Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 824
Filtrar
1.
Front Endocrinol (Lausanne) ; 13: 983924, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36213265

RESUMO

Background: Prematurity is the leading cause of childhood death under the age of five. The aetiology of preterm birth is multifactorial; however, inflammation and infection are the most common causal factors, supporting a potential role for immunomodulation as a therapeutic strategy. 15-Deoxy-Delta-12,14-prostaglandin J2 (15dPGJ2) is an anti-inflammatory prostaglandin and has been shown to delay lipopolysaccharide (LPS) induced preterm labour in mice and improve pup survival. This study explores the immunomodulatory effect of 15dPGJ2 on the transcription factors NF-κB and AP-1, pro-inflammatory cytokines, and contraction associated proteins in human cultured myocytes, vaginal epithelial cell line (VECs) and primary amnion epithelial cells (AECs). Methods: Cells were pre-incubated with 32µM of 15dPGJ2 and stimulated with 1ng/mL of IL-1ß as an in vitro model of inflammation. Western immunoblotting was used to detect phosphorylated p-65 and phosphorylated c-Jun as markers of NF-κB and AP-1 activation, respectively. mRNA expression of the pro-inflammatory cytokines IL-6, IL-8, and TNF-α was examined, and protein expression of COX-2 and PGE2 were detected by western immunoblotting and ELISA respectively. Myometrial contractility was examined ex-vivo using a myograph. Results: 15dPGJ2 inhibited IL-1ß-induced activation of NF-κB and AP-1, and expression of IL-6, IL-8, TNF-α, COX-2 and PGE2 in myocytes, with no effect on myometrial contractility or cell viability. Despite inhibiting IL-1ß-induced activation of NF-κB, expression of IL-6, TNF-α, and COX-2, 15dPGJ2 led to activation of AP-1, increased production of PGE2 and increased cell death in VECs and AECs. Conclusion: We conclude that 15dPGJ2 has differential effects on inflammatory modulation depending on cell type and is therefore unlikely to be a useful therapeutic agent for the prevention of preterm birth.


Assuntos
NF-kappa B , Nascimento Prematuro , Âmnio , Animais , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Ciclo-Oxigenase 2/farmacologia , Citocinas/metabolismo , Dinoprostona/metabolismo , Dinoprostona/farmacologia , Dinoprostona/uso terapêutico , Células Epiteliais/metabolismo , Feminino , Humanos , Recém-Nascido , Inflamação/metabolismo , Interleucina-6 , Interleucina-8/metabolismo , Interleucina-8/farmacologia , Interleucina-8/uso terapêutico , Lipopolissacarídeos , Camundongos , Células Musculares/metabolismo , NF-kappa B/metabolismo , Prostaglandina D2/análogos & derivados , RNA Mensageiro/metabolismo , Fator de Transcrição AP-1/metabolismo , Fator de Transcrição AP-1/farmacologia , Fator de Transcrição AP-1/uso terapêutico , Fator de Necrose Tumoral alfa/metabolismo
2.
Acta Pharmacol Sin ; 43(5): 1251-1263, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-34417577

RESUMO

Transcriptional factor EB (TFEB), a master regulator of autophagy and lysosomal biogenesis, is generally regarded as a pro-survival factor. Here, we identify that besides its effect on autophagy induction, TFEB exerts a pro-apoptotic effect in response to the cyclopentenone prostaglandin 15-deoxy-∆-12,14-prostaglandin J2 (15d-PGJ2). Specifically, 15d-PGJ2 promotes TFEB translocation from the cytoplasm into the nucleus to induce autophagy and lysosome biogenesis via reactive oxygen species (ROS) production rather than mTORC1 inactivation. Surprisingly, TFEB promotes rather than inhibits apoptosis in response to 15d-PGJ2. Mechanistically, ROS-mediated TFEB translocation into the nucleus transcriptionally upregulates the expression of ATF4, which is required for apoptosis elicited by 15d-PGJ2. Additionally, inhibition of TFEB activation by ROS scavenger N-acetyl cysteine or inhibition of protein synthesis by cycloheximide effectively compromises ATF4 upregulation and apoptosis in response to 15d-PGJ2. Collectively, these results indicate that ROS-induced TFEB activation exerts a novel role in promoting apoptosis besides its role in regulating autophagy in response to 15d-PGJ2. This work not only evidences how TFEB is activated by 15d-PGJ2, but also unveils a previously unexplored role of ROS-dependent activation of TFEB in modulating cell apoptosis in response to 15d-PGJ2.


Assuntos
Prostaglandina D2 , Prostaglandinas , Apoptose , Autofagia , Ciclopentanos , Prostaglandina D2/análogos & derivados , Prostaglandina D2/farmacologia , Prostaglandinas/farmacologia , Espécies Reativas de Oxigênio/metabolismo
3.
Int J Mol Sci ; 22(21)2021 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-34769194

RESUMO

Osteosarcoma (OS) is the most common type of bone tumor, and has limited therapy options. 15-Deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2) has striking anti-tumor effects in various tumors. Here, we investigated molecular mechanisms that mediate anti-tumor effects of 15d-PGJ2 in different OS cell lines. Human U2-OS and Saos-2 cells were treated with 15d-PGJ2 and cell survival was measured by MTT assay. Cell proliferation and motility were investigated by scratch assay, the tumorigenic capacity by colony forming assay. Intracellular ROS was estimated by H2DCFDA. Activation of MAPKs and cytoprotective proteins was detected by immunoblotting. Apoptosis was detected by immunoblotting and Annexin V/PI staining. The ex ovo CAM model was used to study growth capability of grafted 15d-PGJ2-treated OS cells, followed by immunohistochemistry with hematoxylin/eosin and Ki-67. 15d-PGJ2 substantially decreased cell viability, colony formation and wound closure capability of OS cells. Non-malignant human osteoblast was less affected by 15d-PGJ2. 15d-PGJ2 induced rapid intracellular ROS production and time-dependent activation of MAPKs (pERK1/2, pJNK and pp38). Tempol efficiently inhibited 15d-PGJ2-induced ERK1/2 activation, while N-acetylcystein and pyrrolidine dithiocarbamate were less effective. Early but weak activation of cytoprotective proteins was overrun by induction of apoptosis. A structural analogue, 9,10-dihydro-15d-PGJ2, did not show toxic effects in OS cells. In the CAM model, we grafted OS tumors with U2-OS, Saos-2 and MG-63 cells. 15d-PGJ2 treatment resulted in significant growth inhibition, diminished tumor tissue density, and reduced tumor cell proliferation for all cell lines. Our in vitro and CAM data suggest 15d-PGJ2 as a promising natural compound to interfere with OS tumor growth.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias Ósseas/tratamento farmacológico , Osteossarcoma/tratamento farmacológico , Prostaglandina D2/análogos & derivados , Animais , Neoplasias Ósseas/metabolismo , Linhagem Celular Tumoral , Galinhas , Ativação Enzimática/efeitos dos fármacos , Humanos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Osteossarcoma/metabolismo , Prostaglandina D2/farmacologia , Espécies Reativas de Oxigênio/metabolismo
4.
Anim Reprod Sci ; 234: 106866, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34626868

RESUMO

The current study was conducted with the aim to investigate effects of PPARγ ligands on synthesis of nuclear receptor κB (NF-κB) and selected cytokines (IL-1ß, IFNγ, TNFα, IL-4, IL-10, LIF) in the pig myometrium on days 14-15 of the estrous cycle (late-luteal phase) and days 14-15 of the gestational period (beginning of embryonic implantation). The myometrial slices were incubated in vitro for 6 h in medium containing PPARγ ligands, agonists: 15d-prostaglandin J2 or pioglitazone, and antagonist - T0070907. The mRNA transcript and protein abundances were evaluated in tissues and culture medium. During the estrous cycle, PPARγ ligands did not have an effect on the mRNA transcript abundance of the immune response mediators used for treatments. The IL-10 protein abundance in the tissue was less when there was inclusions of pioglitazone in the medium, while the treatment with T0070907 resulted in a larger abundance of NF-κB, IL-1ß (in the tissue) and IL-4 (in tissue and culture media). During the gestational period, pioglitazone or PGJ2 suppressed mRNA IFNγ and IL-10 transcript and protein abundances (in the tissue and culture media), whereas there was an enhanced NF-κB protein abundance (in the tissue). Treatment with T0070907 had diverse effects (e.g., for NFκB inhibited mRNA transcript abundance or enhanced protein abundance). The observed changes are related mainly in tissues from pregnant animals. Responses to PPARγ antagonist are indicative of the possible involvement of PPARγ-independent factors as well as ligand-independent activation of the receptor, ligand selectivity/functionality or tissue receptivity to the factors evaluated.


Assuntos
Imunidade/fisiologia , Miométrio/metabolismo , PPAR gama/metabolismo , Suínos/fisiologia , Animais , Antineoplásicos/farmacologia , Benzamidas/farmacologia , Citocinas/genética , Citocinas/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Hipoglicemiantes/farmacologia , PPAR gama/genética , Pioglitazona/farmacologia , Gravidez , Prostaglandina D2/análogos & derivados , Prostaglandina D2/farmacologia , Piridinas/farmacologia , Técnicas de Cultura de Tecidos
5.
Front Endocrinol (Lausanne) ; 12: 708248, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34557159

RESUMO

Purpose: The inhibition of estrogen receptor alpha (ERα) or the activation of ERß can inhibit papillary thyroid cancer (PTC), but the precise mechanism is not known. We aimed to explore the role of ERα and ERß on the production of endogenous peroxisome proliferator-activated receptor gamma (PPARγ) ligands in PTC. Methods: 2 PTC cell lines, 32 pairs of PTC tissues and matched normal thyroid tissues were used in this study. The levels of endogenous PPARγ ligands 15(S)-hydroxyeicosatetraenoic acid (15(S)-HETE), 13-S-hydroxyoctadecadienoic acid (13(S)-HODE), and15-deoxy-Δ12,14-prostaglandin J2 (PGJ2) were measured by ELISA. Results: The levels of PGJ2 and 15(S)-HETE were significantly reduced in PTC, but 13(S)-HODE was not changed. Activation of ERα or inhibition of ERß significantly downregulated the production of PGJ2, 15(S)-HETE and 13(S)-HODE, whereas inhibition of ERα or activation of ERß markedly upregulated the production of these three ligands. Application of endogenous PPARγ ligands inhibited growth, induced apoptosis of cancer cells, and promoted the efficacy of chemotherapy. Conclusion: The levels of endogenous PPARγ ligands PGJ2 and 15(S)-HETE are significantly decreased in PTC. The inhibition of ERα or activation of ERß can inhibit PTC by stimulating the production of endogenous PPARγ ligands to induce apoptosis in cancer cells.


Assuntos
Receptor beta de Estrogênio/metabolismo , Ácidos Hidroxieicosatetraenoicos/metabolismo , Ácidos Linoleicos/metabolismo , PPAR gama/metabolismo , Prostaglandina D2/análogos & derivados , Câncer Papilífero da Tireoide/patologia , Adulto , Apoptose , Movimento Celular , Proliferação de Células , Feminino , Humanos , Ligantes , Masculino , Pessoa de Meia-Idade , Prognóstico , Prostaglandina D2/metabolismo , Câncer Papilífero da Tireoide/metabolismo , Neoplasias da Glândula Tireoide/metabolismo , Neoplasias da Glândula Tireoide/patologia , Células Tumorais Cultivadas
6.
Prostaglandins Other Lipid Mediat ; 156: 106583, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34332056

RESUMO

15-Deoxy-Δ-12,14-prostaglandin J2 (15d-PGJ2) is an endogenous agonist of the ligand dependent transcriptional factor, peroxisome proliferator-activated receptor -gamma (PPAR-γ). Although PPAR-γ mediates some actions of 15d-PGJ2, many actions of 15d-PGJ2 are independent of PPAR-γ. The PPAR-γ signaling pathway has beneficial effects on tumor progression, inflammation, oxidative stress, and angiogenesis in numerous studies. In this review, various studies were analyzed to understand the effects of 15d-PGJ2 in vascular smooth muscle cells (VSMC)s. 15d-PGJ2 inhibits proliferation of VSMCs during vascular remodeling and it alters the expression of contractile proteins and inflammatory components within these cells as well. However, the effects of 15d-PGJ2 as well as its ability to induce PPAR-γ activation remains controversial as contradictory effects of this prostaglandin in VSMCs exist. Understanding the mechanisms by which 15d-PGJ2 elicit beneficial actions whether by PPAR-γ activation or independently, will aid in developing new therapeutic strategies for diseases such as hypertension with an inflammatory component. Although great advances are being made, more research is needed to reach definitive conclusions.


Assuntos
Prostaglandina D2/análogos & derivados
7.
J Immunol Res ; 2021: 5591115, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33997056

RESUMO

Tetranor-PGDM is a metabolite of PGD2. Urinary tetranor-PGDM levels were reported to be increased in some diseases, including food allergy, Duchenne muscular dystrophy, and aspirin-intolerant asthma. In this study, we developed a monoclonal antibody (MAb) and a competitive enzyme immunoassay (EIA) for measuring tetranor-PGDM. Spleen cells isolated from mice immunized with tetranor-PGDM were utilized to generate Ab-producing hybridomas. We chose hybridomas and purified MAb against tetranor-PGDM to develop competitive EIA. The assay evaluated the optimal ionic strength, pH, precision, and reliability. Specificity was determined by cross-reactivity to tetranor-PGEM, tetranor-PGFM, and tetranor-PGAM. Recovery was determined by spiking experiments on artificial urine. Optimal ionic strength was 150 mM NaCl, and optimal pH was pH 7.5. Metabolites other than tetranor-PGDM did not show any significant cross-reactivity in the EIA. The assay exhibited a half-maximal inhibition concentration (IC50) of 1.79 ng/mL, limit of detection (LOD) of 0.0498 ng/mL, and range of quantitation (ROQ) value of 0.252 to 20.2 ng/mL. The intra- and inter-assay variation for tetranor-PGDM was 3.9-6.0% and 5.7-10.4%, respectively. The linearity-dilution effect showed excellent linearity under dilution when artificial urine samples were applied to solid-phase extraction (SPE). After SPE, recovery of tetranor-PGDM in artificial urine averaged from 82.3% to 113.5% and was within acceptable limits (80%-120%). We successfully generated one monoclonal antibody and developed a sensitive competitive EIA. The established EIA would be useful for routine detection and monitoring of tetranor-PGDM in research or diagnostic body fluids.


Assuntos
Anticorpos Monoclonais/imunologia , Técnicas Imunoenzimáticas/métodos , Prostaglandina D2/análogos & derivados , Animais , Anticorpos Monoclonais/isolamento & purificação , Feminino , Camundongos , Modelos Animais , Prostaglandina D2/imunologia , Prostaglandina D2/metabolismo , Prostaglandina D2/urina , Reprodutibilidade dos Testes
8.
Brain Behav Immun ; 95: 462-476, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33964434

RESUMO

Physically active individuals are less likely to develop chronic pain, and physical exercise is an established strategy to control inflammatory diseases. Here, we hypothesized that 1) peripheral pro-inflammatory macrophages phenotype contribute to predisposition of the musculoskeletal to chronic pain, and that 2) activation of PPARγ receptors, modulation of macrophage phenotypes and cytokines through physical exercise would prevent persistent muscle pain. We tested these hypotheses using swimming exercise, pharmacological and immunochemical techniques in a rodent model of persistent muscle hyperalgesia. Swimming prevented the persistent mechanical muscle hyperalgesia most likely through activation of PPARγ receptors, as well as activation of PPARγ receptors by 15d-PGJ2 and depletion of muscle macrophages in sedentary animals. Acute and persistent muscle hyperalgesia were characterized by an increase in pro-inflammatory macrophages phenotype, and swimming and the 15d-PGJ2 prevented this increase and increased anti-inflammatory macrophages phenotype. Finally, IL-1ß concentration in muscle increased in the acute phase, which was also prevented by PPARγ receptors activation through swimming. Besides, swimming increased muscle concentration of IL-10 in both acute and chronic phases, but only in the persistent phase through PPARγ receptors. Our findings suggest physical exercise activates PPARγ receptors and increases anti-inflammatory responses in the muscle tissue by modulating macrophages phenotypes and cytokines, thereby preventing the establishment of persistent muscle hyperalgesia. These results further highlight the potential of physical exercise to prevent chronic muscle pain.


Assuntos
Hiperalgesia , Macrófagos , Músculos/metabolismo , PPAR gama , Condicionamento Físico Animal , Animais , Citocinas , Masculino , Camundongos , Fenótipo , Prostaglandina D2/análogos & derivados
9.
Artigo em Inglês | MEDLINE | ID: mdl-33812217

RESUMO

OBJECTIVE: Cyclooxygenase (COX)-derived prostaglandin E2 (PGE2) is an important lipid mediator in colorectal carcinoma (CRC) pathogenesis. Other lipid mediators derived from lipoxygenases (LOX) have also been implicated in neoplastic processes in the colon. In this study we aimed to characterize lipid mediators, so called oxylipins, in human colon adenomatous polyps. DESIGN: We quantified oxylipins in healthy colon tissue and colorectal adenoma tissue procured during routine colonoscopy examinations. Lipid metabolite profiles were analyzed by liquid chromatography-tandem mass spectrometry. RESULTS: Adenoma tissue showed a distinct prostaglandin profile as compared to normal colon mucosa. Interestingly, PGE2 was not higher in adenoma tissue as compared to normal mucosa. In contrast, we found significantly lower levels of prostaglandin D2, prostaglandin J2, and prostaglandin D1 in adenoma tissue. Furthermore, levels of 5-LOX and 12-LOX pathway products were clearly increased in adenoma biopsy samples. We also investigated the effect of aspirin treatment on prostaglandin profiles in adenoma tissue in a subset of patients and found a trend towards decreased prostaglandin levels in response to aspirin. CONCLUSION: The human data presented here show specific changes of oxylipin profiles in colon adenoma tissue with decreased prostaglandin D2 levels as well as increased 5- and 12-LOX metabolites.


Assuntos
Adenoma/patologia , Colo/patologia , Neoplasias do Colo/patologia , Oxilipinas/metabolismo , Adenoma/metabolismo , Idoso , Araquidonato 5-Lipoxigenase/metabolismo , Estudos de Casos e Controles , Colo/metabolismo , Neoplasias do Colo/metabolismo , Ciclo-Oxigenase 2/metabolismo , Feminino , Humanos , Masculino , Projetos Piloto , Prostaglandina D2/análogos & derivados , Prostaglandina D2/metabolismo , Prostaglandinas D/metabolismo
10.
Front Immunol ; 12: 615803, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33633749

RESUMO

Uncontrolled macrophage functions cause failure to resolve gut inflammation and has been implicated in the pathogenesis of inflammatory bowel disease (IBD). 15-Deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2), one of endogenous lipid mediators formed from arachidonic acid during the inflammatory process, has been reported to terminate inflammation. However, the pro-resolving effect of 15d-PGJ2 on intestinal inflammation and underlying molecular mechanisms remain largely unknown. In the present study, we examined the effects of 15d-PGJ2 on the resolution of dextran sulfate sodium (DSS)-induced murine colitis that mimics human IBD. Pharmacologic inhibition of prostaglandin D synthase (PGDS) responsible for the synthesis of 15d-PGJ2 hampered resolution of inflammation in the colonic mucosa of mice treated with DSS. Notably, intraperitoneal injection of 15d-PGJ2 accelerated the resolution of experimentally induced colitis. 15d-PGJ2 treatment reduced the number of neutrophils and M1 macrophages, while it increased the proportion of M2 macrophages. Moreover, 15d-PGJ2 treated mice exhibited the significantly reduced proportion of macrophages expressing the pro-inflammatory cytokine, IL-6 with concomitant suppression of STAT3 phosphorylation in the colonic mucosa of mice administered 2.5% DSS in drinking water. Taken together, these findings clearly indicate that 15d-PGJ2, endogenously generated from arachidonic acid by cyclooxygenase-2 and PGDS activities in inflamed tissue, promotes resolution of intestinal colitis.


Assuntos
Anti-Inflamatórios/farmacologia , Colite/tratamento farmacológico , Fatores Imunológicos/farmacologia , Prostaglandina D2/análogos & derivados , Animais , Biomarcadores , Colite/etiologia , Colite/patologia , Sulfato de Dextrana/efeitos adversos , Modelos Animais de Doenças , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Prostaglandina D2/farmacologia , Fator de Transcrição STAT3 , Resultado do Tratamento
11.
FEBS Lett ; 595(5): 604-622, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33452674

RESUMO

Signal transducer and activator of transcription 3 (STAT3) has been considered as a potential target for development of anticancer therapeutics. Here, we report a novel mechanism by which the cyclopentenone prostaglandin, 15-deoxy-Δ12,14 -prostaglandin J2 (15d-PGJ2 ) functions as an allosteric inhibitor of STAT3. 15d-PGJ2 inhibits phosphorylation, dimerization, nuclear translocation, and transcriptional activity of STAT3 in H-Ras-transformed human mammary epithelial cells (MCF10A-Ras) through the Michael addition reaction at cysteine 259 of STAT3. Comparative studies with 15d-PGJ2 analogues reveal that both C12-C13 and C9-C10 double bonds conjugated to the carbonyl group in the cyclopentenone ring of 15d-PGJ2 are essential for STAT3 binding. Antiproliferative and pro-apoptotic activities of 15d-PGJ2 in MCF10A-Ras cells are attributable to covalent modification of STAT3 on Cys259, and mimic the effects induced by mutation of this amino acid.


Assuntos
Antineoplásicos/farmacologia , Cisteína/química , Células Epiteliais/efeitos dos fármacos , Prostaglandina D2/análogos & derivados , Proteínas Proto-Oncogênicas p21(ras)/genética , Fator de Transcrição STAT3/antagonistas & inibidores , Sequência de Aminoácidos , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Apoptose/genética , Linhagem Celular Transformada , Proliferação de Células/efeitos dos fármacos , Cisteína/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Regulação Neoplásica da Expressão Gênica , Humanos , Glândulas Mamárias Humanas/metabolismo , Glândulas Mamárias Humanas/patologia , Fosforilação/efeitos dos fármacos , Prostaglandina D2/química , Prostaglandina D2/farmacologia , Ligação Proteica , Multimerização Proteica/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Relação Estrutura-Atividade , Transcrição Gênica
12.
Invest Ophthalmol Vis Sci ; 62(1): 29, 2021 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-33507228

RESUMO

Purpose: Thyroid eye disease (TED) is a condition that causes the tissue behind the eye to become inflamed and can result in excessive fatty tissue accumulation in the orbit. Two subpopulations of fibroblasts reside in the orbit: those that highly express Thy1 (Thy1+) and those with little or no Thy1 (Thy1-). Thy1- orbital fibroblasts (OFs) are more prone to lipid accumulation than Thy1+ OFs. The purpose of this study was to investigate the mechanisms whereby Thy1- OFs more readily accumulate lipid. Methods: We screened Thy1+ and Thy1- OFs for differences in microRNA (miRNA) expression. The effects of increasing miR-130a levels in OFs was investigated by measuring lipid accumulation and visualizing lipid deposits. To determine if adenosine monophosphate-activated protein kinase (AMPK) is important for lipid accumulation, we performed small interfering RNA (siRNA)-mediated knockdown of AMPKß1. We measured AMPK expression and activity using immunoblotting for AMPK and AMPK target proteins. Results: We determined that miR-130a was upregulated in Thy1- OFs and that miR-130a targets two subunits of AMPK. Increasing miR-130a levels enhanced lipid accumulation and reduced expression of AMPKα and AMPKß in OFs. Depletion of AMPK also increased lipid accumulation. Activation of AMPK using AICAR attenuated lipid accumulation and increased phosphorylation of acetyl-CoA carboxylase (ACC) in OFs. Conclusions: These data suggest that when Thy1- OFs accumulate in TED, miR-130a levels increase, leading to a decrease in AMPK activity. Decreased AMPK activity promotes lipid accumulation in TED OFs, leading to excessive fatty tissue accumulation in the orbit.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Fibroblastos/metabolismo , Regulação da Expressão Gênica/fisiologia , Oftalmopatia de Graves/metabolismo , Metabolismo dos Lipídeos/fisiologia , MicroRNAs/genética , Adulto , Idoso , Western Blotting , Células Cultivadas , Feminino , Fibroblastos/efeitos dos fármacos , Inativação Gênica , Humanos , Immunoblotting , Masculino , Pessoa de Meia-Idade , Órbita/citologia , Prostaglandina D2/análogos & derivados , Prostaglandina D2/farmacologia , RNA Interferente Pequeno/farmacologia , Reação em Cadeia da Polimerase em Tempo Real , Antígenos Thy-1/metabolismo
13.
Acta Pharmacol Sin ; 42(3): 422-435, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32694760

RESUMO

Oral administration of curcumin has been shown to inhibit pulmonary fibrosis (PF) despite its extremely low bioavailability. In this study, we investigated the mechanisms underlying the anti-PF effect of curcumin in focus on intestinal endocrine. In bleomycin- and SiO2-treated mice, curcumin (75, 150 mg· kg-1 per day) exerted dose-dependent anti-PF effect when administered orally or rectally but not intravenously, implying an intestinal route was involved in the action of curcumin. We speculated that curcumin might promote the generation of gut-derived factors and the latter acted as a mediator subsequently entering the lungs to ameliorate fibrosis. We showed that oral administration of curcumin indeed significantly increased the expression of gut-derived hepatocyte growth factor (HGF) in colon tissues. Furthermore, in bleomycin-treated mice, the upregulated protein level of HGF in lungs by oral curcumin was highly correlated with its anti-PF effect, which was further confirmed by coadministration of c-Met inhibitor SU11274. Curcumin (5-40 µM) dose-dependently increased HGF expression in primary mouse fibroblasts, macrophages, CCD-18Co cells (fibroblast cell line), and RAW264.7 cells (monocyte-macrophage cell line), but not in primary colonic epithelial cells. In CCD-18Co cells and RAW264.7 cells, curcumin dose-dependently activated PPARγ and CREB, whereas PPARγ antagonist GW9662 (1 µM) or cAMP response element (CREB) inhibitor KG-501 (10 µM) significantly decreased the boosting effect of curcumin on HGF expression. Finally, we revealed that curcumin dose-dependently increased the production of 15-deoxy-Δ12, 14-prostaglandin J2 (15d-PGJ2) in CCD-18Co cells and RAW264.7 cells, which was a common upstream of the two transcription factors. Moreover, both the in vitro and in vivo effects of curcumin were diminished by coadministration of HPGDS-inhibitor-1, an inhibitor of 15d-PGJ2 generation. Together, curcumin promotes the expression of HGF in colonic fibroblasts and macrophages by activating PPARγ and CREB via an induction of 15d-PGJ2, and the HGF enters the lungs giving rise to an anti-PF effect.


Assuntos
Colo/efeitos dos fármacos , Curcumina/uso terapêutico , Fator de Crescimento de Hepatócito/metabolismo , Prostaglandina D2/análogos & derivados , Fibrose Pulmonar/tratamento farmacológico , Administração Oral , Animais , Colo/citologia , Colo/metabolismo , Curcumina/administração & dosagem , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Feminino , Fibroblastos/metabolismo , Humanos , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos ICR , PPAR gama/metabolismo , Prostaglandina D2/metabolismo , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/patologia , Células RAW 264.7 , Regulação para Cima/efeitos dos fármacos
14.
Biol Reprod ; 104(1): 130-143, 2021 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-33112378

RESUMO

Female fertility depends greatly on the capacity of the uterus to recognize and eliminate microbial infections, a major reason of inflammation in the endometrium in many species. This study aimed to determine the in vitro effect of peroxisome proliferator-activated receptor gamma (PPARγ) ligands on the transcriptome genes expression and alternative splicing in the porcine endometrium in the mid-luteal phase of the estrous cycle during LPS-stimulated inflammation using RNA-seq technology. The endometrial slices were incubated in vitro in the presence of LPS and PPARγ agonists-PGJ2 or pioglitazone and antagonist-T0070907. We identified 222, 3, 4, and 62 differentially expressed genes after LPS, PGJ2, pioglitazone, or T0070907 treatment, respectively. In addition, we detected differentially alternative spliced events: after treatment with LPS-78, PGJ2-60, pioglitazone-52, or T0070907-134. These results should become a basis for further studies explaining the mechanism of PPARγ action in the reproductive system in pigs.


Assuntos
Endométrio/efeitos dos fármacos , Inflamação/metabolismo , PPAR gama/agonistas , Pioglitazona/farmacologia , Prostaglandina D2/análogos & derivados , Processamento Alternativo/efeitos dos fármacos , Animais , Benzamidas/farmacologia , Endométrio/metabolismo , Endométrio/patologia , Feminino , Perfilação da Expressão Gênica , Inflamação/induzido quimicamente , Inflamação/genética , Lipopolissacarídeos , PPAR gama/antagonistas & inibidores , PPAR gama/metabolismo , Prostaglandina D2/farmacologia , Piridinas/farmacologia , Suínos
15.
Eur J Pharmacol ; 892: 173733, 2021 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-33220278

RESUMO

Over the past decade, dexmedetomidine (DEX) has been found to possess an anti-inflammatory effect. However, the local anti-inflammatory mechanism of DEX has not been fully clarified. Some intracellular inflammatory pathways lead to negative feedback during the inflammatory process. The cyclooxygenase (COX) cascade synthesizes prostaglandins (PGs) and plays a key role in inflammation, but is known to also have anti-inflammatory properties through an alternative route of a PGD2 metabolite, 15-deoxy-delta-12,14-prostaglandin J2 (15d-PGJ2), and its receptor, peroxisome proliferator-activated receptor gamma (PPARγ). Therefore, we hypothesized that DEX inhibits LPS-induced inflammatory responses through 15d-PGJ2 and/or PPARγ activation, and evaluated the effects of DEX on these responses. The RAW264.7 mouse macrophage-like cells were pre-incubated with DEX, followed by the addition of LPS to induce inflammatory responses. Concentrations of TNFα, IL-6, PGE2, and 15d-PGJ2 in the supernatants of the cells were measured, and gene expressions of PPARγ and COX-2 were evaluated in the cells. Furthermore, we evaluated whether a selective α2 adrenoceptor antagonist, yohimbine or a selective PPARγ antagonist, T0070907, reversed the effects of DEX on the LPS-induced inflammatory responses. DEX inhibited LPS-induced TNFα, IL-6, and PGE2 productions and COX-2 mRNA expression, and the effects of DEX were reversed by yohimbine. On the other hand, DEX significantly increased 15d-PGJ2 production and PPARγ mRNA expression, and yohimbine reversed these DEX's effects. Furthermore, T0070907 reversed the anti-inflammatory effects of DEX on TNFα and IL-6 productions in the cells. These results suggest that DEX inhibits LPS-induced inflammatory responses through PPARγ activation following binding to α2 adrenoceptors.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/farmacologia , Anti-Inflamatórios/farmacologia , Dexmedetomidina/farmacologia , Inflamação/prevenção & controle , Macrófagos/efeitos dos fármacos , PPAR gama/agonistas , Receptores Adrenérgicos beta 2/efeitos dos fármacos , Agonistas de Receptores Adrenérgicos alfa 2/metabolismo , Animais , Anti-Inflamatórios/metabolismo , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Dexmedetomidina/metabolismo , Dinoprostona/metabolismo , Inflamação/induzido quimicamente , Inflamação/metabolismo , Interleucina-6/metabolismo , Lipopolissacarídeos/toxicidade , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , PPAR gama/genética , PPAR gama/metabolismo , Prostaglandina D2/análogos & derivados , Prostaglandina D2/metabolismo , Ligação Proteica , Células RAW 264.7 , Receptores Adrenérgicos beta 2/metabolismo , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo
16.
Int Immunopharmacol ; 84: 106565, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32388213

RESUMO

Natural or synthetic ligands for peroxisome proliferator-activated receptor gamma (PPAR-γ) represent an interesting tool for pharmacological interventions to treat inflammatory conditions. In particular, PPAR-γ activation prevents pain and inflammation in the temporomandibular joint (TMJ) by decreasing cytokine release and stimulating the synthesis of endogenous opioids. The goal of this study was to clarify whether PPAR-γ activation induces macrophage polarization, inhibiting inflammatory cytokine release and leukocyte recruitment. In addition, we investigated the involvement of heme oxygenase 1 (HO-1) in downstream events after PPAR-γ activation. Our results demonstrate that PPAR-γ activation ablates cytokine release by Bone Marrow-Derived Macrophages (BMDM) in vitro. 15d-PGJ2 induces the PPAR-γ heterodimer activation from rat macrophages, with macrophage polarization from M1-like cells toward M2-like cells. This response is mediated through HO-1. PPAR-γ activation diminished neutrophil migration induced by carrageenan, which was also HO-1 dependent. Ca2+/calmodulin expression did not change after PPAR-γ activation indicating that is not required for the activation of the intracellular L-arginine/NO/cGMP/K+ATP channel pathway. In summary, the anti-inflammatory actions induced by PPAR-γ activation involve macrophage polarization. HO-1 expression is increased and HO-1 activity is required for the suppression of neutrophil migration.


Assuntos
Heme Oxigenase-1/imunologia , Macrófagos/imunologia , Neutrófilos/fisiologia , PPAR gama/imunologia , Anilidas/farmacologia , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/imunologia , Carragenina/farmacologia , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Citocinas/imunologia , Humanos , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Neutrófilos/efeitos dos fármacos , Óxido Nítrico/imunologia , Prostaglandina D2/análogos & derivados , Prostaglandina D2/farmacologia , Ratos Wistar , Articulação Temporomandibular/efeitos dos fármacos , Articulação Temporomandibular/imunologia
17.
Blood ; 136(2): 235-246, 2020 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-32350523

RESUMO

Anemic stress induces stress erythropoiesis, which rapidly generates new erythrocytes to restore tissue oxygenation. Stress erythropoiesis is best understood in mice where it is extramedullary and occurs primarily in the spleen. However, both human and mouse stress erythropoiesis use signals and progenitor cells that are distinct from steady-state erythropoiesis. Immature stress erythroid progenitors (SEPs) are derived from short-term hematopoietic stem cells. Although the SEPs are capable of self-renewal, they are erythroid restricted. Inflammation and anemic stress induce the rapid proliferation of SEPs, but they do not differentiate until serum erythropoietin (Epo) levels increase. Here we show that rather than directly regulating SEPs, Epo promotes this transition from proliferation to differentiation by acting on macrophages in the splenic niche. During the proliferative stage, macrophages produce canonical Wnt ligands that promote proliferation and inhibit differentiation. Epo/Stat5-dependent signaling induces the production of bioactive lipid mediators in macrophages. Increased production of prostaglandin J2 (PGJ2) activates peroxisome proliferator-activated receptor γ (PPARγ)-dependent repression of Wnt expression, whereas increased production of prostaglandin E2 (PGE2) promotes the differentiation of SEPs.


Assuntos
Diferenciação Celular , Células Eritroides/metabolismo , Macrófagos/metabolismo , Receptores da Eritropoetina/metabolismo , Transdução de Sinais , Baço/metabolismo , Nicho de Células-Tronco , Animais , Dinoprostona/genética , Dinoprostona/metabolismo , Células Eritroides/citologia , Humanos , Macrófagos/citologia , Camundongos , Camundongos Transgênicos , PPAR gama/genética , PPAR gama/metabolismo , Prostaglandina D2/análogos & derivados , Prostaglandina D2/genética , Prostaglandina D2/metabolismo , Receptores da Eritropoetina/genética , Fator de Transcrição STAT5/genética , Fator de Transcrição STAT5/metabolismo , Baço/citologia
18.
Cell Rep ; 30(13): 4399-4417.e7, 2020 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-32234476

RESUMO

Oxidized lipids play a critical role in a variety of diseases with two faces: pro- and anti-inflammatory. The molecular mechanisms of this Janus-faced activity remain largely unknown. Here, we have identified that cyclopentenone-containing prostaglandins such as 15d-PGJ2 and structurally related oxidized phospholipid species possess a dual and opposing bioactivity in inflammation, depending on their concentration. Exposure of dendritic cells (DCs)/macrophages to low concentrations of such lipids before Toll-like receptor (TLR) stimulation instigates an anti-inflammatory response mediated by nuclear factor erythroid 2-related factor 2 (Nrf2)-dependent inhibition of nuclear factor κB (NF-κB) activation and downstream targets. By contrast, high concentrations of such lipids upon TLR activation of DCs/macrophages result in inflammatory apoptosis characterized by mitochondrial depolarization and caspase-8-mediated interleukin (IL)-1ß maturation independently of Nrf2 and the classical inflammasome pathway. These results uncover unexpected pro- and anti-inflammatory activities of physiologically relevant lipid species generated by enzymatic and non-enzymatic oxidation dependent on their concentration, a phenomenon known as hormesis.


Assuntos
Anti-Inflamatórios/farmacologia , Ciclopentanos/farmacologia , Inflamação/patologia , Prostaglandinas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Antígenos CD40/metabolismo , Caspase 8/metabolismo , Morte Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Inflamassomos/metabolismo , Inflamação/genética , Interleucinas/genética , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , NF-kappa B/metabolismo , Oxirredução , Fenótipo , Prostaglandina D2/análogos & derivados , Prostaglandina D2/química , Prostaglandina D2/farmacologia , Transdução de Sinais , Células Th1/efeitos dos fármacos , Receptores Toll-Like/metabolismo , Transcrição Gênica/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
19.
Am J Respir Crit Care Med ; 201(12): 1488-1498, 2020 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-32142372

RESUMO

Rationale: Aspirin-exacerbated respiratory disease is characterized by severe asthma, nonsteroidal antiinflammatory drug hypersensitivity, nasal polyposis, and leukotriene overproduction. Systemic corticosteroid therapy does not completely suppress lifelong aspirin hypersensitivity. Omalizumab efficacy against aspirin-exacerbated respiratory disease has not been investigated in a randomized manner.Objectives: To evaluate omalizumab efficacy against aspirin hypersensitivity, leukotriene E4 overproduction, and symptoms during an oral aspirin challenge in patients with aspirin-exacerbated respiratory disease using a randomized design.Methods: We performed a double-blind, randomized, crossover, placebo-controlled, single-center study at Sagamihara National Hospital between August 2015 and December 2016. Atopic patients (20-79 yr old) with aspirin-exacerbated respiratory disease diagnosed by systemic aspirin challenge were randomized (1:1) to a 3-month treatment with omalizumab or placebo, followed by a >18-week washout period (crossover design). The primary endpoint was the difference in area under logarithm level of urinary leukotriene E4 concentration versus time curve in the intent-to-treat population during an oral aspirin challenge.Measurements and Main Results: Sixteen patients completed the study and were included in the analysis. The area under the logarithm level of urinary leukotriene E4 concentration versus time curve during an oral aspirin challenge was significantly lower in the omalizumab phase (median [interquartile range], 51.1 [44.5-59.8]) than in the placebo phase (80.8 [interquartile range, 65.4-87.8]) (P < 0.001). Ten of 16 patients (62.5%) developed oral aspirin tolerance up to cumulative doses of 930 mg in the omalizumab phase (P < 0.001).Conclusions: Omalizumab treatment inhibited urinary leukotriene E4 overproduction and upper/lower respiratory tract symptoms during an oral aspirin challenge, resulting in aspirin tolerance in 62.5% of the patients with aspirin-exacerbated respiratory disease.


Assuntos
Antialérgicos/uso terapêutico , Anti-Inflamatórios não Esteroides/efeitos adversos , Aspirina/efeitos adversos , Asma Induzida por Aspirina/tratamento farmacológico , Omalizumab/uso terapêutico , Adulto , Idoso , Área Sob a Curva , Asma Induzida por Aspirina/etiologia , Asma Induzida por Aspirina/fisiopatologia , Asma Induzida por Aspirina/urina , Estudos Cross-Over , Método Duplo-Cego , Feminino , Volume Expiratório Forçado , Humanos , Leucotrieno E4/urina , Masculino , Pessoa de Meia-Idade , Prostaglandina D2/análogos & derivados , Prostaglandina D2/urina , Adulto Jovem
20.
Braz Oral Res ; 34: e013, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32074213

RESUMO

This study evaluated the effect of a cyclopentenone-type PG, 15-Deoxy-Δ12,14-PG J2 (15d-PGJ2), and lectin (ScLL) on the viability of human gingival fibroblasts (HGFs), and on IL-6 and TGFß-1 release by these fibroblasts, stimulated with lipopolysaccharide (LPS). HGFs were stimulated with LPS 10 µg/ml and treated with 15d-PGJ2 1 and 2 µg/ml, and ScLL 2 and 5 µg/ml, for 1 and 3h, and then evaluated for viability by MTT assay. Supernatant was collected to detect IL-6 and TGFß-1 release, by ELISA. Positive control was cells kept in Dulbecco's Modified Eagle's Medium, and negative control was those kept in LPS. Data were analyzed by ANOVA and Dunnett's test (α = 0.05). No significant difference was found in viability among experimental groups at 1h (p > 0.05). Percentage of ScLL 5 µg/ml viable cells was similar to that of positive control at evaluated periods (p > 0.05), whereas the other groups had lower levels than the positive control (p < 0.05). IL-6 release was statistically higher for ScLL 5 µg/ml and 15d-PGJ2 2 µg/ml at 1h, compared with the other treated groups and positive control (p < 0.05). No significant differences were found among the groups at 3h (p > 0.05), except for ScLL 2 µg/ml and 15d-PGJ2 1 µg/ml, which showed lower IL-6 release compared with that of negative control (p < 0.05). No significant difference was found among the groups for TGFß-1 release (p > 0.05). Results indicated that ScLL 5 µg/ml did not interfere in viability, and ScLL 2 µg/ml and 15d-PGJ2 1 µg/ml demonstrated reduced IL-6 release. Tested substances had no effect on TGFß-1 release.


Assuntos
Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Interleucina-6/metabolismo , Lipopolissacarídeos/farmacologia , Lectinas de Plantas/farmacologia , Prostaglandina D2/análogos & derivados , Fator de Crescimento Transformador beta1/metabolismo , Análise de Variância , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Gengiva/citologia , Humanos , Prostaglandina D2/farmacologia , Valores de Referência , Estatísticas não Paramétricas , Fatores de Tempo , Fator de Crescimento Transformador beta1/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA