Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 126
Filter
1.
Cells ; 11(11)2022 06 02.
Article in English | MEDLINE | ID: mdl-35681514

ABSTRACT

Cyclooxygenase-2 (COX-2) plays a critical role in regulating innate immunity and metabolism by producing prostaglandins (PGs) and other lipid mediators. However, the implication of adipose COX-2 in obesity remains largely unknown. Using adipocyte-specific COX-2 knockout (KO) mice, we showed that depleting COX-2 in adipocytes promoted white adipose tissue development accompanied with increased size and number of adipocytes and predisposed diet-induced adiposity, obesity, and insulin resistance. The increased size and number of adipocytes by COX-2 KO were reversed by the treatment of prostaglandin E2 (PGE2) but not PGI2 and PGD2 during adipocyte differentiation. PGE2 suppresses PPARγ expression through the PKA pathway at the early phase of adipogenesis, and treatment of PGE2 or PKA activator isoproterenol diminished the increased lipid droplets in size and number in COX-2 KO primary adipocytes. Administration of PGE2 attenuated increased fat mass and fat percentage in COX-2 deficient mice. Taken together, our study demonstrated the suppressing effect of adipocyte COX-2 on adipogenesis and reveals that COX-2 restrains adipose tissue expansion via the PGE2-mediated paracrine mechanism and prevents the development of obesity and related metabolic disorders.


Subject(s)
Adipogenesis , Cyclooxygenase 2 , Dinoprostone , Obesity , Animals , Cyclooxygenase 2/deficiency , Cyclooxygenase 2/genetics , Diet, High-Fat/adverse effects , Dinoprostone/metabolism , Mice , Mice, Knockout , Obesity/genetics
2.
Am J Physiol Regul Integr Comp Physiol ; 322(2): R126-R135, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34984926

ABSTRACT

Many lung diseases are caused by an excessive inflammatory response, and inflammatory lung diseases are often modeled using lipopolysaccharide (LPS) in mice. Cyclooxygenase-2 (COX-2) encoded by the Ptgs2 gene is induced in response to inflammatory stimuli including LPS. The objective of this study was to test the hypothesis that mice deficient in COX-2 (Ptgs2-/-) will be protected from LPS-induced lung injury. Wild-type (WT; CD1 mice) and Ptgs2-/- mice (on a CD1 background) were treated with LPS or vehicle for 24 h. LPS treatment resulted in histological evidence of lung injury, which was attenuated in the Ptgs2-/- mice. LPS treatment increased the mRNA levels for tumor necrosis factor-α, interleukin-10, and monocyte chemoattractant protein-1 in the lungs of WT mice, and the LPS-induced increases in these levels were attenuated in the Ptgs2-/- mice. The protein levels of active caspase-3 and caspase-9 were lower in the LPS-treated lungs of Ptgs2-/- mice than in LPS-treated WT mice, as were the number of terminal deoxynucleotide transferase dUTP nick end labeling-positive cells in lung sections. LPS exposure resulted in a greater lung wet-to-dry weight ratio (W/D) in WT mice, suggestive of pulmonary edema, while in LPS-treated Ptgs2-/- mice, the W/D was not different from controls and less than in LPS-treated WT mice. These results demonstrate that COX-2 is involved in the inflammatory response to LPS and suggest that COX-2 not only acts as a downstream participant in the inflammatory response, but also acts as a regulator of the inflammatory response likely through a feed-forward mechanism following LPS stimulation.


Subject(s)
Acute Lung Injury/prevention & control , Apoptosis , Cyclooxygenase 2/deficiency , Lung/enzymology , Pneumonia/prevention & control , Acute Lung Injury/chemically induced , Acute Lung Injury/enzymology , Acute Lung Injury/pathology , Animals , Caspase 3/metabolism , Caspase 9/metabolism , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Cyclooxygenase 2/genetics , Disease Models, Animal , Female , Interleukin-10/genetics , Interleukin-10/metabolism , Lipopolysaccharides , Lung/pathology , Male , Mice, Knockout , Pneumonia/chemically induced , Pneumonia/enzymology , Pneumonia/pathology , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
3.
Gastroenterology ; 158(8): 2195-2207.e6, 2020 06.
Article in English | MEDLINE | ID: mdl-32084424

ABSTRACT

BACKGROUND AND AIMS: Visceral hypersensitivity is common in patients with irritable bowel syndrome (IBS). We investigated whether inflammatory molecules, such as histamine and proteases, activate prostaglandin-endoperoxide synthase 2 (also called COX2) to increase the synthesis of prostaglandin E2 (PGE2) by mast cells, which activates the receptor PTGER2 (also called EP2) in the dorsal root ganglia to promote visceral hypersensitivity. METHODS: We used an enzyme-linked immunosorbent assay to measure levels of spontaneous release of molecules from mast cells in colonic mucosa from patients with IBS with diarrhea (IBS-D; 18 women and 5 men; aged 28-60 years), healthy individuals (controls, n = 24), mice, and rats. We measured visceromotor responses to colorectal distension in rodents after intracolonic administration of colon biopsy supernatants, histamine, PGE2, a small interfering RNA against EP2, or an agonist of F2R like trypsin receptor 1 (F2RL1, also called protease-activated receptor 2 [PAR2]). We investigated the role of COX2, produced by mast cells, in mediation of visceral hypersensitivity using mice with the Y385F substitution in Ptgs2 (Ptgs2Y385F mice), mast cell-deficient (W/WV) mice, and W/WV mice given injections of mast cells derived from wild-type or Ptgs2Y385F mice. RESULTS: Colon biopsies from patients with IBS-D had increased levels of PGE2, based on enzyme-linked immunosorbent assay, and COX2 messenger RNA and protein, compared with control biopsies. Immunohistochemistry showed that most of the COX2 was in mast cells. Intracolonic infusions of rats with IBS-D biopsy supernatants generated a 3- to 4-fold increase in visceromotor responses to colorectal distension; this was associated with significant increases in PGE2, histamine, and tryptase in the colonic mucosa. These increases were prevented by a mast cell stabilizer, COX2 inhibitor, or knockdown of EP2. Intracolonic administration of supernatants from biopsies of patients with IBS-D failed to induce visceral hypersensitivity or increase the level of PGE2 in W/WV and Ptgs2Y385Fmice. Reconstitution of mast cells in W/WV mice restored the visceral hypersensitivity response. CONCLUSIONS: Abnormal synthesis of PGE2 by colonic mast cells appears to induce visceral hypersensitivity in patients with IBS-D.


Subject(s)
Colon/metabolism , Dinoprostone/metabolism , Intestinal Mucosa/metabolism , Irritable Bowel Syndrome/complications , Mast Cells/metabolism , Tissue Extracts/metabolism , Abdominal Pain/etiology , Abdominal Pain/metabolism , Abdominal Pain/physiopathology , Adult , Animals , Case-Control Studies , Cells, Cultured , Colon/innervation , Cyclooxygenase 2/deficiency , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Diarrhea/etiology , Diarrhea/metabolism , Diarrhea/physiopathology , Female , Humans , Hyperalgesia/etiology , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Intestinal Mucosa/innervation , Irritable Bowel Syndrome/metabolism , Irritable Bowel Syndrome/physiopathology , Male , Mast Cells/pathology , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Rats, Wistar , Sensory Receptor Cells/metabolism , Tissue Extracts/administration & dosage
4.
Genes Brain Behav ; 18(1): e12506, 2019 01.
Article in English | MEDLINE | ID: mdl-30027581

ABSTRACT

Prostaglandin E2 (PGE2) is an endogenous lipid molecule involved in normal brain development. Cyclooxygenase-2 (COX2) is the main regulator of PGE2 synthesis. Emerging clinical and molecular research provides compelling evidence that abnormal COX2/PGE2 signaling is associated with autism spectrum disorder (ASD). We previously found that COX2 knockout mice had dysregulated expression of many ASD genes belonging to important biological pathways for neurodevelopment. The present study is the first to show the connection between irregular COX2/PGE2 signaling and autism-related behaviors in male and female COX2-deficient knockin, (COX)-2- , mice at young (4-6 weeks) or adult (8-11 weeks) ages. Autism-related behaviors were prominent in male (COX)-2- mice for most behavioral tests. In the open field test, (COX)-2- mice traveled more than controls and adult male (COX)-2- mice spent less time in the center indicating elevated hyperactive and anxiety-linked behaviors. (COX)-2- mice also buried more marbles, with males burying more than females, suggesting increased anxiety and repetitive behaviors. Young male (COX)-2- mice fell more frequently in the inverted screen test revealing motor deficits. The three-chamber sociability test found that adult female (COX)-2- mice spent less time in the novel mouse chamber indicative of social abnormalities. In addition, male (COX)-2- mice showed altered expression of several autism-linked genes: Wnt2, Glo1, Grm5 and Mmp9. Overall, our findings offer new insight into the involvement of disrupted COX2/PGE2 signaling in ASD pathology with age-related differences and greater impact on males. We propose that (COX)-2- mice might serve as a novel model system to study specific types of autism.


Subject(s)
Autistic Disorder/genetics , Behavior, Animal , Cyclooxygenase 2/genetics , Animals , Autistic Disorder/physiopathology , Cyclooxygenase 2/deficiency , Dinoprostone/metabolism , Disease Models, Animal , Female , Lactoylglutathione Lyase/genetics , Lactoylglutathione Lyase/metabolism , Male , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Receptor, Metabotropic Glutamate 5/genetics , Receptor, Metabotropic Glutamate 5/metabolism , Wnt2 Protein/genetics , Wnt2 Protein/metabolism
5.
Physiol Res ; 67(5): 809-812, 2018 11 14.
Article in English | MEDLINE | ID: mdl-30044110

ABSTRACT

We investigated and evaluated post-irradiation survival in cyclooxygenase-2-deficient (COX-2 KO) mice. Thirty-day survival following exposure of COX-2 KO mice to a lethal dose of 8.5 Gy of gamma-rays was observed to be statistically significantly lower in both males and females, as well as when the sexes were merged, in comparisons with their wild-type counterparts. These findings were related to the previous observations concerning the detrimental influence of the COX-2 genetic disruption on hematopoiesis in sublethally irradiated mice. Deteriorated post-irradiation survival of COX-2 KO mice confirmed the previously anticipated conclusion regarding negative influence of the antiinflammatory action of COX-2 deficiency under the conditions of exposure of the animals to ionizing radiation.


Subject(s)
Cyclooxygenase 2/deficiency , Cyclooxygenase 2/radiation effects , Gamma Rays/adverse effects , Animals , Female , Male , Mice , Mice, Knockout , Survival Rate/trends
6.
Genomics Proteomics Bioinformatics ; 15(6): 339-351, 2017 12.
Article in English | MEDLINE | ID: mdl-29247872

ABSTRACT

The differential expression of two closelyassociated cyclooxygenase isozymes, COX-1 and COX-2, exhibited functions beyond eicosanoid metabolism. We hypothesized that COX-1 or COX-2 knockout lung fibroblasts may display altered protein profiles which may allow us to further differentiate the functional roles of these isozymes at the molecular level. Proteomic analysis shows constitutive production of macrophage migration inhibitory factor (MIF) in lung fibroblasts derived from COX-2-/- but not wild-type (WT) or COX-1-/- mice. MIF was spontaneously released in high levels into the extracellular milieu of COX2-/- fibroblasts seemingly from the preformed intracellular stores, with no change in the basal gene expression of MIF. The secretion and regulation of MIF in COX-2-/- was "prostaglandin-independent." GO analysis showed that concurrent with upregulation of MIF, there is a significant surge in expression of genes related to fibroblast growth, FK506 binding proteins, and isomerase activity in COX-2-/- cells. Furthermore, COX-2-/- fibroblasts also exhibit a significant increase in transcriptional activity of various regulators, antagonists, and co-modulators of p53, as well as in the expression of oncogenes and related transcripts. Integrative Oncogenomics Cancer Browser (IntroGen) analysis shows downregulation of COX-2 and amplification of MIF and/or p53 activity during development of glioblastomas, ependymoma, and colon adenomas. These data indicate the functional role of the MIF-COX-p53 axis in inflammation and cancer at the genomic and proteomic levels in COX-2-ablated cells. This systematic analysis not only shows the proinflammatory state but also unveils a molecular signature of a pro-oncogenic state of COX-1 in COX-2 ablated cells.


Subject(s)
Cyclooxygenase 2/deficiency , Fibroblasts/metabolism , Intramolecular Oxidoreductases/metabolism , Lung/cytology , Macrophage Migration-Inhibitory Factors/metabolism , Proteomics/methods , Tumor Suppressor Protein p53/metabolism , Animals , Arachidonic Acid/pharmacology , Carcinogenesis/drug effects , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Line , Cyclooxygenase 1/deficiency , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Indomethacin/pharmacology , Interleukin-1beta/pharmacology , Intramolecular Oxidoreductases/genetics , Macrophage Migration-Inhibitory Factors/genetics , Membrane Proteins/deficiency , Membrane Proteins/metabolism , Mice, Inbred C57BL , Models, Biological , Oncogenes , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tumor Suppressor Protein p53/genetics , Up-Regulation/drug effects , Up-Regulation/genetics
7.
Proc Natl Acad Sci U S A ; 114(47): 12596-12601, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29109289

ABSTRACT

Mass spectrometry imaging (MSI) was used to elucidate host lipids involved in the inflammatory signaling pathway generated at the host-pathogen interface during a septic bacterial infection. Using Francisella novicida as a model organism, a bacterial lipid virulence factor (endotoxin) was imaged and identified along with host phospholipids involved in the splenic response in murine tissues. Here, we demonstrate detection and distribution of endotoxin in a lethal murine F. novicida infection model, in addition to determining the temporally and spatially resolved innate lipid inflammatory response in both 2D and 3D renderings using MSI. Further, we show that the cyclooxygenase-2-dependent lipid inflammatory pathway is responsible for lethality in F. novicida infection due to overproduction of proinflammatory effectors including prostaglandin E2. The results of this study emphasize that spatial determination of the host lipid components of the immune response is crucial to identifying novel strategies to effectively address highly pathogenic and lethal infections stemming from bacterial, fungal, and viral origins.


Subject(s)
Cyclooxygenase 2/immunology , Dinoprostone/immunology , Francisella/pathogenicity , Gram-Negative Bacterial Infections/immunology , Host-Pathogen Interactions , Spleen/immunology , Animals , Cyclooxygenase 2/deficiency , Cyclooxygenase 2/genetics , Dinoprostone/biosynthesis , Eicosanoids/immunology , Eicosanoids/metabolism , Endotoxins/biosynthesis , Endotoxins/toxicity , Female , Francisella/physiology , Gene Expression , Gram-Negative Bacterial Infections/metabolism , Gram-Negative Bacterial Infections/mortality , Gram-Negative Bacterial Infections/pathology , Immunity, Innate , Inflammation , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Imaging , Phospholipids/immunology , Phospholipids/metabolism , Signal Transduction , Spleen/metabolism , Spleen/pathology , Survival Analysis
8.
Am J Physiol Renal Physiol ; 313(2): F430-F439, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28490532

ABSTRACT

Enhanced expression of cyclooxygenase 2 (COX2) in podocytes contributes to glomerular injury in diabetic kidney disease, but some basal level of podocyte COX2 expression might be required to promote podocyte attachment and/or survival. To investigate the role of podocyte COX2 expression in diabetic kidney disease, we deleted COX2 specifically in podocytes in a mouse model of Type 1 diabetes mellitus (Akita mice). Podocyte-specific knockout (KO) of COX2 did not affect renal morphology or albuminuria in nondiabetic mice. Albuminuria was significantly increased in wild-type (WT) and KO Akita mice compared with nondiabetic controls, and the increase in albuminuria was significantly greater in KO Akita mice compared with WT Akita mice at both 16 and 20 wk of age. At the 20-wk time point, mesangial expansion was also increased in WT and KO Akita mice compared with nondiabetic animals, and these histologic abnormalities were not improved by KO of COX2. Tubular injury was seen only in diabetic mice, but there were no significant differences between groups. Thus, KO of COX2 enhanced albuminuria and did not improve the histopathologic features of diabetic kidney disease. These data suggest that 1) KO of COX2 in podocytes does not ameliorate diabetic kidney disease in Akita mice, and 2) some basal level of podocyte COX2 expression in podocytes is necessary to attenuate the adverse effects of diabetes on glomerular filtration barrier function.


Subject(s)
Albuminuria/enzymology , Cyclooxygenase 2/deficiency , Diabetic Nephropathies/enzymology , Podocytes/enzymology , Albuminuria/genetics , Albuminuria/pathology , Albuminuria/urine , Animals , Biomarkers/blood , Biomarkers/urine , Blood Glucose/metabolism , Blood Pressure , Cyclooxygenase 2/genetics , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Diabetic Nephropathies/urine , Disease Models, Animal , Eicosanoids/urine , Genetic Predisposition to Disease , Integrases/genetics , Intracellular Signaling Peptides and Proteins/genetics , Male , Membrane Proteins/genetics , Mice, 129 Strain , Mice, Knockout , Phenotype , Podocytes/ultrastructure , Promoter Regions, Genetic , Renin/metabolism , Severity of Illness Index
9.
Sci Rep ; 7(1): 1657, 2017 05 10.
Article in English | MEDLINE | ID: mdl-28490739

ABSTRACT

Ultraviolet (UV) radiation exposure induces immunosuppression, which contributes to the development of cutaneous malignancies. We investigated the effects of honokiol, a phytochemical found in plants of the genus Magnolia, on UVB-induced immunosuppression using contact hypersensitivity (CHS) as a model in C3H/HeN mice. Topical application of honokiol (0.5 and 1.0 mg/cm2 skin area) had a significant preventive effect on UVB-induced suppression of the CHS response. The inflammatory mediators, COX-2 and PGE2, played a key role in this effect, as indicated by honokiol inhibition of cyclooxygenase-2 (COX-2) expression and PGE2 production in the UVB-exposed skin. Honokiol application also inhibited UVB-induced DNA hypermethylation and its elevation of the levels of TET enzyme, which is responsible for DNA demethylation in UVB-exposed skin. This was consistent with the restoration of the CHS response in mice treated with the DNA demethylating agent, 5-aza-2'-deoxycytidine, after UVB exposure. There was no significant difference in the levels of inhibition of UVB-induced immunosuppression amongst mice that were treated topically with available anti-cancer drugs (imiquimod and 5-fluorouracil). This study is the first to show that honokiol has the ability to inhibit UVB-induced immunosuppression in preclinical model and, thus, has potential for use as a chemopreventive strategy for UVB radiation-induced malignancies.


Subject(s)
Biphenyl Compounds/pharmacology , DNA Methylation/genetics , Immunosuppression Therapy , Inflammation/genetics , Lignans/pharmacology , Skin/immunology , Skin/pathology , Ultraviolet Rays , Animals , Biphenyl Compounds/administration & dosage , Cyclooxygenase 2/deficiency , Cyclooxygenase 2/metabolism , Cyclooxygenase Inhibitors/pharmacology , Dermatitis, Contact/immunology , Female , Inflammation Mediators/metabolism , Lignans/administration & dosage , Methyltransferases/metabolism , Mice , Models, Biological , Protective Agents/pharmacology , Proto-Oncogene Proteins/metabolism , Skin/drug effects , Skin/radiation effects
10.
Thromb Haemost ; 117(8): 1486-1497, 2017 07 26.
Article in English | MEDLINE | ID: mdl-28536720

ABSTRACT

Deep-vein thrombosis (DVT) is a common condition that often leads to pulmonary thromboembolism (VTE) and death. The role of prostaglandin-endoperoxide synthase (PTGS)2 in arterial thrombosis has been well established, whereas its impact in venous thrombosis remains unclear. Here, we showed that PTGS2 deletion predisposes to venous thrombosis as suggested by greater clot firmness and clot elasticity, by higher plasma levels of functional fibrinogen, factor VIII and PAI-1 activity, and proved by bigger thrombi detected after inferior vena cava ligation (IVCL) compared to WT mice. PTGS2-/- thrombi have greater fibrin content, higher number of F4/80+, TF+ and ANXA2+ cells, and lower S100A10+ cells. Remarkably, monocyte depletion reduced thrombus size in mutant mice, suggesting an important role of PTGS2-/- monocytes in this experimental setting. Interestingly, PTGS2 deletion reduced membrane ANXA2, and total S100A10, promoted assembly of ANXA2/p50NF-kB complex and its nuclear accumulation, and induced TF in peritoneal macrophages, whereas ANXA2 silencing decreased dramatically TF. Finally, Carbaprostacyclin treatment prevented venous thrombus formation induced by IVCL in mutant mice, reduced the ANXA2 binding to p50NF-kB subunit and its nuclear trafficking, and decreased TF in PTGS2-/- macrophages. PTGS2 deletion, changing the natural distribution of ANXA2 in monocytes/macrophages, increases TF expression and activity predisposing to venous thrombosis. Interestingly, Carbaprostacyclin treatment, inhibiting nuclear ANXA2 trafficking, controls monocyte TF activity and prevents DVT occurrence. Our data are of help in elucidating the mechanisms by which PTGS2 inhibition increases DVT risk, and suggest a new role for ANXA2 in venous thrombosis.


Subject(s)
Annexin A2/metabolism , Blood Coagulation , Cyclooxygenase 2/deficiency , Gene Deletion , Monocytes/enzymology , Venous Thrombosis/enzymology , Animals , Annexin A2/genetics , Antigens, Differentiation/metabolism , Blood Coagulation/drug effects , Cyclooxygenase 2/genetics , Disease Models, Animal , Epoprostenol/analogs & derivatives , Epoprostenol/pharmacology , Factor VIII/metabolism , Fibrinogen/metabolism , Fibrinolytic Agents/pharmacology , Genetic Predisposition to Disease , Mice , Mice, Knockout , Monocytes/drug effects , NF-kappa B p50 Subunit/metabolism , Phenotype , Plasminogen Activator Inhibitor 1/blood , Platelet Aggregation , Protein Transport , RAW 264.7 Cells , S100 Proteins/metabolism , Thromboplastin/metabolism , Time Factors , Venous Thrombosis/blood , Venous Thrombosis/genetics , Venous Thrombosis/prevention & control
11.
Physiol Res ; 66(4): 673-676, 2017 09 22.
Article in English | MEDLINE | ID: mdl-28406698

ABSTRACT

We investigated hematopoiesis in untreated and ionizing radiation-exposed cyclooxygenase-2-deficient (COX-2 KO) mice. We performed a complex hematological analysis of 16 parameters in untreated COX-2 KO male mice or COX-2 KO male mice irradiated with the dose of 4 Gy of gamma-rays and their wildtype littermates. At baseline, hematopoiesis was increased in COX-2-deficient mice, but attenuated by irradation in COX-2-deficient mice compared to wildtype. To conclude, the anti-inflammatory action of the COX-2 genetic disruption plays a positive role in hematopoiesis under basal conditions but is detrimental following radiation exposure.


Subject(s)
Cyclooxygenase 2/deficiency , Cyclooxygenase 2/radiation effects , Hematopoiesis/physiology , Hematopoiesis/radiation effects , Radiation, Ionizing , Animals , Male , Mice , Mice, 129 Strain , Mice, Knockout
12.
Am J Physiol Renal Physiol ; 312(6): F1044-F1055, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28274925

ABSTRACT

Deficiency of cyclooxygenase-2 (COX-2) activity in the early postnatal period causes impairment of kidney development leading to kidney insufficiency. We hypothesize that impaired NaCl reabsorption during the first days of life is a substantial cause for nephrogenic defects observed in COX-2-/- mice and that salt supplementation corrects these defects. Daily injections of NaCl (0.8 mg·g-1·day-1) for the first 10 days after birth ameliorated impaired kidney development in COX-2-/- pups resulting in an increase in glomerular size and fewer immature superficial glomeruli. However, impaired renal subcortical growth was not corrected. Increasing renal tubular flow by volume load or injections of KCl did not relieve the renal histomorphological damage. Administration of torsemide and spironolactone also affected nephrogenesis resulting in diminished glomeruli and cortical thinning. Treatment of COX-2-/- pups with NaCl/DOCA caused a stronger mitigation of glomerular size and induced a slight but significant growth of cortical tissue mass. After birth, renal mRNA expression of NHE3, NKCC2, ROMK, NCCT, ENaC, and Na+/K+-ATPase increased relative to postnatal day 2 in wild-type mice. However, in COX-2-/- mice, a significantly lower expression was observed for NCCT, whereas NaCl/DOCA treatment significantly increased NHE3 and ROMK expression. Long-term effects of postnatal NaCl/DOCA injections indicate improved kidney function with normalization of pathologically enhanced creatinine and urea plasma levels; also, albumin excretion was observed. In summary, we present evidence that salt supplementation during the COX-2-dependent time frame of nephrogenesis partly reverses renal morphological defects in COX-2-/- mice and improves kidney function.


Subject(s)
Cyclooxygenase 2/deficiency , Kidney/drug effects , Sodium Chloride, Dietary/administration & dosage , Urogenital Abnormalities/drug therapy , Animals , Animals, Newborn , Cyclooxygenase 2/genetics , Desoxycorticosterone Acetate/administration & dosage , Disease Models, Animal , Epithelial Sodium Channels/genetics , Epithelial Sodium Channels/metabolism , Female , Gene Expression Regulation, Developmental , Genetic Predisposition to Disease , Kidney/abnormalities , Kidney/enzymology , Kidney/growth & development , Male , Mice, Inbred C57BL , Mice, Knockout , Mineralocorticoid Receptor Antagonists/pharmacology , Morphogenesis , Phenotype , Potassium Channels, Inwardly Rectifying/genetics , Potassium Channels, Inwardly Rectifying/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sodium Potassium Chloride Symporter Inhibitors/administration & dosage , Sodium-Hydrogen Exchanger 3 , Sodium-Hydrogen Exchangers/genetics , Sodium-Hydrogen Exchangers/metabolism , Sodium-Potassium-Exchanging ATPase/genetics , Sodium-Potassium-Exchanging ATPase/metabolism , Solute Carrier Family 12, Member 1/genetics , Solute Carrier Family 12, Member 1/metabolism , Solute Carrier Family 12, Member 3/genetics , Solute Carrier Family 12, Member 3/metabolism , Spironolactone/administration & dosage , Sulfonamides/administration & dosage , Torsemide , Urogenital Abnormalities/enzymology , Urogenital Abnormalities/genetics , Urogenital Abnormalities/physiopathology
13.
Kidney Int ; 91(4): 818-829, 2017 04.
Article in English | MEDLINE | ID: mdl-28040266

ABSTRACT

Deletion of cyclooxygenase-2 (COX-2) causes impairment of postnatal kidney development. Here we tested whether the renin angiotensin system contributes to COX-2-dependent nephrogenesis in mice after birth and whether a rescue of impaired renal development and function in COX-2-/- mice was achievable. Plasma renin concentration in mouse pups showed a birth peak and a second peak around day P8 during the first 10 days post birth. Administration of the angiotensin II receptor AT1 antagonist telmisartan from day P1 to P3 did not result in cortical damage. However, telmisartan treatment from day P3 to P8, the critical time frame of renal COX-2 expression, led to hypoplastic glomeruli, a thinned subcapsular cortex and maturational arrest of superficial glomeruli quite similar to that observed in COX-2-/- mice. In contrast, AT2 receptor antagonist PD123319 was without any effect on renal development. Inhibition of the renin angiotensin system by aliskiren and enalapril caused similar glomerular defects as telmisartan. Administration of the AT1 receptor agonist L162313 to COX-2-/- pups improved kidney growth, ameliorated renal defects, but had no beneficial effect on reduced cortical mass. L162313 rescued impaired renal function by reducing serum urea and creatinine and mitigated pathologic albumin excretion. Moreover, glomerulosclerosis in the kidneys of COX-2-/- mice was reduced. Thus, angiotensin II-AT1-receptor signaling is necessary for COX-2-dependent normal postnatal nephrogenesis and maturation.


Subject(s)
Angiotensin II/metabolism , Cyclooxygenase 2/metabolism , Nephrons/enzymology , Receptor, Angiotensin, Type 1/metabolism , Renin-Angiotensin System , Signal Transduction , Age Factors , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin II Type 2 Receptor Blockers/pharmacology , Animals , Animals, Newborn , Creatinine/blood , Cyclooxygenase 2/deficiency , Cyclooxygenase 2/genetics , Dinoprostone/metabolism , Dose-Response Relationship, Drug , Female , Genotype , Male , Mice, Inbred C57BL , Mice, Knockout , Nephrons/drug effects , Nephrons/growth & development , Nephrons/pathology , Phenotype , Receptor, Angiotensin, Type 1/drug effects , Receptor, Angiotensin, Type 2/drug effects , Receptor, Angiotensin, Type 2/metabolism , Renin/blood , Renin-Angiotensin System/drug effects , Signal Transduction/drug effects , Urea/blood
14.
Genomics Proteomics Bioinformatics ; 14(2): 81-93, 2016 04.
Article in English | MEDLINE | ID: mdl-27012456

ABSTRACT

The constitutively-expressed cyclooxygenase 1 (COX-1) and the inducible COX-2 are both involved in the conversion of arachidonic acid (AA) to prostaglandins (PGs). However, the functional roles of COX-1 at the cellular level remain unclear. We hypothesized that by comparing differential gene expression and eicosanoid metabolism in lung fibroblasts from wild-type (WT) mice and COX-2(-/-) or COX-1(-/-) mice may help address the functional roles of COX-1 in inflammation and other cellular functions. Compared to WT, the number of specifically-induced transcripts were altered descendingly as follows: COX-2(-/-)>COX-1(-/-)>WT+IL-1ß. COX-1(-/-) or COX-2(-/-) cells shared about 50% of the induced transcripts with WT cells treated with IL-1ß, respectively. An interactive "anti-inflammatory, proinflammatory, and redox-activated" signature in the protein-protein interactome map was observed in COX-2(-/-) cells. The augmented COX-1 mRNA (in COX-2(-/-) cells) was associated with the upregulation of mRNAs for glutathione S-transferase (GST), superoxide dismutase (SOD), NAD(P)H dehydrogenase quinone 1 (NQO1), aryl hydrocarbon receptor (AhR), peroxiredoxin, phospholipase, prostacyclin synthase, and prostaglandin E synthase, resulting in a significant increase in the levels of PGE2, PGD2, leukotriene B4 (LTB4), PGF1α, thromboxane B2 (TXB2), and PGF2α. The COX-1 plays a dominant role in shifting AA toward the LTB4 pathway and anti-inflammatory activities. Compared to WT, the upregulated COX-1 mRNA in COX-2(-/-) cells generated an "eicosanoid storm". The genomic characteristics of COX-2(-/-) is similar to that of proinflammatory cells as observed in IL-1ß induced WT cells. COX-1(-/-) and COX-2(-/-) cells exhibited compensation of various eicosanoids at the genomic and metabolic levels.


Subject(s)
Cyclooxygenase 1/metabolism , Eicosanoids/metabolism , Genomics , Metabolomics , Animals , Cells, Cultured , Cyclooxygenase 1/deficiency , Cyclooxygenase 1/genetics , Cyclooxygenase 2/deficiency , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression/drug effects , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Inflammation/metabolism , Inflammation/pathology , Interleukin-1beta/pharmacology , Mice , Mice, Knockout , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , Phospholipases A2/metabolism , Prostaglandins/metabolism , Protein Interaction Maps , Protein Isoforms/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism
15.
Br J Pharmacol ; 173(9): 1541-55, 2016 May.
Article in English | MEDLINE | ID: mdl-26856544

ABSTRACT

BACKGROUND AND PURPOSE: Prostanoids derived from COX-2 and EP receptors are involved in vascular remodelling in different cardiovascular pathologies. This study evaluates the contribution of COX-2 and EP1 receptors to vascular remodelling and function in hypertension. EXPERIMENTAL APPROACH: Spontaneously hypertensive rats (SHR) and angiotensin II (AngII)-infused (1.44 mg · kg(-1) · day(-1), 2 weeks) mice were treated with the COX-2 inhibitor celecoxib (25 mg · kg(-1) · day(-1) i.p) or with the EP1 receptor antagonist SC19220 (10 mg · kg(-1) · day(-1) i.p.). COX-2(-/-) mice with or without AngII infusion were also used. KEY RESULTS: Celecoxib and SC19220 treatment did not modify the altered lumen diameter and wall : lumen ratio in mesenteric resistance arteries from SHR-infused and/or AngII-infused animals. However, both treatments and COX-2 deficiency decreased the augmented vascular stiffness in vessels from hypertensive animals. This was accompanied by diminished vascular collagen deposition, normalization of altered elastin structure and decreased connective tissue growth factor and plasminogen activator inhibitor-1 gene expression. COX-2 deficiency and SC19220 treatment diminished the increased vasoconstrictor responses and endothelial dysfunction induced by AngII infusion. Hypertensive animals showed increased mPGES-1 expression and PGE2 production in vascular tissue, normalized by celecoxib. Celecoxib treatment also decreased AngII-induced macrophage infiltration and TNF-α expression. Macrophage conditioned media (MCM) increased COX-2 and collagen type I expression in vascular smooth muscle cells; the latter was reduced by celecoxib treatment. CONCLUSIONS AND IMPLICATIONS: COX-2 and EP1 receptors participate in the increased extracellular matrix deposition and vascular stiffness, the impaired vascular function and inflammation in hypertension. Targeting PGE2 receptors might have benefits in hypertension-associated vascular damage.


Subject(s)
Cyclooxygenase 2/metabolism , Dibenz(b,f)(1,4)oxazepine-10(11H)-carboxylic acid, 8-chloro-, 2-acetylhydrazide/pharmacology , Dinoprostone/metabolism , Hypertension/drug therapy , Receptors, Prostaglandin E, EP1 Subtype/metabolism , Vascular Stiffness/drug effects , Animals , Celecoxib/administration & dosage , Celecoxib/chemistry , Celecoxib/pharmacology , Cells, Cultured , Cyclooxygenase 2/deficiency , Cyclooxygenase 2 Inhibitors/pharmacology , Dibenz(b,f)(1,4)oxazepine-10(11H)-carboxylic acid, 8-chloro-, 2-acetylhydrazide/administration & dosage , Dibenz(b,f)(1,4)oxazepine-10(11H)-carboxylic acid, 8-chloro-, 2-acetylhydrazide/chemistry , Dose-Response Relationship, Drug , Humans , Hypertension/metabolism , Male , Mice , Rats , Rats, Inbred SHR , Rats, Wistar , Receptors, Prostaglandin E, EP1 Subtype/antagonists & inhibitors , Structure-Activity Relationship
16.
Arterioscler Thromb Vasc Biol ; 36(1): 108-21, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26543101

ABSTRACT

OBJECTIVE: Chronic administration of selective cyclooxygenase-2 (COX-2) inhibitors leads to an increased risk of adverse cardiovascular events, including myocardial infarction and stroke. Vascular smooth muscle cell (VSMC) calcification, a common complication of chronic kidney disease, is directly related to cardiovascular morbidity and mortality. Here, we tested whether specific COX-2 inhibition affects vascular calcification during chronic renal failure. APPROACH AND RESULTS: The COX-2-specific inhibitors NS398 and SC236 significantly increased high-phosphate (Pi)-induced VSMC calcification. Similarly, COX-2(-/-) VSMCs, COX-2(-/-) aortas rings treated with high Pi and adenine diet-induced COX-2(-/-) chronic renal failure mice displayed enhanced calcium deposition. Metabolomic analysis revealed the differential suppression of PGE2 production by COX-1- and COX-2-specific inhibitors in high-Pi-stimulated VSMCs, indicating the involvement of PGE2 during COX-2 inhibition-aggravated vascular calcification. Indeed, exogenous PGE2 reduced alkaline phosphatase activity, osteogenic transdifferentiation, apoptosis, and calcification of VSMCs. In accordance, downregulation of microsomal prostaglandin E synthase (mPGES)-1 in VSMCs, mPGES-1(-/-) aorta with high-Pi stimulation and mPGES-1(-/-) chronic renal failure mice resulted in enhanced vascular mineralization. Further applications of RNAi and specific antagonists for PGE2 receptors indicated EP4 may mediate PGE2-inhibited vascular calcification. CONCLUSIONS: Our data revealed the pivotal role of COX-2-mPGES-1-PGE2 axis in vascular calcification. The selective inhibition of COX-2 or mPGES-1 may increase the risk of calcification and subsequent adverse cardiovascular events during chronic renal failure.


Subject(s)
Aortic Diseases/prevention & control , Dinoprostone/metabolism , Intramolecular Oxidoreductases/metabolism , Kidney Failure, Chronic/enzymology , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Prostaglandin-Endoperoxide Synthases/metabolism , Vascular Calcification/prevention & control , Adenine , Animals , Aorta, Abdominal/enzymology , Aorta, Abdominal/pathology , Aorta, Thoracic/enzymology , Aorta, Thoracic/pathology , Aortic Diseases/chemically induced , Aortic Diseases/enzymology , Aortic Diseases/genetics , Calcitriol , Cells, Cultured , Cyclooxygenase 2/deficiency , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/toxicity , Disease Models, Animal , Humans , Intramolecular Oxidoreductases/deficiency , Intramolecular Oxidoreductases/genetics , Kidney Failure, Chronic/chemically induced , Kidney Failure, Chronic/genetics , Male , Metabolomics/methods , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Nephrectomy , Phosphates , Prostaglandin-E Synthases , Rats, Sprague-Dawley , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Signal Transduction , Time Factors , Vascular Calcification/chemically induced , Vascular Calcification/enzymology , Vascular Calcification/genetics
17.
Dev Biol ; 409(2): 473-88, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26586201

ABSTRACT

Hirschsprung Disease (HSCR) is a potentially deadly birth defect characterized by the absence of the enteric nervous system (ENS) in distal bowel. Although HSCR has clear genetic causes, no HSCR-associated mutation is 100% penetrant, suggesting gene-gene and gene-environment interactions determine HSCR occurrence. To test the hypothesis that certain medicines might alter HSCR risk we treated zebrafish with medications commonly used during early human pregnancy and discovered that ibuprofen caused HSCR-like absence of enteric neurons in distal bowel. Using fetal CF-1 mouse gut slice cultures, we found that ibuprofen treated enteric neural crest-derived cells (ENCDC) had reduced migration, fewer lamellipodia and lower levels of active RAC1/CDC42. Additionally, inhibiting ROCK, a RHOA effector and known RAC1 antagonist, reversed ibuprofen effects on migrating mouse ENCDC in culture. Ibuprofen also inhibited colonization of Ret+/- mouse bowel by ENCDC in vivo and dramatically reduced bowel colonization by chick ENCDC in culture. Interestingly, ibuprofen did not affect ENCDC migration until after at least three hours of exposure. Furthermore, mice deficient in Ptgs1 (COX 1) and Ptgs2 (COX 2) had normal bowel colonization by ENCDC and normal ENCDC migration in vitro suggesting COX-independent effects. Consistent with selective and strain specific effects on ENCDC, ibuprofen did not affect migration of gut mesenchymal cells, NIH3T3, or WT C57BL/6 ENCDC, and did not affect dorsal root ganglion cell precursor migration in zebrafish. Thus, ibuprofen inhibits ENCDC migration in vitro and bowel colonization by ENCDC in vivo in zebrafish, mouse and chick, but there are cell type and strain specific responses. These data raise concern that ibuprofen may increase Hirschsprung disease risk in some genetically susceptible children.


Subject(s)
Cell Movement/drug effects , Enteric Nervous System/cytology , Ibuprofen/pharmacology , Intestines/cytology , Neural Stem Cells/cytology , Actin Cytoskeleton/metabolism , Animals , Caspase 3/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Chickens , Cyclooxygenase 1/deficiency , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/deficiency , Cyclooxygenase 2/metabolism , Enzyme Activation/drug effects , Membrane Proteins/deficiency , Membrane Proteins/metabolism , Mesoderm/cytology , Mice , Models, Biological , NIH 3T3 Cells , Neural Stem Cells/drug effects , Neurons/cytology , Neurons/drug effects , Organ Culture Techniques , PPAR gamma/metabolism , Pseudopodia/drug effects , Pseudopodia/metabolism , Zebrafish , rac1 GTP-Binding Protein/metabolism , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism
18.
Oncogene ; 35(18): 2390-7, 2016 05 05.
Article in English | MEDLINE | ID: mdl-26257064

ABSTRACT

Adrenergic stimulation adversely affects tumor growth and metastasis, but the underlying mechanisms are not well understood. Here, we uncovered a novel mechanism by which catecholamines induce inflammation by increasing prostaglandin E2 (PGE2) levels in ovarian cancer cells. Metabolic changes in tumors isolated from patients with depression and mice subjected to restraint stress showed elevated PGE2 levels. Increased metabolites, PTGS2 and PTGES protein levels were found in Skov3-ip1 and HeyA8 cells treated with norepinephrine (NE), and these changes were shown to be mediated by ADRB2 receptor signaling. Silencing PTGS2 resulted in significantly decreased migration and invasion in ovarian cancer cells in the presence of NE and decreased tumor burden and metastasis in restraint stress orthotopic models. In human ovarian cancer samples, concurrent increased ADRB2, PTGS2 and PTGES expression was associated with reduced overall and progression-free patient survival. In conclusion, increased adrenergic stimulation results in increased PGE2 synthesis via ADRB2-Nf-kB-PTGS2 axis, which drives tumor growth and metastasis.


Subject(s)
Dinoprostone/biosynthesis , Norepinephrine/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Signal Transduction , Animals , Cell Line, Tumor , Cyclooxygenase 2/deficiency , Cyclooxygenase 2/genetics , Female , Gene Silencing , Humans , Mice , Neoplasm Metastasis , Prostaglandin-E Synthases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...