Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
2.
Int Immunopharmacol ; 121: 110491, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37329807

ABSTRACT

15-deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2) exhibited potential to alleviate liver inflammation in chronic injury but was less studied in acute injury. Acute liver injury was associated with elevated macrophage migration inhibitory factor (MIF) levels in damaged hepatocytes. This study aimed to investigate the regulatory mechanism of hepatocyte-derived MIF by 15d-PGJ2 and its subsequent impact on acute liver injury. In vivo, mouse models were established by carbon tetrachloride (CCl4) intraperitoneal injection, with or without 15d-PGJ2 administration. 15d-PGJ2 treatment reduced the necrotic areas induced by CCl4. In the same mouse model constructed using enhanced green fluorescent protein (EGFP)-labeled bone marrow (BM) chimeric mice, 15d-PGJ2 reduced CCl4 induced BM-derived macrophage (BMM, EGFP+F4/80+) infiltration and inflammatory cytokine expression. Additionally, 15d-PGJ2 down-regulated liver and serum MIF levels; liver MIF expression was positively correlated with BMM percentage and inflammatory cytokine expression. In vitro, 15d-PGJ2 inhibited Mif expression in hepatocytes. In primary hepatocytes, reactive oxygen species inhibitor (NAC) showed no effect on MIF inhibition by 15d-PGJ2; PPARγ inhibitor (GW9662) abolished 15d-PGJ2 suppressed MIF expression and antagonists (troglitazone, ciglitazone) mimicked its function. In Pparg silenced AML12 cells, the suppression of MIF by 15d-PGJ2 was weakened; 15d-PGJ2 promoted PPARγ activation in AML 12 cells and primary hepatocytes. Furthermore, the conditioned medium of recombinant MIF- and lipopolysaccharide-treated AML12 respectively promoted BMM migration and inflammatory cytokine expression. Conditioned medium of 15d-PGJ2- or siMif-treated injured AML12 suppressed these effects. Collectively, 15d-PGJ2 activated PPARγ to suppress MIF expression in injured hepatocytes, reducing BMM infiltration and pro-inflammatory activation, ultimately alleviating acute liver injury.


Subject(s)
Chemical and Drug Induced Liver Injury , Macrophage Migration-Inhibitory Factors , Prostaglandin D2 , Animals , Mice , Culture Media, Conditioned , Hepatocytes , Liver , Macrophage Migration-Inhibitory Factors/metabolism , PPAR gamma , Prostaglandin D2/therapeutic use , Prostaglandin D2/pharmacology , Prostaglandins , Chemical and Drug Induced Liver Injury/drug therapy
3.
Transl Vis Sci Technol ; 12(5): 5, 2023 05 01.
Article in English | MEDLINE | ID: mdl-37133840

ABSTRACT

Purpose: The purpose of this study was to investigate the impact of prostaglandin D2 (PGD2) receptor 2 (DP2) on choroidal neovascularization (CNV) formation in mice. Methods: Using a laser-induced CNV model, the CNV size of wild-type (WT) mice treated with DP2 antagonist (CAY10471 or OC000459) was compared with that of untreated mice. Vascular endothelial growth factor (VEGF) and MCP-1 levels were also compared between the two groups. Similar experiments were performed comparing DP2 knockout (DP2KO) mice with WT mice (8 and 56 weeks old). The number of infiltrating macrophages to laser spots was also compared between the WT and DP2KO mice. We administered a DP2 antagonist to 15-methyl PGD2 (a DP2 agonist)-stimulated ARPE-19 cells and measured VEGF secretion by enzyme-linked immunosorbent assay. Tube formation assay was performed on human umbilical vein endothelial cells with or without a DP2 antagonist. Results: CNV sizes were significantly smaller in mice treated with CAY10471 or OC000459 than in those treated with vehicle. Similarly, the CNV size of DP2KO mice was significantly smaller than that of WT mice. The number of macrophages at laser spots in DP2KO mice was significantly lower than that in WT mice. The VEGF concentration of lasered DP2KO mice's eyes was significantly lower than that of lasered WT mice' eyes. DP2 antagonist treatment suppressed VEGF secretion in ARPE-19 cells under 15-methyl PGD2 stimulation. The tube formation assay suggested that lumen formation was inhibited by a DP2 antagonist. Conclusions: DP2 blockade attenuated choroidal neovascularization. Translational Relevance: Drugs targeting DP2 are potentially a novel treatment for age-related macular degeneration.


Subject(s)
Choroidal Neovascularization , Vascular Endothelial Growth Factor A , Mice , Humans , Animals , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/therapeutic use , Prostaglandin D2/pharmacology , Prostaglandin D2/therapeutic use , Choroidal Neovascularization/drug therapy , Lasers , Human Umbilical Vein Endothelial Cells/metabolism , Mice, Knockout
4.
Oxid Med Cell Longev ; 2022: 1571705, 2022.
Article in English | MEDLINE | ID: mdl-35437456

ABSTRACT

The pathogenesis of cerebral ischemia-reperfusion (I/R) injury is complex and does not exhibit an effective strategy. Maternal inflammation represents one of the most important factors involved in the etiology of brain injury in newborns. We aimed to investigate the effect of maternal inflammation on offspring susceptibility to cerebral I/R injury and the mechanisms by which it exerts its effects. Pregnant SD rats were intraperitoneally injected with LPS (300 µg/kg/day) at gestational days 11, 14, and 18. Pups were subjected to MCAO/R on postnatal day 60. Primary neurons were obtained from postnatal day 0 SD rats and subjected to OGD/R. Neurological deficits, brain injury, neuronal viability, neuronal damage, and neuronal apoptosis were assessed. Oxidative stress and inflammation were evaluated, and the expression levels of COX-2/PGD2/DP pathway-related proteins and apoptotic proteins were detected. Maternal LPS exposure significantly increased the levels of oxidative stress and inflammation, significantly activated the COX-2/PGD2/DP2 pathway, and increased proapoptotic protein expression. However, maternal LPS exposure significantly decreased the antiapoptotic protein expression, which subsequently increased neurological deficits and cerebral I/R injury in offspring rats. The corresponding results were observed in primary neurons. Moreover, these effects of maternal LPS exposure were reversed by a COX-2 inhibitor and DP1 agonist but exacerbated by a DP2 agonist. In conclusion, maternal inflammatory exposure may increase offspring susceptibility to cerebral I/R injury. Moreover, the underlying mechanism might be related to the activation of the COX-2/PGD2/DP2 pathway. These findings provide a theoretical foundation for the development of therapeutic drugs for cerebral I/R injury.


Subject(s)
Brain Injuries , Brain Ischemia , Reperfusion Injury , Animals , Apoptosis , Apoptosis Regulatory Proteins , Brain Ischemia/drug therapy , Cyclooxygenase 2/metabolism , Female , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Lipopolysaccharides/therapeutic use , Pregnancy , Prostaglandin D2/pharmacology , Prostaglandin D2/therapeutic use , Rats , Rats, Sprague-Dawley , Reperfusion Injury/pathology , Signal Transduction
5.
Med Hypotheses ; 143: 110080, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32683221

ABSTRACT

Coronaviruses including severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2, also known as 2019-nCoV especially in China) replicate and divide in host cells. During this they are partly hidden from the innate immune responses although inflammatory consequences of viral replication still occur. We propose that anti-inflammatory antiviral prostaglandins may not only restrict viral replication but also prevent inflammatory responses in the lungs and other vital organs that are known to be part of the immuno-pathogenesis of coronavirus disease-19 (COVID-19). The combination of anti-inflammatory antiviral prostaglandins with interferons may lead to the clearance of viruses inside growth-restricted infected cells. However, further experimental studies and clinical trials should be conducted to evaluate the safety and efficacy of these possible therapies.


Subject(s)
Betacoronavirus , Coronavirus Infections/drug therapy , Coronavirus Infections/immunology , Pneumonia, Viral/drug therapy , Pneumonia, Viral/immunology , Prostaglandin D2/therapeutic use , Animals , Anti-Inflammatory Agents/therapeutic use , Antiviral Agents/therapeutic use , Betacoronavirus/drug effects , Betacoronavirus/immunology , Betacoronavirus/pathogenicity , COVID-19 , Coronavirus Infections/etiology , Host Microbial Interactions/drug effects , Host Microbial Interactions/immunology , Humans , Models, Biological , Pandemics , Pneumonia, Viral/etiology , SARS-CoV-2 , Translational Research, Biomedical , COVID-19 Drug Treatment
6.
Arch Biochem Biophys ; 677: 108183, 2019 11 30.
Article in English | MEDLINE | ID: mdl-31704099

ABSTRACT

The prostaglandin D2 metabolite, 15-deoxy-Δ12,14-Prostaglandin J2 (15d-PGJ2), exerts an anti-inflammatory effect through peroxisome proliferator-activated receptor γ (PPARγ)-dependent and -independent anti-inflammatory actions. In the present study, we focused on heme oxygenase-1 (HO-1) induced by 15d-PGJ2, and evaluated the effects of enema treatment with 15d-PGJ2 in the development of intestinal inflammation using a murine colitis model. Acute colitis was induced with dextran sulfate sodium (DSS) in male C57BL/6 mice (8 weeks old) and NF-E2-related factor-2 (Nrf2) deficient mice. Mice were rectally administered 15d-PGJ2 (1 µM, 0.2 mL: 66.9 ng) daily during DSS administration. Intestinal expression of HO-1 mRNA and protein after rectal administration of 15d-PGJ2 was evaluated by real-time PCR and western blotting, respectively. A disease activity index (DAI) was determined on a daily basis for each animal, and consisted of a calculated score based on changes in body weight, stool consistency, and intestinal bleeding. Tissue-associated myeloperoxidase (MPO) activity as an index of neutrophil infiltration and mRNA expression levels of TNF-α, IFN-γ, and IL-17A were measured in the colonic mucosa. In addition, we evaluated the effects of co-treatment with a HO-1 inhibitor, zinc protoporphyrin IX (ZnPP), or a specific PPARγ antagonist, GW9662. As a result, rectal administration of 15d-PGJ2 markedly induced HO-1 protein and mRNA expression in the colonic mucosa. Treatment with 15d-PGJ2 ameliorated the increase in DAI score and MPO activity and the mRNA expression levels of TNF-α, IFN-γ, and IL-17A after DSS administration. These effects of 15d-PGJ2 against intestinal inflammation were negated by co-treatment with ZnPP, but not with GW9662. In Nrf2 deficient mice, the rectal administration of 15d-PGJ2 did not affect colonic HO-1 expression and activity of DSS-induced colitis. These results demonstrate that 15d-PGJ2 inhibits development of intestinal inflammation in mice via PPAR-independent and Nrf2-HO-1-dependent mechanisms.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Colitis/drug therapy , Heme Oxygenase-1/metabolism , Inflammation/drug therapy , Membrane Proteins/metabolism , Prostaglandin D2/analogs & derivatives , Administration, Rectal , Animals , Anti-Inflammatory Agents/administration & dosage , Colitis/chemically induced , Colon/cytology , Colon/pathology , Dextran Sulfate , Male , Mice, Inbred C57BL , NF-E2-Related Factor 2/metabolism , Prostaglandin D2/administration & dosage , Prostaglandin D2/therapeutic use
7.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1864(5): 609-618, 2019 05.
Article in English | MEDLINE | ID: mdl-30684679

ABSTRACT

Pain is one of the cardinal signs of inflammation and is present in many inflammatory conditions. Therefore, anti-inflammatory drugs such as NSAIDs also have analgesic properties. We previously showed that prostaglandin D2-glycerol ester (PGD2-G), endogenously produced by cyclooxygenase-2 from the endocannabinoid 2-arachidonoylglycerol, has anti-inflammatory effects in vitro and in vivo that are partly mediated by DP1 receptor activation. In this work, we investigated its effect in a model of carrageenan-induced inflammatory pain. PGD2-G decreased hyperalgesia and edema, leading to a faster recovery. Moreover, PGD2-G decreased carrageenan-induced inflammatory markers in the paw as well as inflammatory cell recruitment. The effects of PGD2-G were independent from metabolite formation (PGD2 and 15d-PGJ2-G) or DP1 receptor activation in this model. Indeed PGD2 delayed recovery from hyperalgesia while 15d-PGJ2-G worsened the edema. However, while PGD2-G decreased hyperalgesia in this model of inflammatory pain, it had no effect when tested in the capsaicin-induced pain model. While the targets mediating the effects of this bioactive lipid in inflammatory pain remain to be elucidated, our findings further support the interest of anti-inflammatory lipid mediators in the management of inflammatory pain.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Hyperalgesia/drug therapy , Inflammation/drug therapy , Prostaglandin D2/therapeutic use , Animals , Carrageenan , Esterification , Hyperalgesia/chemically induced , Inflammation/chemically induced , Male , Mice
8.
J Biol Chem ; 292(35): 14544-14555, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28684424

ABSTRACT

Macrophages use various cell-surface receptors to sense their environment and undergo polarized responses. The cytokines, interleukin (IL)-4 and IL-13, released from T-helper type 2 (Th2) cells, drive macrophage polarization toward an alternatively activated phenotype (M2). This phenotype is associated with the expression of potent pro-resolving mediators, such as the prostaglandin (PG) D2-derived cyclopentenone metabolite, 15d-PGJ2, produced by the cyclooxygenase (Ptgs; Cox) pathway. Interestingly, IL-4 treatment of bone marrow-derived macrophages (BMDMs) significantly down-regulates Cox-2 protein expression, whereas Cox-1 levels are significantly increased. This phenomenon not only challenges the dogma that Cox-1 is only developmentally regulated, but also demonstrates a novel mechanism in which IL-4-dependent regulation of Cox-1 involves the activation of the mechanistic target of rapamycin complex (mTORC). Using specific chemical inhibitors, we demonstrate here that IL-4-dependent Cox-1 up-regulation occurs at the post-transcriptional level via the Fes-Akt-mTORC axis. Activation of AMP-activated protein kinase (AMPK) by metformin, inhibition of mTORC by torin 1, or CRISPR/Cas9-mediated genetic knock-out of tuberous sclerosis complex-2 (Tsc2) blocked the IL-4-dependent expression of Cox-1 and the ability of macrophages to polarize to M2. However, use of 15d-PGJ2 partially rescued the effects of AMPK activation, suggesting the importance of Cox-1 in macrophage polarization as also observed in a model of gastrointestinal helminth clearance. In summary, these findings suggest a new paradigm where IL-4-dependent up-regulation of Cox-1 expression may play a key role in tissue homeostasis and wound healing during Th2-mediated immune responses, such as parasitic infections.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Interleukin-4/metabolism , Macrophage Activation , Macrophages/metabolism , Membrane Proteins/agonists , Models, Immunological , AMP-Activated Protein Kinases/chemistry , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cells, Cultured , Cyclooxygenase 1/genetics , Cyclooxygenase 1/metabolism , Enzyme Activation/drug effects , Enzyme Induction/drug effects , Enzyme Inhibitors/pharmacology , HEK293 Cells , Humans , Immunomodulation/drug effects , Interleukin-4/genetics , Ligands , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/immunology , Macrophages/pathology , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Metformin/pharmacology , Metformin/therapeutic use , Mice, Inbred C57BL , Nippostrongylus/drug effects , Nippostrongylus/growth & development , Nippostrongylus/immunology , Prostaglandin D2/analogs & derivatives , Prostaglandin D2/metabolism , Prostaglandin D2/therapeutic use , Recombinant Proteins/metabolism , Strongylida Infections/immunology , Strongylida Infections/metabolism , Strongylida Infections/pathology , Strongylida Infections/prevention & control
9.
Chin Med J (Engl) ; 130(3): 347-356, 2017 02 05.
Article in English | MEDLINE | ID: mdl-28139520

ABSTRACT

BACKGROUND: 15-Deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2), one of the major metabolites from prostaglandin D2 in arachidonic acid metabolic pathway, has potential anti-inflammatory properties. The objective of this study was to explore the effects of 15d-PGJ2-loaded poly(D,L-lactide-co-glycolide) nanocapsules (15d-PGJ2-NC) on inflammatory responses and bone regeneration in local bone defect. METHODS: The study was conducted on 96 Wistar rats from June 2014 to March 2016. Saline, unloaded nanoparticles, free 15d-PGJ2or 15d-PGJ2-NC, were delivered through a collagen vehicle inside surgically created transcortical defects in rat femurs. Interleukin-6 (IL-6), interleukin-1 beta (IL-1ß), and tumor necrosis factor-alpha (TNF-α) levels in the surrounding soft tissue were analyzed by Western blot and in the defect by quantitative real-time polymerase chain reaction over 14 days. Simultaneously, bone morphogenetic protein-6 (BMP-6) and platelet-derived growth factor-B (PDGF-B) messenger RNA (mRNA) in the defect were examined. New bone formation and EphrinB2 and osteoprotegerin (OPG) protein expression in the cortical defect were observed by Masson's Trichrome staining and immunohistochemistry over 28 days. Data were analyzed by one-way analysis of variance. Least-significant difference and Dunnett's T3 methods were used with a bilateral P< 0.05. RESULTS: Application of l5d-PGJ2-NC (100 µg/ml) in the local bone defect significantly decreased IL-6, IL-1ß, and TNF-α mRNA and protein, compared with saline-treated controls (P < 0.05). l5d-PGJ2-NC upregulated BMP-6 and PDGF-B mRNA (P < 0.05). New bone formation was observed in the cortical defect in l5d-PGJ2-NC-treated animals from 7th day onward (P < 0.001). Expression of EphrinB2 and OPG presented early on day 3 and persisted through day 28 in 15d-PGJ2-NC group (P < 0.05). CONCLUSION: Stable l5d-PGJ2-NC complexes were prepared that could attenuate IL-6, IL-1ß, and TNF-α expression, while increasing new bone formation and growth factors related to bone regeneration.


Subject(s)
Bone Regeneration/drug effects , Inflammation/drug therapy , Prostaglandin D2/analogs & derivatives , Animals , Bone Morphogenetic Protein 6/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Male , Platelet-Derived Growth Factor/metabolism , Prostaglandin D2/therapeutic use , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha/metabolism
10.
Acta Pharmacol Sin ; 38(5): 672-687, 2017 May.
Article in English | MEDLINE | ID: mdl-28216619

ABSTRACT

Hepatic ischemia-reperfusion (I/R) injury is a common clinical impairment that occurs in many circumstances and leads to poor prognosis. Both apoptosis and autophagy have been shown to contribute to cell death in hepatic I/R injury. 15-Deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2) is one of the best-studied anti-inflammatory prostaglandins, which has been verified to exert anti-inflammatory and cell-protective functions in various types of cells and animal models. In this study we explored the effects of 15d-PGJ2 on both apoptosis and autophagy in mouse hepatic I/R injury and its possible mechanisms. A model of segmental (70%) hepatic warm ischemia was established in Balb/c mice, and the pathological changes in serum and liver tissues were detected at 6, 12, and 24 h post-surgery, while 15d-PGJ2 (2.5, 7.5, 15 µg, iv) was administered 30 min prior the surgery. Pretreatment with 15d-PGJ2 (7.5, 15 µg) significantly ameliorated I/R-induced hepatic injury evidenced by dose-dependent reduction of serum ALT and AST levels as well as alleviated tissue damages. 15d-PGJ2 pretreatment significantly decreased the serum TNF-α and IL-1ß levels and the hepatic expression of F4/80, a major biomarker of macrophages. 15d-PGJ2 pretreatment upregulated the Bcl-2/Bax ratio, thus reducing the number of apoptotic cells in the livers. 15d-PGJ2 pretreatment considerably suppressed the expression of Beclin-1 and LC3, thus decreasing the number of autophagosomes in the livers. Furthermore, 15d-PGJ2 pretreatment activated Nrf2 and inhibited a ROS/HIF1α/BNIP3 pathway in the livers. Pretreatment with the PPARγ receptor blocker GW9662 (2 µg, ip) partly reversed the protective effects of 15d-PGJ2 on hepatic I/R injury. In conclusion, our results confirm the protective effect of 15d-PGJ2 on hepatic I/R injury, an effect that may rely on a reduction in the activation of Kupffer cells and on activation of the Nrf2 pathway, which lead to inhibition of ROS generation, apoptosis, and autophagy.


Subject(s)
Antioxidants/metabolism , Apoptosis/drug effects , Autophagy/drug effects , Liver Diseases/drug therapy , Prostaglandin D2/analogs & derivatives , Protective Agents/therapeutic use , Reperfusion Injury/drug therapy , Animals , Kupffer Cells/drug effects , Kupffer Cells/pathology , Liver/blood supply , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Diseases/metabolism , Liver Diseases/pathology , Male , Mice, Inbred BALB C , Prostaglandin D2/administration & dosage , Prostaglandin D2/therapeutic use , Protective Agents/administration & dosage , Reactive Oxygen Species/metabolism , Reperfusion Injury/metabolism , Reperfusion Injury/pathology
11.
Mediators Inflamm ; 2016: 9626427, 2016.
Article in English | MEDLINE | ID: mdl-27872515

ABSTRACT

The prostaglandin, 15-deoxy Δ12,14-prostaglandin J2 (15d-PGJ2), is a lipid mediator that plays an important role in the control of chronic inflammatory disease. However, the role of prostanoid in rheumatoid arthritis (RA) is not well determined. We demonstrated the therapeutic effect of 15d-PGJ2 in an experimental model of arthritis. Daily administration of 15d-PGJ2 attenuated the severity of CIA, reducing the clinical score, pain, and edema. 15d-PGJ2 treatment was associated with a marked reduction in joint levels of proinflammatory cytokines. Although the mRNA expression of ROR-γt was profoundly reduced, FOXP3 was enhanced in draining lymph node cells from 15d-PGJ2-treated arthritic mice. The specific and polyclonal CD4+ Th17 cell responses were limited during the addition of prostaglandin to cell culture. Moreover, in vitro 15d-PGJ2 increased the expression of FOXP3, GITR, and CTLA-4 in the CD4+CD25- population, suggesting the induction of Tregs on conventional T cells. Prostanoid addition to CD4+CD25- cells selectively suppressed Th17 differentiation and promoted the enhancement of FOXP3 under polarization conditions. Thus, 15d-PGJ2 ameliorated symptoms of collagen-induced arthritis by regulating Th17 differentiation, concomitant with the induction of Tregs, and, consequently, protected mice from diseases aggravation. Altogether, these results indicate that 15d-PGJ2 may represent a potential therapeutic strategy in RA.


Subject(s)
Arthritis, Experimental/drug therapy , Arthritis, Experimental/metabolism , CD4 Antigens/metabolism , Forkhead Transcription Factors/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Prostaglandin D2/analogs & derivatives , Th17 Cells/drug effects , Th17 Cells/metabolism , Animals , Arthritis, Experimental/immunology , Male , Mice , Mice, Inbred DBA , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , PPAR gamma/agonists , PPAR gamma/metabolism , Prostaglandin D2/pharmacology , Prostaglandin D2/therapeutic use
12.
J Nutr Biochem ; 38: 86-92, 2016 12.
Article in English | MEDLINE | ID: mdl-27732913

ABSTRACT

Hypertension can be programmed in response to nutritional insults in early life. Maternal high-fructose (HF) intake induced programmed hypertension in adult male offspring, which is associated with renal programming and arachidonic acid metabolism pathway. We examined whether early treatment with a soluble epoxide hydrolase (SEH) inhibitor, 12-(3-adamantan-1-yl-ureido)-dodecanoic acid (AUDA) or 15-Deoxy-Δ12,14-prostagandin J2 (15dPGJ2) can prevent HF-induced programmed hypertension. Pregnant Sprague Dawley rats received regular chow or chow supplemented with fructose (60% diet by weight) during the whole period of pregnancy and lactation. Four groups of male offspring were studied: control, HF, HF+AUDA and HF+15dPGJ2. In HF+AUDA group, mother rats received AUDA 25 mg/L in drinking water during lactation. In the HF+15dPGJ2 group, male offspring received 15dPGJ2 1.5 mg/kg body weight by subcutaneous injection once daily for 1 week after birth. Rats were sacrificed at 12 weeks of age. Maternal HF-induced programmed hypertension is associated with increased renal protein level of SEH and oxidative stress, which early AUDA therapy prevents. Comparison of AUDA and 15dPGJ2 treatments demonstrated that AUDA was more effective in preventing HF-induced programmed hypertension. AUDA therapy increases angiotensin converting enzyme-2 (ACE2) protein levels and PGE2 levels in adult offspring kidney exposed to maternal HF. 15dPGJ2 therapy increases plasma asymmetric dimethylarginine (ADMA) levels and decreases L-arginine-to-ADMA ratio. Better understanding of the impact of arachidonic acid pathway, especially inhibition of SEH, on renal programming may aid in developing reprogramming strategy to prevent programmed hypertension in children exposed to antenatal HF intake.


Subject(s)
Adamantane/analogs & derivatives , Antihypertensive Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , Epoxide Hydrolases/antagonists & inhibitors , Hypertension/prevention & control , Kidney/drug effects , Lauric Acids/therapeutic use , Prostaglandin D2/analogs & derivatives , Adamantane/therapeutic use , Angiotensin-Converting Enzyme 2 , Animals , Antihypertensive Agents/administration & dosage , Biomarkers/blood , Biomarkers/metabolism , Cyclooxygenase 2/chemistry , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Diet, Carbohydrate Loading/adverse effects , Enzyme Repression/drug effects , Epoxide Hydrolases/metabolism , Female , Fetal Development/drug effects , Fructose/adverse effects , Hypertension/etiology , Hypertension/metabolism , Hypertension/pathology , Injections, Subcutaneous , Intramolecular Oxidoreductases/antagonists & inhibitors , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Kidney/metabolism , Kidney/pathology , Lactation , Lipocalins/antagonists & inhibitors , Lipocalins/genetics , Lipocalins/metabolism , Male , Maternal Nutritional Physiological Phenomena , Peptidyl-Dipeptidase A/chemistry , Peptidyl-Dipeptidase A/metabolism , Pregnancy , Prostaglandin D2/administration & dosage , Prostaglandin D2/metabolism , Prostaglandin D2/therapeutic use , Rats , Rats, Sprague-Dawley
13.
Exp Parasitol ; 170: 100-108, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27693222

ABSTRACT

Trypanosoma cruzi, the etiological agent of Chagas' disease, causes an intense inflammatory response in several tissues, including the liver. Since this organ is central to metabolism, its infection may be reflected in the outcome of the disease. 15-deoxy-Δ12,14 prostaglandin J2 (15dPGJ2), a natural agonist of peroxisome-proliferator activated receptor (PPAR) γ, has been shown to exert anti-inflammatory effects in the heart upon T. cruzi infection. However, its role in the restoration of liver function and reduction of liver inflammation has not been studied yet. BALB/c mice were infected with T. cruzi. The effects of in vivo treatment with 15dPGJ2 on liver inflammation and fibrosis, as well as on the GOT/GPT ratio were studied and the role of NF-κB pathway on 15dPGJ2-mediated effects was analysed. 15dPGJ2 reduced liver inflammatory infiltrates, proinflammatory enzymes and cytokines expression, restored the De Ritis ratio values to normal, reduced the deposits of interstitial and perisinusoidal collagen, reduced the expression of the pro-fibrotic cytokines and inhibited the translocation of the p65 NF-κB subunit to the nucleus. Thus, we showed that 15dPGJ2 is able to significantly reduce the inflammatory response and fibrosis and reduced enzyme markers of liver damage in mice infected with T. cruzi.


Subject(s)
Chagas Disease/drug therapy , Immunologic Factors/therapeutic use , Liver/pathology , Prostaglandin D2/analogs & derivatives , Animals , Biomarkers , Chagas Disease/pathology , Cytokines/drug effects , DNA, Protozoan/analysis , DNA, Protozoan/genetics , Liver/metabolism , Liver/parasitology , Liver Cirrhosis/prevention & control , Male , Mice , Mice, Inbred BALB C , NF-kappa B/antagonists & inhibitors , PPAR gamma/antagonists & inhibitors , PPAR gamma/genetics , PPAR gamma/metabolism , Parasite Load , Prostaglandin D2/pharmacology , Prostaglandin D2/therapeutic use , Protozoan Proteins/analysis , Protozoan Proteins/genetics , Real-Time Polymerase Chain Reaction , Trypanosoma cruzi/genetics , Trypanosoma cruzi/physiology
14.
Biomed Pharmacother ; 80: 183-192, 2016 May.
Article in English | MEDLINE | ID: mdl-27133055

ABSTRACT

OBJECTIVE: In this study, we confirmed a protective effect of 15d-PGJ2 in concanavalin A (ConA)-induced fulminant hepatitis in mice and investigated the potential mechanism. MATERIALS AND METHODS: Balb/C mice were injected with ConA (25mg/kg) to induce acute fulminant hepatitis, and 15d-PGJ2 (2.5-10µg) was administered 30min after the ConA injection. The histological grade, pro-inflammatory cytokine and ROS levels, apoptosis and autophagy activity, the expression of HO-1, Nrf2, JNK and Bcl-2 activity were determined 2, 4, and 8h after the ConA injection. RESULTS: Following ConA challenge, the expression of cytokines tumor necrosis factor α (TNF-α) and interleukin 1ß (IL-1ß) was up-regulated. Treatment with 15d-PGJ2 reduced the pathological effects of ConA-induced fulminant hepatitis and significantly reduced the levels of TNF-α, IL-1ß and ROS after injection. 15d-PGJ2 inhibited apoptosis and autophagic cell death, facilitated Nrf2 nuclear translocation, increased HO-1 expression and suppressed the JNK activation. CONCLUSION: 15d-PGJ2 alleviates ConA-induced acute liver injury in mice by up-regulating the anti-oxidative stress factor HO-1 and reducing the production of cytokines and ROS, thereby inhibiting hepatic cell autophagy probably induced by ROS.


Subject(s)
Autophagy/drug effects , Heme Oxygenase-1/metabolism , Hepatocytes/pathology , Liver/injuries , Liver/pathology , Prostaglandin D2/analogs & derivatives , Up-Regulation/drug effects , Acute Disease , Animals , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Concanavalin A , Disease Models, Animal , Enzyme Activation/drug effects , Hepatitis/drug therapy , Hepatitis/pathology , Hepatocytes/drug effects , Hepatocytes/ultrastructure , Interleukin-1beta/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Liver/drug effects , Liver/ultrastructure , Male , Mice, Inbred BALB C , NF-E2-Related Factor 2/metabolism , Phagosomes/drug effects , Phagosomes/metabolism , Phagosomes/ultrastructure , Prostaglandin D2/pharmacology , Prostaglandin D2/therapeutic use , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/metabolism
15.
Comput Math Methods Med ; 2016: 3628124, 2016.
Article in English | MEDLINE | ID: mdl-28044089

ABSTRACT

Researchers have observed that response of tumor cells to treatment varies depending on whether the cells are grown in monolayer, as in vitro spheroids or in vivo. This study uses data from the literature on monolayer treatment of SK-N-SH neuroblastoma cells with 15-deoxy-PGJ2 and couples it with data on growth rates for untreated SK-N-SH neuroblastoma cells grown as multicellular spheroids. A linear model is constructed for untreated and treated monolayer data sets, which is tuned to growth, death, and cell cycle data for the monolayer case for both control and treatment with 15-deoxy-PGJ2. The monolayer model is extended to a five-dimensional nonlinear model of in vitro tumor spheroid growth and treatment that includes compartments of the cell cycle (G1, S, G2/M) as well as quiescent (Q) and necrotic (N) cells. Monolayer treatment data for 15-deoxy-PGJ2 is used to derive a prediction of spheroid response under similar treatments. For short periods of treatment, spheroid response is less pronounced than monolayer response. The simulations suggest that the difference in response to treatment of monolayer versus spheroid cultures observed in laboratory studies is a natural consequence of tumor spheroid physiology rather than any special resistance to treatment.


Subject(s)
Antineoplastic Agents/therapeutic use , Neuroblastoma/drug therapy , Prostaglandin D2/analogs & derivatives , Spheroids, Cellular/drug effects , Algorithms , Cell Cycle/drug effects , Cell Division , Cell Line, Tumor , Cell Survival , Dose-Response Relationship, Drug , Humans , Models, Biological , Models, Statistical , Prostaglandin D2/therapeutic use
16.
Mol Neurobiol ; 53(6): 3626-3645, 2016 08.
Article in English | MEDLINE | ID: mdl-26108185

ABSTRACT

Peroxisome proliferator-activated receptor-gamma (PPAR-γ), a stress-induced transcription factor, protects neurons against ischemic stroke insult by reducing oxidative stress. NADPH oxidase (NOX) activation, a major driving force in ROS generation in the setting of reoxygenation/reperfusion, constitutes an important pathogenetic mechanism of ischemic brain damage. In the present study, both transient in vitro oxygen-glucose deprivation and in vivo middle cerebral artery (MCA) occlusion-reperfusion experimental paradigms of ischemic neuronal death were used to investigate the interaction between PPAR-γ and NOX. With pharmacological (PPAR-γ antagonist GW9662), loss-of-function (PPAR-γ siRNA), and gain-of-function (Ad-PPAR-γ) approaches, we first demonstrated that 15-deoxy-∆(12,14)-PGJ2 (15d-PGJ2), via selectively attenuating p22phox expression, inhibited NOX activation and the subsequent ROS generation and neuronal death in a PPAR-γ-dependent manner. Secondly, results of promoter analyses and subcellular localization studies further revealed that PPAR-γ, via inhibiting hypoxia-induced NF-κB nuclear translocation, indirectly suppressed NF-κB-driven p22phox transcription. Noteworthily, postischemic p22phox siRNA treatment not only reduced infarct volumes but also improved functional outcome. In summary, we report a novel transrepression mechanism involving PPAR-γ downregulation of p22phox expression to suppress the subsequent NOX activation, ischemic neuronal death, and brain infarct. Identification of a PPAR-γ → NF-κB → p22phox neuroprotective signaling cascade opens a new avenue for protecting the brain against ischemic insult.


Subject(s)
Apoptosis , Brain Ischemia/pathology , Cytochrome b Group/metabolism , NADPH Oxidases/metabolism , NF-kappa B/metabolism , Neurons/pathology , PPAR gamma/metabolism , Transcription, Genetic , Animals , Apoptosis/drug effects , Base Sequence , Brain Ischemia/complications , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cells, Cultured , Cerebral Cortex/pathology , Cerebral Infarction/complications , Cerebral Infarction/drug therapy , Cerebral Infarction/pathology , Cytosol/metabolism , Down-Regulation/drug effects , Glucose/deficiency , Male , Mice , Neurons/drug effects , Neurons/metabolism , Oxidation-Reduction , Oxygen , Promoter Regions, Genetic/genetics , Prostaglandin D2/analogs & derivatives , Prostaglandin D2/pharmacology , Prostaglandin D2/therapeutic use , Protein Binding/drug effects , Protein Transport/drug effects , RNA, Small Interfering/metabolism , Rats, Long-Evans , Reactive Oxygen Species/metabolism , Transcription, Genetic/drug effects
17.
Eur J Pharmacol ; 740: 58-65, 2014 Oct 05.
Article in English | MEDLINE | ID: mdl-25016088

ABSTRACT

The aim of this study was to evaluate the peripheral effect of 15-deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2) in albumin-induced arthritis in temporomandibular joint (TMJ) of rats. Antigen-induced arthritis (AIA) was generated in rats with methylated bovine serum albumin (mBSA) diluted in complete Freund׳s adjuvant. Pretreatment with an intra-articular injection of 15d-PGJ2 (100 ng/TMJ) before mBSA intra-articular injection (10 µg/TMJ) (challenge) in immunized rats significantly reduced the albumin-induced arthritis inflammation. The results demonstrated that 15d-PGJ2 was able to inhibit plasma extravasation, leukocyte migration and the release of inflammatory cytokines IL-6, IL-12, IL-18 and the chemokine CINC-1 in the TMJ tissues. In addition, 15d-PGJ2 was able to increase the expression of the anti-adhesive molecule CD55 and the anti-inflammatory cytokine IL-10. Taken together, it is possible to suggest that 15d-PGJ2 inhibit leukocyte infiltration and subsequently inflammatory process, through a shift in the balance of the pro- and anti-adhesive properties. Thus, 15d-PGJ2 might be used as a potential anti-inflammatory drug to treat arthritis-induced inflammation of the temporomandibular joint.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Arthritis, Experimental/drug therapy , Prostaglandin D2/analogs & derivatives , Temporomandibular Joint Disorders/drug therapy , Animals , Anti-Inflammatory Agents/pharmacology , Antigens , Arthritis, Experimental/immunology , CD55 Antigens/immunology , Cell Movement/drug effects , Cytokines/immunology , Freund's Adjuvant , Injections, Intra-Articular , Intercellular Adhesion Molecule-1/immunology , Leukocytes/drug effects , Leukocytes/physiology , Male , Prostaglandin D2/pharmacology , Prostaglandin D2/therapeutic use , Rats, Wistar , Serum Albumin, Bovine , Temporomandibular Joint Disorders/immunology
18.
Chin Med J (Engl) ; 127(5): 815-20, 2014.
Article in English | MEDLINE | ID: mdl-24571868

ABSTRACT

BACKGROUND: A proinflammatory milieu emerging in the lung due to neutrophil accumulation and activation is a key in the pathogenesis of acute lung injury (ALI). 15-deoxy-Δ(12, 14)-prostaglandin J2 (15d-PGJ2), one of the terminal products of the cyclooxygenase-2 pathway, is known to be the endogenous ligand of peroxisome proliferator-activated receptor γ (PPAR-γ) with multiple physiological properties. Growing evidence indicates that 15d-PGJ2 has anti-inflammatory, antiproliferative, cytoprotective and pro-resolving effects. We investigated whether 15d-PGJ2 has a protective effect against endotoxin-induced acute lung injury in rats. METHODS: Twenty-four male Wistar rats were randomly assigned into four groups (n = 6 per group): sham+vehicle group, sham+15d-PGJ2 group, LPS+vehicle group, and LPS+15d-PGJ2 group. The rats were given either lipopolysaccharide (LPS, 6 mg/kg intravenously) or saline, and pretreated with 15d-PGJ2 (0.3 mg/kg intravenously) or its vehicle (dimethyl sulphoxide) 30 minutes before LPS. Histological alterations, wet/dry weight (W/D) ratio and myeloperoxidase (MPO) activity as well as tumor necrosis factor (TNF)-α and cytokine-induced neutrophil chemoattractant-1 (CINC-1) levels were determined in lung tissues four hours after LPS injection. Immunohistochemical analysis for intercellular adhesion molecule-1 (ICAM-1) expression and Western blotting analysis for nuclear factor (NF)-κB p65 translocation and IκBα protein levels were also studied. RESULTS: 15d-PGJ2 pretreatment significantly attenuated LPS-induced lung injury, and reduced the increased W/D ratio, MPO activity, TNF-α, CINC-1 levels, and ICAM-1 expression in the lung. 15d-PGJ2 also suppressed the nuclear NF-κB p65 translocation and increased cytosolic IκBα levels. CONCLUSIONS: 15d-PGJ2 protects against endotoxin-induced acute lung injury, most likely through the reduction of proinflammatory protein levels during endotoxemia subsequent to the inhibition of NF-κB activation.


Subject(s)
Acute Lung Injury/chemically induced , Acute Lung Injury/drug therapy , Lipopolysaccharides/toxicity , Prostaglandin D2/analogs & derivatives , Acute Lung Injury/immunology , Animals , Chemokine CXCL1/metabolism , I-kappa B Proteins/metabolism , Intercellular Adhesion Molecule-1/metabolism , Male , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Prostaglandin D2/therapeutic use , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha/metabolism
19.
J Alzheimers Dis ; 40(2): 245-56, 2014.
Article in English | MEDLINE | ID: mdl-24413616

ABSTRACT

Mitochondria are essential cytoplasmic organelles, critical for cell survival and death. Recent mitochondrial research revealed that mitochondrial dynamics-the balance of fission and fusion in normal mitochondrial dynamics--is an important cellular mechanism in eukaryotic cell and is involved in the maintenance of mitochondrial morphology, structure, number, distribution, and function. Research into mitochondria and cell function has revealed that mitochondrial dynamics is impaired in a large number of aging and neurodegenerative diseases, and in several inherited mitochondrial diseases, and that this impairment involves excessive mitochondrial fission, resulting in mitochondrial structural changes and dysfunction, and cell damage. Attempts have been made to develop molecules to reduce mitochondrial fission while maintaining normal mitochondrial fusion and function in those diseases that involve excessive mitochondrial fission. This review article discusses mechanisms of mitochondrial fission in normal and diseased states of mammalian cells and discusses research aimed at developing therapies, such as Mdivi, Dynasore and P110, to prevent or to inhibit excessive mitochondrial fission.


Subject(s)
Mitochondrial Diseases/therapy , Mitochondrial Dynamics/physiology , Oxidative Stress/physiology , Animals , Humans , Hydrazones/therapeutic use , Mitochondrial Diseases/metabolism , Mitochondrial Dynamics/drug effects , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/metabolism , Oxidative Stress/drug effects , Prostaglandin D2/analogs & derivatives , Prostaglandin D2/therapeutic use , Quinazolinones/therapeutic use
20.
J Physiol Pharmacol ; 65(6): 867-76, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25554991

ABSTRACT

Cyclosporine A(CsA) is an immunosuppressor frequently used in the transplant surgery and in the treatment of autoimmune diseases. The therapeutic benefits of CsA are often limited by it's main side effect-nephrotoxicity. Mechanisms of chronic CsA- induced renal damage include: activation of renin-angiotensin-aldosterone system, upregulation of transforming growth factor beta (TGF-ß), oxidative stress. This study was undertaken to investigate the protective effect of the peroxisome-proliferator-activated receptors gamma (PPARs-γ) agonists: rosiglitazone and 15-deoxy-Δ12,14-prostaglandin J2 (PGDJ2), against CsA-induced kidney injury in male Wistar rats. CsA was administered subcutaneously at a dose of 15 mg/kg/day for 28 days. Both PPAR-γ agonists were given for 28 days 0.5 hour before the administration of CsA. Rosiglitazone was administered orally at a dose of 8 mg/kg/day and PGDJ2 was given intraperitoneally at a dose of 30 µg/kg/day. CsA induced renal failure was evidenced by increased serum levels of urea, uric acid and creatinine. Serum concentrations of GSH and GSSG, lipid peroxidation products as well as NAD+/NADH, NADP+/NADPH and ADP/ATP ratios showed, that CsA induced oxidative stress and evoked an imbalanced red-ox state in the kidney. Light and electron microscope studies showed degenerative changes within renal tubules with damage to their mitochondria, interstitial fibrosis and arteriolopathy. Immunohistochemical expression of profibrotic TGF-ß was assessed. The biochemical and morphological changes induced by CsA were limited by administration of both rosiglitazone and PGDJ2. Ultrastructural examination of renal tubular epithelial cells showed marked improvement within mitochondria. Our results indicate that both PPAR-γ agonists used in the experiment may play an important role in protecting against CsA-induced damage in the kidney.


Subject(s)
Kidney Diseases/drug therapy , PPAR gamma/agonists , Prostaglandin D2/analogs & derivatives , Protective Agents/therapeutic use , Thiazolidinediones/therapeutic use , Animals , Creatinine/blood , Cyclosporine , Disease Models, Animal , Glutathione/metabolism , Glutathione Disulfide/metabolism , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Kidney Diseases/chemically induced , Kidney Diseases/metabolism , Kidney Diseases/pathology , Male , NAD/metabolism , NADP/metabolism , Prostaglandin D2/pharmacology , Prostaglandin D2/therapeutic use , Protective Agents/pharmacology , Rats, Wistar , Rosiglitazone , Thiazolidinediones/pharmacology , Urea/blood , Uric Acid/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...