contraction by triggering Ca2+ influx and release of arachidonic acid and PGE2. Thus, Piezo1 could acts as a regulator of intraocular pressure (IOP) within the conventional outflow pathway and could be a novel therapeutic strategy to modulate IOP in glaucoma patients.


Subject(s)
Dinoprostone/metabolism , Ion Channels/metabolism , Trabecular Meshwork/metabolism , Aqueous Humor/metabolism , Biomechanical Phenomena/physiology , Calcium/metabolism , Calcium Channels/metabolism , Cells, Cultured , Dinoprostone/physiology , Female , Gene Expression/genetics , Glaucoma/metabolism , Homeostasis , Humans , Intraocular Pressure/physiology , Ion Channels/physiology , Male , Mechanoreceptors/metabolism , Mechanoreceptors/physiology , Middle Aged , Primary Cell Culture , Pyrazines/pharmacology , Thiadiazoles/pharmacology , Trabecular Meshwork/physiology
8.
Exp Eye Res ; 205: 108507, 2021 04.
Article in English | MEDLINE | ID: mdl-33609510

ABSTRACT

Proliferative retinopathies, such as proliferative diabetic retinopathy (PDR) and retinopathy of prematurity (ROP) are major causes of visual impairment and blindness in industrialized countries. Prostaglandin E2 (PGE2) is implicated in cellular proliferation and migration via E-prostanoid receptor (EP4R). The aim of this study was to investigate the role of PGE2/EP4R signaling in the promotion of retinal neovascularisation. In a streptozotocin (STZ)-induced diabetic model and an oxygen-induced retinopathy (OIR) model, rats received an intravitreal injection of PGE2, cay10598 (an EP4R agonist) or AH23848 (an EP4R antagonist). Optical coherence tomography, retinal histology and biochemical markers were assessed. Treatment with PGE2 or cay10598 accelerated pathological retinal angiogenesis in STZ and OIR-induced rat retina, which was ameliorated in rats pretreated with AH23848. Serum VEGF-A was upregulated in the PGE2-treated diabetic rats vs non-treated diabetic rats and significantly downregulated in AH23848-treated diabetic rats. PGE2 or cay10598 treatment also significantly accelerated endothelial tip-cell formation in new-born rat retina. In addition, AH23848 treatment attenuated PGE2-or cay10598-induced proliferation and migration by repressing the EGF receptor (EGFR)/Growth factor receptor bound protein 2-associated binder protein 1 (Gab1)/Akt/NF-κB/VEGF-A signaling network in human retinal microvascular endothelial cells (hRMECs). PGE2/EP4R signaling network is thus a potential therapeutic target for pathological intraocular angiogenesis.


Subject(s)
Dinoprostone/physiology , ErbB Receptors/metabolism , Phosphoproteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Retinal Neovascularization/physiopathology , Animals , Animals, Newborn , Biphenyl Compounds/pharmacology , Blotting, Western , Cell Movement/drug effects , Cell Proliferation/drug effects , Diabetes Mellitus, Experimental , Disease Models, Animal , Electrophoretic Mobility Shift Assay , Endothelium, Vascular/metabolism , Intravitreal Injections , Male , NF-kappa B/metabolism , Oxygen/toxicity , Phosphorylation , Pyrrolidinones/pharmacology , Rats, Sprague-Dawley , Receptors, Prostaglandin E, EP4 Subtype/agonists , Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors , Retinal Neovascularization/metabolism , Retinal Vessels/metabolism , Signal Transduction/physiology , Tetrazoles/pharmacology , Vascular Endothelial Growth Factor A/metabolism
9.
Blood ; 137(4): 500-512, 2021 01 28.
Article in English | MEDLINE | ID: mdl-33507291

ABSTRACT

Glucocorticoid (GC) resistance remains a clinical challenge in pediatric acute lymphoblastic leukemia where response to GC is a reliable prognostic indicator. To identify GC resistance pathways, we conducted a genome-wide, survival-based, short hairpin RNA screen in murine T-cell acute lymphoblastic leukemia (T-ALL) cells. Genes identified in the screen interfere with cyclic adenosine monophosphate (cAMP) signaling and are underexpressed in GC-resistant or relapsed ALL patients. Silencing of the cAMP-activating Gnas gene interfered with GC-induced gene expression, resulting in dexamethasone resistance in vitro and in vivo. We demonstrate that cAMP signaling synergizes with dexamethasone to enhance cell death in GC-resistant human T-ALL cells. We find the E prostanoid receptor 4 expressed in T-ALL samples and demonstrate that prostaglandin E2 (PGE2) increases intracellular cAMP, potentiates GC-induced gene expression, and sensitizes human T-ALL samples to dexamethasone in vitro and in vivo. These findings identify PGE2 as a target for GC resensitization in relapsed pediatric T-ALL.


Subject(s)
Cyclic AMP/physiology , Dexamethasone/pharmacology , Dinoprostone/pharmacology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Second Messenger Systems/drug effects , 1-Methyl-3-isobutylxanthine/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Child , Chromogranins/antagonists & inhibitors , Colforsin/pharmacology , Cyclic AMP/pharmacology , Dexamethasone/administration & dosage , Dinoprostone/administration & dosage , Dinoprostone/antagonists & inhibitors , Dinoprostone/physiology , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/physiology , Female , GTP-Binding Protein alpha Subunits, Gs/antagonists & inhibitors , GTP-Binding Protein alpha Subunits, Gs/deficiency , Gene Expression Regulation, Leukemic/drug effects , Humans , Male , Mice , Models, Animal , Molecular Targeted Therapy , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Radiation Chimera , Receptors, Glucocorticoid/biosynthesis , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/physiology , Receptors, Prostaglandin E, EP4 Subtype/biosynthesis , Receptors, Prostaglandin E, EP4 Subtype/genetics , Xenograft Model Antitumor Assays
10.
J Vasc Surg Venous Lymphat Disord ; 9(6): 1535-1544, 2021 11.
Article in English | MEDLINE | ID: mdl-33482378

ABSTRACT

OBJECTIVE: The vital pathogenesis of varicose veins includes remodeling of the extracellular matrix and decreased vascular tone. Prostaglandin E2 (PGE2), a small molecule substance and inflammatory medium that belongs to the arachidonic acid derivatives, has the capacity to influence the expression of metalloproteinase and the vascular tone of the venous wall. The purpose of the present study was to investigate the role of PGE2 in the development of varicose veins in lower limbs. METHODS: The collected venous specimens were analyzed using hematoxylin and eosin, Masson's trichrome, and immunohistochemical staining. Transforming growth factor (TGF)-ß1, PGE2, CD31, and α-smooth muscle actin antibody were used to detect the expression and distribution of these proteins. The effect of PGE2 on the proliferation, migration, and tube formation capacity of human umbilical vein endothelial cells (HUVECs) was detected in vitro. The effect of TGF-ß1 on the expression of PGE2 and matrix metalloproteinases (MMPs) was assessed using Western blotting. Quantitative reverse transcription polymerase chain reaction was used to evaluate the effect of PGE2 on the expression of nitric oxide synthase (NOS) and other genes. RESULTS: The expression of PGE2 and TGF-ß1 in varicose veins was upregulated in the media tunica and intima tunica, and a strong positive correlation was found between PGE2 and TGF-ß1 expression in both varicose veins (95% confidence interval, 0.5207-0.9582; R = 0.848; P = .0005) and normal veins (95% confidence interval, 0.2530-0.8532; R = 0.643; P = .003). PGE2 promoted the migration and tube formation ability of HUVECs. Moreover, PGE2 also upregulated the expression of MMP-1 and TGF-ß1 in HUVECs and increased the mRNA level of inducible NOS. CONCLUSIONS: PGE2 can affect the remodeling of the extracellular matrix and reduce the elasticity of the vascular walls by promoting the synthesis of TGF-ß1 and MMP-1. PGE2 can also reduce the tension of the great saphenous vein by promoting the expression of inducible NOS, thus aggravating the blood stasis.


Subject(s)
Dinoprostone/physiology , Lower Extremity/blood supply , Nitric Oxide Synthase Type II/physiology , Transforming Growth Factor beta1/physiology , Varicose Veins/etiology , Cohort Studies , Disease Progression , Female , Humans , Male , Middle Aged , Retrospective Studies , Signal Transduction
11.
J Cyst Fibros ; 20(4): 673-677, 2021 07.
Article in English | MEDLINE | ID: mdl-33250436

ABSTRACT

BACKGROUND: Exposure to particulate matter (PM) air pollution is associated with adverse health outcomes in children with cystic fibrosis (CF). Airway macrophages (AM) phagocytose and retain inhaled PM in vivo, and the area of carbon in AM reflects both inhaled PM dose and phagocytic function. Since airway prostaglandin-E2 (PGE2) is increased in CF, and PGE2 suppresses AM phagocytosis, we sought evidence for PGE2-mediated suppression of AM phagocytosis of inhaled carbonaceous PM in CF. METHODS: After informed consent, urine was obtained from 20 controls and 24 CF children. In the subgroup of older children, at least one induced sputum was done in 20 controls and 19 CF children. Urinary tetranor PGEM, the major metabolite of PGE2, and sputum PGE2 were measured by mass spectrometry. The area of carbon in AM was determined by image analysis. Exposure to PM was assessed by modelling and personal monitoring. The effect of either PGE2 or CF sputum supernatant on phagocytosis of diesel exhaust particle (DEP) by AM was assessed in vitro. Data were analysed by t-test. RESULTS: Both urinary tetranor PGEM (P<0.05), and sputum PGE2 (P<0.05) were increased in CF . Despite no difference in PM exposure between groups, the area of phagocytosed carbon by AM was decreased in children with CF (P<0.01). PGE2 suppressed phagocytosis of DEP by AM from both controls and CF (P<0.0001). CF sputum supernatant suppressed phagocytosis of DEP by AM (P<0.0001) in a PGE2-dependent manner. CONCLUSION: Increased PGE2 in the CF airway suppresses phagocytosis of inhaled PM by AM.


Subject(s)
Cystic Fibrosis , Dinoprostone/physiology , Macrophages/physiology , Particulate Matter , Phagocytosis , Child , Cystic Fibrosis/immunology , Cystic Fibrosis/urine , Female , Humans , Inhalation , Male , Particulate Matter/analysis , Particulate Matter/urine , Sputum/chemistry
12.
Int J Radiat Biol ; 97(2): 170-178, 2021.
Article in English | MEDLINE | ID: mdl-33164600

ABSTRACT

PURPOSE: Radioresistance is highly correlated with radiotherapy failure in clinical cancer treatment. In the current study, we sought to examine the efficacy of Celecoxib and Afatinib co-treatment as radiosensitizers in the management of non-small cell lung cancer (NSCLC) A549 cells. MATERIALS AND METHODS: Generally, A549 cells were cultured with the treatment of Celecoxib and/or Afatinib for 24 h. Then, the cells were exposed to irradiation at 2 Gy/min for 1 min. After the end of treatment, cell viability, clonogenic survival, apoptosis and Prostaglandin E2 (PGE2) Elisa assays were performed. Transcriptional levels of Cyclooxygenase-2 (COX-2) affected by Celecoxib and/or Afatinib were measured by RT-qPCR. Posttranscriptional level of epidermal growth factor receptor (EGFR)-related gene was measured by Western blotting analysis. RESULTS: Here, we, for the first time, reported that the co-treatment of Celecoxib and Afatinib regulates the resistance of NSCLC A549 cells to radiation. The co-treatment of Celecoxib and Afatinib sensitized radiotherapy through the radiation-induced loss of cell viability and colony formation, as well as apoptosis. Mechanistically, Celecoxib and Afatinib-treated cells showed the inhibition of COX-2 and EGFR expression, which may be responsible for the A549 cells' increased resistance to radiation. CONCLUSION: Our results suggested that Celecoxib and Afatinib regulate cell sensitivity to apoptosis, and thus modulate the resistance of NSCLC to radiation.


Subject(s)
Afatinib/administration & dosage , Carcinoma, Non-Small-Cell Lung/radiotherapy , Celecoxib/administration & dosage , Lung Neoplasms/radiotherapy , Radiation Tolerance/drug effects , A549 Cells , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/pathology , Cell Survival/drug effects , Cyclooxygenase 2/physiology , Dinoprostone/physiology , Drug Synergism , ErbB Receptors/physiology , Humans , Lung Neoplasms/pathology
13.
Bioessays ; 42(12): e2000198, 2020 12.
Article in English | MEDLINE | ID: mdl-33174637

ABSTRACT

The outbreak of a new, potentially fatal virus, SARS-COV-2, which started in December 2019 in Wuhan, China, and since developed into a pandemic has stimulated research for an effective treatment and vaccine. For this research to be successful, it is necessary to understand the pathology of the virus. So far, we know that this virus can harm different organs of the body. Although the exact mechanisms are still unknown, this phenomenon may result from the body's secretion of prostaglandin E2 (PGE2), which is involved in several inflammation and immunity pathways. Noticeably, the expression of this molecule can lead to a cytokine storm causing a variety of side effects. In this paper, we discuss those side effects in SARS-COV-2 infection separately to determine whether PGE2 is, indeed, an important causative factor. Lastly, we propose a mechanism by which PGE2 production increases in response to COVID-19 disease and suggest the possible direct relation between PGE2 levels and the severity of this disease. Also see the video abstract here: https://youtu.be/SnPFAcjxxKw.


Subject(s)
COVID-19/epidemiology , COVID-19/pathology , Dinoprostone/physiology , Pandemics , SARS-CoV-2/pathogenicity , Aging/physiology , Angiotensin-Converting Enzyme 2/metabolism , COVID-19/immunology , Dinoprostone/metabolism , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation/complications , Inflammation/epidemiology , Inflammation/pathology , Inflammation/virology , Male , Phenotype , SARS-CoV-2/immunology , Severity of Illness Index , Signal Transduction/immunology
14.
Life Sci Alliance ; 3(11)2020 11.
Article in English | MEDLINE | ID: mdl-32820026

ABSTRACT

Alveolar macrophages (AMs) are resident immune cells of the lung that are critical for host defense. AMs are capable of proliferative renewal, yet their numbers are known to decrease with aging and increase with cigarette smoking. The mechanism by which AM proliferation is physiologically restrained, and whether dysregulation of this brake contributes to altered AM numbers in pathologic circumstances, however, remains unknown. Mice of advanced age exhibited diminished basal AM numbers and contained elevated PGE2 levels in their bronchoalveolar lavage fluid (BALF) as compared with young mice. Exogenous PGE2 inhibited AM proliferation in an E prostanoid receptor 2 (EP2)-cyclic AMP-dependent manner. Furthermore, EP2 knockout (EP2 KO) mice exhibited elevated basal AM numbers, and their AMs resisted the ability of PGE2 and aged BALF to inhibit proliferation. In contrast, increased numbers of AMs in mice exposed to cigarette smoking were associated with reduced PGE2 levels in BALF and were further exaggerated in EP2 KO mice. Collectively, our findings demonstrate that PGE2 functions as a tunable brake on AM numbers under physiologic and pathophysiological conditions.


Subject(s)
Macrophages, Alveolar/metabolism , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Aging/physiology , Animals , Bronchoalveolar Lavage Fluid/immunology , Dinoprostone/metabolism , Dinoprostone/physiology , Female , Lung/immunology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Prostaglandin E, EP2 Subtype/genetics , Receptors, Prostaglandin E, EP2 Subtype/physiology , Smoking/adverse effects
15.
J Physiol Pharmacol ; 71(5)2020 10.
Article in English | MEDLINE | ID: mdl-33475092

ABSTRACT

Prenatal treatment with magnesium sulfate (MgSO4) has neuroprotective effects in very preterm infants but its use has been associated with an increased rate of patent ductus arteriosus (DA). MgSO4 is a vasodilator and thus may exert a direct relaxant effect in the DA. We aimed to investigate the vasoactive effects of MgSO4 in the DA using the chicken embryo as experimental model. DA rings from 15-d (E15), 17-d (E17) and 19-d (E19) chicken embryos (total incubation: 21-d) were mounted in a wire myograph for isometric tension recordings. Exposure of DA rings to 21% O2 induced a tonic contraction which was relaxed by MgSO4 (2.4 - 7.2 mmol L-1) in a concentration-dependent manner (mean maximal relaxation E19: 51.4%, SE 6.3; EC50: 3.5 mmol L-1, SE 0.7). The relaxation evoked by MgSO4 was not significantly different between E15, E17 and E19 DA and was not affected by removal of the endothelium or by the presence of the nitric oxide synthase inhibitor L-NAME, the soluble guanylate cyclase inhibitor ODQ, or the cyclooxygenase inhibitor indomethacin. In contrast, when the DA rings were incubated in Ca2+-free solution, or in the presence of inhibitors of L-type Ca2+ channels (nifedipine), or large-conductance Ca2+-activated K+ (BKCa) channels (iberiotoxin), MgSO4-induced relaxation was impaired. Preincubation of the DA rings with MgSO4 concentrations ranging from 0 to 6.0 mmol L-1 did not significantly affect O2-induced contraction that was only impaired by a concentration of 7.2 mmol L-1. In conclusion, MgSO4 induced endothelium-independent relaxation of chicken DA and this relaxation appeared to be mediated through stimulation of BKCa channels and blockade of transmembrane flux of extracellular Ca2+. However, O2-induced DA contraction was only impaired by suprapharmacological concentrations of MgSO4 (> 6.0 mmol L-1). Therefore, our data suggest that the higher incidence of patent DA in preterm infants exposed to MgSO4 is unlikely to be due to a direct pharmacological effect of the drug on the DA.


Subject(s)
Ductus Arteriosus/drug effects , Magnesium Sulfate/pharmacology , Animals , Calcium/metabolism , Chick Embryo , Dinoprostone/physiology , Ductus Arteriosus/physiology , Ductus Arteriosus, Patent/chemically induced , Nitric Oxide/physiology , Vasodilation/drug effects
16.
Biochem Biophys Res Commun ; 521(2): 347-352, 2020 01 08.
Article in English | MEDLINE | ID: mdl-31668810

ABSTRACT

We had previously reported a prostaglandin E synthase (bmPGES) in the silkworm Bombyx mori that catalyzes the isomerization of PGH2 to PGE2. The present study aimed to provide a genome-editing characterization of bmPGES in B. mori. Results showed bmPGES gene disruption to result in a reduced content of PGE2. The change affected the expression of chorion genes and egg formation in silkworms. Collectively, the results indicated that bmPGES could be involved in reproduction of B. mori. Therefore, this study provides insights into the physiological role of bmPGES and PGE2 in silkworms.


Subject(s)
Ovum/growth & development , Prostaglandin-E Synthases/physiology , Animals , Bombyx , Chorion , Dinoprostone/deficiency , Dinoprostone/physiology , Gene Editing , Reproduction
17.
J Endocrinol ; 244(1): 163-175, 2020 01.
Article in English | MEDLINE | ID: mdl-31600720

ABSTRACT

Changes in zebrafish testicular gene expression induced by follicle-stimulating hormone (Fsh) or anti-Mullerian hormone (Amh) suggested that Amh inhibition and Fsh stimulation of spermatogenesis involved up and downregulation, respectively, of prostaglandin (PG) signaling. We found that Sertoli cells contacting type A undifferentiated (Aund) and differentiating (Adiff) spermatogonia expressed a key enzyme of PG production (Ptgs2); previous work showed that Sertoli cells contacting Adiff and B spermatogonia and spermatocytes showed ptges3b expression, an enzyme catalyzing PGE2 production. In primary testis tissue cultures, PGE2, but not PGD2 or PGF2α, reduced the mitotic activity of Adiff and their development into B spermatogonia. Vice versa, inhibiting PG production increased the mitotic activity of Adiff and B spermatogonia. Studies with pharmacological PG receptor antagonists suggest that an Ep4 receptor mediates the inhibitory effects on the development of spermatogonia, and cell-sorting experiments indicated this receptor is expressed mainly by testicular somatic cells. Combined inhibition of PG and steroid production moreover reduced the mitotic activity of Aund spermatogonia and led to their partial depletion, suggesting that androgens (and/or other testicular steroids), supported by PGE2, otherwise prevent depletion of Aund. Androgens also decreased testicular PGE2 production, increased the transcript levels of the enzyme-catabolizing PGs and decreased PGE2 receptor ptger4b transcript levels. Also Fsh potentially reduced, independent of androgens, PGE2 production by decreasing ptges3b transcript levels. Taken together, our results indicate that PGE2, via Ep4 receptors, favors self-renewal in conjunction with androgens and, independent of Fsh and androgens, inhibits differentiating divisions of spermatogonia.


Subject(s)
Androgens/metabolism , Cell Differentiation/genetics , Dinoprostone/physiology , Follicle Stimulating Hormone/metabolism , Spermatogonia/metabolism , Animals , Cell Culture Techniques , Male , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Signal Transduction/genetics , Testis/cytology , Zebrafish
18.
Brain Res ; 1724: 146442, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31513790

ABSTRACT

Prostaglandin E2 (PGE2) and proton are typical inflammatory mediators. They play a major role in pain processing and hypersensitivity through activating their cognate receptors expressed in terminals of nociceptive sensory neurons. However, it remains unclear whether there is an interaction between PGE2 receptors and proton-activated acid-sensing ion channels (ASICs). Herein, we show that PGE2 enhanced the functional activity of ASICs in rat dorsal root ganglion (DRG) neurons through EP1 and EP4 receptors. In the present study, PGE2 concentration-dependently increased ASIC currents in DRG neurons. It shifted the proton concentration-response curve upwards, without change in the apparent affinity of proton for ASICs. Moreover, PGE2 enhancement of ASIC currents was partially blocked by EP1 or EP4 receptor antagonist. PGE2 failed to enhance ASIC currents when simultaneous blockade of both EP1 and EP4 receptors. PGE2 enhancement was partially suppressed after inhibition of intracellular PKC or PKA signaling, and completely disappeared after concurrent blockade of both PKC and PKA signaling. PGE2 increased significantly the expression levels of p-PKCε and p-PKA in DRG cells. PGE2 also enhanced proton-evoked action potentials in rat DRG neurons. Finally, peripherally administration of PGE2 dose-dependently exacerbated acid-induced nocifensive behaviors in rats through EP1 and EP4 receptors. Our results indicate that PGE2 enhanced the electrophysiological activity of ASICs in DRG neurons and contributed to acidosis-evoked pain, which revealed a novel peripheral mechanism underlying PGE2 involvement in hyperalgesia by sensitizing ASICs in primary sensory neurons.


Subject(s)
Acid Sensing Ion Channels/metabolism , Ganglia, Spinal/metabolism , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Acid Sensing Ion Channels/physiology , Action Potentials/drug effects , Animals , Dinoprostone/metabolism , Dinoprostone/physiology , Hyperalgesia/metabolism , Male , Neurons/metabolism , Nociceptors/metabolism , Pain/physiopathology , Pain Measurement/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Prostaglandin E, EP2 Subtype/physiology , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Sensory Receptor Cells/drug effects , Signal Transduction/drug effects
19.
Brain Res ; 1721: 146335, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31302096

ABSTRACT

Prevalence of prior stressful experience is linked to high incidence of chronic pain. Stress, particularly repeated stress, is known to induce maladaptive neuroplasticity along peripheral and central pain transmission pathways. These maladaptive neuroplastic events facilitate sensitization of nociceptive neurons and transition from acute to chronic pain. Pro-inflammatory and pain mediators are involved in inducing neuroplasticity. Pain mediators such as prostaglandin E2 (PGE2), EP4 receptor and transient receptor potential vanilloid-1 (TRPV1) contribute to the genesis of chronic pain. In this study, we examined the role of PGE2/EP4 signaling and TRPV1 signaling in repeated restraint stress-induced prolongation of sensitization pain, a model for transition from acute to chronic pain, in both in vivo and in vitro models. We found that pre-exposure to single restraint stress induced analgesia that masked sensitization pain evoked by subsequent PGE2 challenge. However, pre-exposure to 3d consecutive restraint stress not only prolonged sensitization pain, but also increased stress hormone corticosterone (CORT) in serum, COX2 levels in paw skin, and EP4 and TRPV1 levels in dorsal root ganglion (DRG) and paw skin. Pre-exposure to CORT for 3d, not 1d, also prolonged sensitization pain evoked by PGE2. Co-injection of glucocorticoid receptor (GR) antagonist RU486, COX2 inhibitor NS-398, EP4 receptor antagonist L161,982 or TRPV1 antagonist capsazepine prevented 3d restraint stress prolonged sensitization pain evoked by PGE2. In DRG cultures, CORT increased EP4 and TRPV1 protein levels through GR activation. These data suggest that PGE2/EP4 signaling and TRPV1 signaling in peripheral pain pathway contribute to repeated stress-predisposed transition from acute to chronic pain.


Subject(s)
Pain/metabolism , Receptors, Prostaglandin E, EP4 Subtype/metabolism , TRPV Cation Channels/metabolism , Animals , Dinoprostone/metabolism , Dinoprostone/physiology , Ganglia, Spinal/metabolism , Hyperalgesia/metabolism , Male , Neuralgia/metabolism , Nociceptors/metabolism , Pain Measurement , Rats , Rats, Sprague-Dawley , Receptors, Prostaglandin E, EP4 Subtype/physiology , Sensory Receptor Cells/drug effects , Stress, Psychological/metabolism , Stress, Psychological/physiopathology , TRPV Cation Channels/physiology
20.
Endocr J ; 66(8): 691-699, 2019 Aug 29.
Article in English | MEDLINE | ID: mdl-31105125

ABSTRACT

The cyclooxygenase2 (COX-2) enzyme catalyzes the first step of prostanoid biosynthesis, and is known for its crucial role in the pathogenesis of several inflammatory diseases including type 2 diabetes mellitus (T2DM). Although a variety of studies revealed that COX-2 played a role in the IL-1ß induced ß cell dysfunction, the molecular mechanism remains unclear. Here, using a cDNA microarray and in silico analysis, we demonstrated that inflammatory responses were upregulated in human T2DM islets compared with non-diabetic (ND) islets. COX-2 expression was significantly enhanced in human T2DM islets, correlated with the high inflammation level. PGE2, the catalytic product of COX-2, downregulated the functional gene expression of PDX1, NKX6.1, and MAFA and blunted the glucose induced insulin secretion of human islets. Conversely, inhibition of COX-2 activity by a pharmaceutical inhibitor prevented the ß-cell dysfunction induced by IL-1ß. COX-2 inhibitor also abrogated the IL-1ß autostimulation in ß cells, which further resulted in reduced COX-2 expression in ß cells. Together, our results revealed that COX-2/PGE2 signaling was involved in the regulation of IL-1ß autostimulation, thus forming an IL-1ß/COX-2/PGE2 pathway loop, which may result in the high inflammation level in human T2DM islets and the inflammatory impairment of ß cells. Breaking this IL-1ß/COX-2/PGE2 pathway loop provides a potential therapeutic strategy to improve ß cell function in the treatment of T2DM patients.


Subject(s)
Cyclooxygenase 2/physiology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Dinoprostone/physiology , Interleukin-1beta/physiology , Islets of Langerhans/physiopathology , Adult , Animals , Cells, Cultured , Diabetes Mellitus, Type 2/pathology , Dinoprostone/metabolism , Feedback, Physiological/physiology , Female , Humans , Inflammation/metabolism , Inflammation/physiopathology , Insulin-Secreting Cells/pathology , Insulin-Secreting Cells/physiology , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Male , Mice , Middle Aged , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...