Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Clin Invest ; 131(7)2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33529171

RESUMO

Tregs restrain both the innate and adaptive immune systems to maintain homeostasis. Allergic airway inflammation, characterized by a Th2 response that results from a breakdown of tolerance to innocuous environmental antigens, is negatively regulated by Tregs. We previously reported that prostaglandin I2 (PGI2) promoted immune tolerance in models of allergic inflammation; however, the effect of PGI2 on Treg function was not investigated. Tregs from mice deficient in the PGI2 receptor IP (IP KO) had impaired suppressive capabilities during allergic airway inflammatory responses compared with mice in which PGI2 signaling was intact. IP KO Tregs had significantly enhanced expression of immunoglobulin-like transcript 3 (ILT3) compared with WT Tregs, which may contribute to the impairment of the IP KO Treg's ability to suppress Th2 responses. Using fate-mapping mice, we reported that PGI2 signaling prevents Treg reprogramming toward a pathogenic phenotype. PGI2 analogs promoted the differentiation of naive T cells to Tregs in both mice and humans via repression of ß-catenin signaling. Finally, a missense variant in IP in humans was strongly associated with chronic obstructive asthma. Together, these data support that PGI2 signaling licenses Treg suppressive function and that PGI2 is a therapeutic target for enhancing Treg function.


Assuntos
Asma/imunologia , Reprogramação Celular/imunologia , Epoprostenol/imunologia , Tolerância Imunológica , Transdução de Sinais/imunologia , Linfócitos T Reguladores/imunologia , Animais , Asma/genética , Asma/patologia , Reprogramação Celular/genética , Doença Crônica , Epoprostenol/genética , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Receptores de Epoprostenol/genética , Receptores de Epoprostenol/imunologia , Transdução de Sinais/genética , Linfócitos T Reguladores/patologia
2.
Pharmacol Rev ; 72(4): 910-968, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32962984

RESUMO

Prostaglandins are derived from arachidonic acid metabolism through cyclooxygenase activities. Among prostaglandins (PGs), prostacyclin (PGI2) and PGE2 are strongly involved in the regulation of homeostasis and main physiologic functions. In addition, the synthesis of these two prostaglandins is significantly increased during inflammation. PGI2 and PGE2 exert their biologic actions by binding to their respective receptors, namely prostacyclin receptor (IP) and prostaglandin E2 receptor (EP) 1-4, which belong to the family of G-protein-coupled receptors. IP and EP1-4 receptors are widely distributed in the body and thus play various physiologic and pathophysiologic roles. In this review, we discuss the recent advances in studies using pharmacological approaches, genetically modified animals, and genome-wide association studies regarding the roles of IP and EP1-4 receptors in the immune, cardiovascular, nervous, gastrointestinal, respiratory, genitourinary, and musculoskeletal systems. In particular, we highlight similarities and differences between human and rodents in terms of the specific roles of IP and EP1-4 receptors and their downstream signaling pathways, functions, and activities for each biologic system. We also highlight the potential novel therapeutic benefit of targeting IP and EP1-4 receptors in several diseases based on the scientific advances, animal models, and human studies. SIGNIFICANCE STATEMENT: In this review, we present an update of the pathophysiologic role of the prostacyclin receptor, prostaglandin E2 receptor (EP) 1, EP2, EP3, and EP4 receptors when activated by the two main prostaglandins, namely prostacyclin and prostaglandin E2, produced during inflammatory conditions in human and rodents. In addition, this comparison of the published results in each tissue and/or pathology should facilitate the choice of the most appropriate model for the future studies.


Assuntos
Receptores de Prostaglandina E/metabolismo , Animais , Doenças Autoimunes/imunologia , Doenças Autoimunes/metabolismo , Dinoprostona/imunologia , Dinoprostona/metabolismo , Epoprostenol/imunologia , Epoprostenol/metabolismo , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Camundongos , Polimorfismo de Nucleotídeo Único , Multimerização Proteica , Ratos , Receptores de Prostaglandina E/química , Receptores de Prostaglandina E/genética , Receptores de Prostaglandina E/imunologia , Especificidade da Espécie
3.
EMBO J ; 39(14): e103454, 2020 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-32484988

RESUMO

The alarm cytokine interleukin-1ß (IL-1ß) is a potent activator of the inflammatory cascade following pathogen recognition. IL-1ß production typically requires two signals: first, priming by recognition of pathogen-associated molecular patterns leads to the production of immature pro-IL-1ß; subsequently, inflammasome activation by a secondary signal allows cleavage and maturation of IL-1ß from its pro-form. However, despite the important role of IL-1ß in controlling local and systemic inflammation, its overall regulation is still not fully understood. Here we demonstrate that peritoneal tissue-resident macrophages use an active inhibitory pathway, to suppress IL-1ß processing, which can otherwise occur in the absence of a second signal. Programming by the transcription factor Gata6 controls the expression of prostacyclin synthase, which is required for prostacyclin production after lipopolysaccharide stimulation and optimal induction of IL-10. In the absence of secondary signal, IL-10 potently inhibits IL-1ß processing, providing a previously unrecognized control of IL-1ß in tissue-resident macrophages.


Assuntos
Epoprostenol/imunologia , Interleucina-10/imunologia , Interleucina-1beta/imunologia , Macrófagos Peritoneais/imunologia , Animais , Epoprostenol/genética , Fator de Transcrição GATA6/genética , Fator de Transcrição GATA6/imunologia , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Interleucina-10/genética , Interleucina-1beta/genética , Macrófagos Peritoneais/patologia , Camundongos , Camundongos Transgênicos
4.
J Immunol ; 198(1): 461-471, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27895167

RESUMO

In allergic asthma, inhalation of airborne allergens such as the house dust mite (HDM) effectively activates both innate and adaptive immunity in the lung mucosa. To determine the role of the eicosanoid PGI2 and its receptor IP during allergic airway sensitization, HDM responses in mice lacking a functional IP receptor (i.e., PGI2 IP receptor-deficient [IP-/-]) were compared with wild type (WT) mice. Surprisingly, IP-/- mice had increased numbers of pulmonary CD3-NK1.1+Ly49b+ NK cells producing IFN-γ that was inversely associated with the number of type 2 innate lymphoid cells (ILC2s) expressing IL-33Rα and IL-13 compared with WT animals. This phenomenon was associated with elevated CX3CL1 levels in the airways of IP-/- mice and treatment with a neutralizing Ab to CX3CL1 reduced IFN-γ production by the lung NK cells. Remarkably, IP-/- mice were less responsive to HDM challenge than WT counterparts because intranasal instillation of the allergen induced markedly reduced levels of airway eosinophils, CD4+ lymphocyte infiltration, and mucus production, as well as depressed levels of CCL2 chemokine and Th2 cytokines. NK cells were responsible for such attenuated responses because depletion of NK1.1+ cells in IP-/- mice restored both the HDM-induced lung inflammation and ILC2 numbers, whereas transfer of CD3-NK1.1+ NK cells into the airways of WT hosts suppressed the inflammatory response. Collectively, these data demonstrate a hitherto unknown role for PGI2 in regulating the number and properties of NK cells resident in lung tissue and reveal a role for NK cells in limiting lung tissue ILC2s and preventing allergic inflammatory responses to inhaled HDM allergen.


Assuntos
Antígenos de Dermatophagoides/imunologia , Epoprostenol/imunologia , Células Matadoras Naturais/imunologia , Receptores de Epoprostenol/imunologia , Hipersensibilidade Respiratória/imunologia , Animais , Modelos Animais de Doenças , Citometria de Fluxo , Imuno-Histoquímica , Pulmão/citologia , Pulmão/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pyroglyphidae/imunologia
5.
Am J Reprod Immunol ; 73(6): 545-56, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25648617

RESUMO

PROBLEM: We investigated the effect of Xianziyizhen recipe capsule (XRC), a kidney-tonifying herb, on the PGI2-PPARδ signaling pathway at the maternal-fetal interface in embryo implantation dysfunction (EID) mice. METHOD OF STUDY: Intragastric administration of Progynova (estradiol) or XRC was performed in EID mouse model, following experimental induction of kidney deficiency by co-treatment with chemotherapy drug hydroxyurea and antiprogesterone mifepristone. The PPARδ and IL-11 mRNA expression in endometrium were detected by real-time relative reverse transcription-polymerase chain reaction (RT-PCR). Further, the protein expression of COX-2, PGI2, MMP-9, and TIMP-3 was detected in endometrial glandular epithelium and in stromal cells by immunohistochemical (IHC) assay. RESULTS: The results showed that hydroxyurea and mifepristone-induced EID were associated with significantly lower PPARδ and IL-11 mRNA levels in endometrium and reduced COX-2, PGI2, MMP-9, and TIMP-3 levels in endometrial glandular epithelium, compared with normal controls. However, XRC and Progynova treatment reversed these effects, leading to significant increases in PPARδ and IL-11 mRNA expression, and COX-2, PGI2, MMP-9 and TIMP-3 protein levels, when compared with the levels observed in EID mice. CONCLUSION: These results strongly suggested that XRC is beneficial in EID treatment and that XRC may mediate its effects through regulation of the PGI2-PPARδ signaling pathway.


Assuntos
Medicamentos de Ervas Chinesas/farmacologia , Implantação do Embrião/efeitos dos fármacos , Endométrio/imunologia , Epoprostenol/imunologia , Receptores Citoplasmáticos e Nucleares/imunologia , Animais , Ciclo-Oxigenase 2/biossíntese , Ciclo-Oxigenase 2/imunologia , Implantação do Embrião/imunologia , Endométrio/citologia , Células Epiteliais/citologia , Células Epiteliais/imunologia , Epoprostenol/biossíntese , Feminino , Interleucina-11/biossíntese , Interleucina-11/imunologia , Metaloproteinase 9 da Matriz/biossíntese , Metaloproteinase 9 da Matriz/imunologia , Camundongos , Gravidez , Receptores Citoplasmáticos e Nucleares/metabolismo , Células Estromais/citologia , Células Estromais/imunologia , Inibidor Tecidual de Metaloproteinase-3/biossíntese , Inibidor Tecidual de Metaloproteinase-3/imunologia
6.
J Immunol ; 189(11): 5402-10, 2012 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-23089395

RESUMO

Given that TLRs and sphingosine-1-phosphate (S1P) are key players in inflammation, we explored the potential interplay between TLRs and S1P in the adhesion/inflammatory pathways in primary human endothelial cells. As determined by Western blot and flow cytometry, cells treated with LPS (a TLR4 ligand) and S1P showed significantly enhanced expression of adhesion molecules such as ICAM-1 and E-selectin compared with the effect of either ligand alone. Cell-type differences on E-selectin upregulation were observed. In contrast, no cooperation effect on ICAM-1 or E-selectin was observed with a TLR2/TLR1 ligand. Consistent with an increase in adhesion molecule expression, endothelial cell treatment with LPS plus S1P significantly enhanced adhesion of PBMCs under shear stress conditions compared with the effect of either ligand alone and exhibited comparable levels of cell adhesion strength as those after TNF-α treatment. Moreover, LPS and S1P cooperated to increase the expression of proinflammatory molecules such as IL-6, cyclooxygenase-2, and prostacyclin, as determined by ELISA and Western blot. The analysis of signaling pathways revealed the synergistic phosphorylation of ERK upon LPS plus S1P treatment of HUVEC and human aortic endothelial cells and cell-type differences on p38 and NF-κB activation. Moreover, pharmacological and small interfering RNA experiments disclosed the involvement of S1P(1/3) and NF-κB in the cooperation effect and that cell origin determines the S1P receptors and signaling routes involved. Sphingosine kinase activity induction upon LPS plus S1P treatment suggests S1P- Sphingosine kinase axis involvement. In summary, LPS and S1P cooperate to increase proinflammatory molecules in endothelial cells and, in turn, to augment leukocyte adhesion, thus exacerbating S1P-mediated proadhesive/proinflammatory properties.


Assuntos
Endotélio Vascular/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Inflamação/imunologia , Lipopolissacarídeos/farmacologia , Lisofosfolipídeos/farmacologia , Esfingosina/análogos & derivados , Adesão Celular/efeitos dos fármacos , Células Cultivadas , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/imunologia , Sinergismo Farmacológico , Selectina E/genética , Selectina E/imunologia , Endotélio Vascular/imunologia , Endotélio Vascular/patologia , Epoprostenol/genética , Epoprostenol/imunologia , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/imunologia , Regulação da Expressão Gênica/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/imunologia , Células Endoteliais da Veia Umbilical Humana/patologia , Humanos , Inflamação/genética , Inflamação/patologia , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/imunologia , Interleucina-6/genética , Interleucina-6/imunologia , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/patologia , NF-kappa B/genética , NF-kappa B/imunologia , Especificidade de Órgãos , Transdução de Sinais/efeitos dos fármacos , Esfingosina/farmacologia , Receptores Toll-Like/genética , Receptores Toll-Like/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/imunologia
7.
PLoS One ; 7(5): e33518, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22590492

RESUMO

BACKGROUND: Prostaglandin I(2) (PGI(2)), a lipid mediator currently used in treatment of human disease, is a critical regulator of adaptive immune responses. Although PGI(2) signaling suppressed Th1 and Th2 immune responses, the role of PGI(2) in Th17 differentiation is not known. METHODOLOGY/PRINCIPAL FINDINGS: In mouse CD4(+)CD62L(+) naïve T cell culture, the PGI(2) analogs iloprost and cicaprost increased IL-17A and IL-22 protein production and Th17 differentiation in vitro. This effect was augmented by IL-23 and was dependent on PGI(2) receptor IP signaling. In mouse bone marrow-derived CD11c(+) dendritic cells (BMDCs), PGI(2) analogs increased the ratio of IL-23/IL-12, which is correlated with increased ability of BMDCs to stimulate naïve T cells for IL-17A production. Moreover, IP knockout mice had delayed onset of a Th17-associated neurological disease, experimental autoimmune encephalomyelitis (EAE), and reduced infiltration of IL-17A-expressing mononuclear cells in the spinal cords compared to wild type mice. These results suggest that PGI(2) promotes in vivo Th17 responses. CONCLUSION: The preferential stimulation of Th17 differentiation by IP signaling may have important clinical implications as PGI(2) and its analogs are commonly used to treat human pulmonary hypertension.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Encefalomielite Autoimune Experimental/tratamento farmacológico , Epoprostenol/farmacologia , Inibidores da Agregação Plaquetária/farmacologia , Receptores de Epoprostenol/imunologia , Células Th17/imunologia , Animais , Antineoplásicos/imunologia , Antineoplásicos/farmacologia , Diferenciação Celular/imunologia , Células Cultivadas , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Epoprostenol/análogos & derivados , Epoprostenol/genética , Epoprostenol/imunologia , Feminino , Humanos , Iloprosta/imunologia , Iloprosta/farmacologia , Interleucina-12/genética , Interleucina-12/imunologia , Interleucina-17/genética , Interleucina-17/imunologia , Interleucina-23/genética , Interleucina-23/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Inibidores da Agregação Plaquetária/imunologia , Receptores de Epoprostenol/genética , Medula Espinal/imunologia , Medula Espinal/patologia , Células Th17/patologia
8.
Cell Immunol ; 273(2): 109-14, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22321156

RESUMO

Follicular dendritic cells (FDCs) are an essential cellular component of the germinal center (GC) and are believed to exert regulatory effects on the various stages of GC reactions. According to our previous reports, human FDCs express prostacyclin synthase, and prostacyclin analogues augment adhesion and co-stimulatory molecules on the surface of activated B cells. These findings prompted us to investigate whether FDCs would contribute to the antigen-presenting capability of B cells by using the well-established FDC-like cells, HK cells, and tonsillar B cells. Our results show that HK cells significantly enhance the expression levels of CD54, CD80, and CD86 on the surface of activated B cells. The enhancing effect of HK cells on CD86 is impeded by indomethacin and an EP4 antagonist, implying that a certain prostaglandin is mediating the up-regulation. Prostacyclin indeed recapitulates the enhancing effect on CD86, which is inhibited by EP4 as well as IP antagonists. B cells co-cultured with HK cells exhibit an augmented APC activity, which is inhibited by CD86 neutralization. These results reveal another unrecognized function of human FDC.


Assuntos
Linfócitos B/imunologia , Antígeno B7-2/imunologia , Células Dendríticas Foliculares/imunologia , Imunidade Inata , Apresentação de Antígeno , Linfócitos B/citologia , Linfócitos B/efeitos dos fármacos , Antígeno B7-2/antagonistas & inibidores , Antígeno B7-2/genética , Antígeno B7-2/metabolismo , Adesão Celular/imunologia , Comunicação Celular/genética , Comunicação Celular/imunologia , Técnicas de Cocultura , Sistema Enzimático do Citocromo P-450/imunologia , Sistema Enzimático do Citocromo P-450/metabolismo , Células Dendríticas Foliculares/citologia , Células Dendríticas Foliculares/efeitos dos fármacos , Epoprostenol/imunologia , Epoprostenol/metabolismo , Citometria de Fluxo , Expressão Gênica , Humanos , Indometacina/farmacologia , Oxirredutases Intramoleculares/imunologia , Oxirredutases Intramoleculares/metabolismo , Ativação Linfocitária , Tonsila Palatina/citologia , Tonsila Palatina/imunologia , Cultura Primária de Células , Antagonistas de Prostaglandina/farmacologia , Receptores de Prostaglandina E Subtipo EP4/antagonistas & inibidores , Receptores de Prostaglandina E Subtipo EP4/imunologia , Linfócitos T/citologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia
9.
Prostaglandins Other Lipid Mediat ; 96(1-4): 21-6, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21864703

RESUMO

Prostaglandin (PG)I(2) has important regulatory functions on the innate and adaptive immune systems. Recent experimental evidence reveals that PGI(2) modulates the development and function of CD4+ T cells subsets, including Th1, Th2, and Th17 cell responses. In vitro and in vivo studies support that PGI(2) generally has an inhibitory effect on Th1 and Th2 activation, differentiation, and cytokine production. In contrast, PGI(2) seems to enhance Th17-favoring polarization conditions, resulting in Th17 cytokine production. Therefore, PGI(2) may either promote or inhibit individual CD4+ cell subsets and impact adaptive immune responses.


Assuntos
Diferenciação Celular/imunologia , Epoprostenol/imunologia , Imunidade Inata , Transdução de Sinais/imunologia , Células Th1/imunologia , Células Th17/imunologia , Células Th2/imunologia , Imunidade Adaptativa/efeitos dos fármacos , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Citocinas/biossíntese , Epoprostenol/análogos & derivados , Epoprostenol/metabolismo , Epoprostenol/farmacologia , Humanos , Iloprosta/farmacologia , Imunidade Inata/efeitos dos fármacos , Ativação Linfocitária/efeitos dos fármacos , Camundongos , Camundongos Knockout , Prostaglandinas Sintéticas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Células Th1/citologia , Células Th1/efeitos dos fármacos , Células Th17/citologia , Células Th17/efeitos dos fármacos , Células Th2/citologia , Células Th2/efeitos dos fármacos , Vasodilatadores/farmacologia
10.
Am J Reprod Immunol ; 64(6): 376-83, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20482519

RESUMO

PROBLEM: To evaluate whether impaired endothelial function and endothelial inflammatory response occur in parallel in the women with preeclampsia. METHOD OF STUDY: Venous blood was drawn from normal (n=40) and severe preeclamptic (sPE) (n=40) pregnant women when they were admitted to the L&D Unit and 24 hrs after delivery. Plasma and serum samples were extracted and measured for 6-keto PGF1α and TXB(2) (stable metabolites of PGI2 and TXA2), and intercellular adhesion molecule (ICAM) and vascular cell adhesion molecule (VCAM) by ELISA. Data are analyzed by Mann-Whitney test and paired t-test. The statistical significance is set as P<0.05. Results Plasma 6-keto PGF1α levels were significantly reduced at admission and 24hr after delivery in sPE compared to normal pregnant controls, P<0.01. The ratio of 6-keto PGF1α and TXB(2) was significant less in sPE than that in normal pregnant controls before delivery. There was no significant difference for ICAM and VCAM levels between normal and patients with sPE before and after delivery. CONCLUSION: Maternal 6-keto PGF1α levels and the ratio of 6-keto PGF1α and TXB(2) were decreased in patients with sPE compared to normal pregnant controls. In contrast, maternal ICAM and VCAM levels were not different between the two groups. These data suggest that serum ICAM and VCAM levels may not be sensitive inflammatory biomarkers for preeclampsia.


Assuntos
Biomarcadores/metabolismo , Endotélio/metabolismo , Epoprostenol/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Pré-Eclâmpsia/imunologia , Tromboxano A2/metabolismo , Molécula 1 de Adesão de Célula Vascular/metabolismo , Adolescente , Adulto , Endotélio/imunologia , Epoprostenol/análogos & derivados , Epoprostenol/genética , Epoprostenol/imunologia , Feminino , Regulação da Expressão Gênica/imunologia , Humanos , Inflamação , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/imunologia , Pré-Eclâmpsia/genética , Pré-Eclâmpsia/metabolismo , Pré-Eclâmpsia/fisiopatologia , Gravidez , Tromboxano A2/análogos & derivados , Tromboxano A2/genética , Tromboxano A2/imunologia , Molécula 1 de Adesão de Célula Vascular/genética , Molécula 1 de Adesão de Célula Vascular/imunologia
11.
J Immunol ; 183(11): 6981-8, 2009 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-19915063

RESUMO

Increasing evidence indicates that pulmonary arterial hypertension is a vascular inflammatory disease. Prostacyclin (PGI(2)) is widely used to treat pulmonary arterial hypertension and is believed to benefit patients largely through vasodilatory effects. PGI(2) is also increasingly believed to have anti-inflammatory effects, including decreasing leukocyte cytokine production, yet few mechanistic details exist to explain how these effects are mediated at the transcriptional level. Because activated monocytes are critical sources of MCP-1 and other cytokines in cardiovascular inflammation, we examined the effects of iloprost on IFN-gamma- and IL-6-stimulated cytokine production in human monocytes. We found that iloprost inhibited IFN-gamma- and IL-6-induced MCP-1, IL-8, RANTES, and TNF-alpha production in monocytes, indicating wide-ranging anti-inflammatory action. We found that activation of STAT1 was critical for IFN-gamma-induced MCP-1 production and demonstrated that iloprost inhibited STAT1 activation by several actions as follows: 1) iloprost inhibited the phosphorylation of STAT1-S727 in the transactivation domain, thereby reducing recruitment of the histone acetylase and coactivator CBP/p300 to STAT1; 2) iloprost selectively inhibited activation of JAK2 but not JAK1, both responsible for activation of STAT1 via phosphorylation of STAT1-Y701, resulting in reduced nuclear recruitment and activation of STAT1; and 3) SOCS-1, which normally terminates IFN-gamma-signaling, was not involved in iloprost-mediated inhibition of STAT1, indicating divergence from the classical pathway for terminating IFN-gamma-signaling. We conclude that PGI(2) exerts anti-inflammatory action by inhibiting STAT1-induced cytokine production, in part by targeting the transactivation domain-induced recruitment of the histone acetylase CBP/p300.


Assuntos
Anti-Inflamatórios/farmacologia , Epoprostenol/farmacologia , Monócitos/efeitos dos fármacos , Fator de Transcrição STAT1/metabolismo , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Fatores de Transcrição de p300-CBP/metabolismo , Western Blotting , Citocinas/biossíntese , Citocinas/efeitos dos fármacos , Citocinas/imunologia , Ensaio de Desvio de Mobilidade Eletroforética , Epoprostenol/análogos & derivados , Epoprostenol/imunologia , Regulação da Expressão Gênica/imunologia , Humanos , Iloprosta/farmacologia , Imunoprecipitação , Interferon gama/imunologia , Interferon gama/metabolismo , Monócitos/imunologia , Monócitos/metabolismo , RNA Interferente Pequeno , Fator de Transcrição STAT1/imunologia , Transdução de Sinais/imunologia , Proteína 1 Supressora da Sinalização de Citocina , Proteínas Supressoras da Sinalização de Citocina/imunologia , Fatores de Transcrição de p300-CBP/imunologia
13.
Respirology ; 8(4): 467-72, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14629650

RESUMO

OBJECTIVE: Cytokines have been implicated in the pathophysiology of pulmonary hypertension. We sought to explore the possibility that prostacyclin is a link. METHODOLOGY: We tested the effects of the cytokines interleukin-1beta (IL-1beta), tumour necrosis factor-alpha (TNF-alpha), and macrophage migration inhibitory factor (MIF) on arachidonic acid metabolism of pulmonary artery smooth muscle cells (PASMCs) with special regard to prostacyclin (PGI2) that protects against pulmonary hypertension. Cultured rat PASMCs were treated with IL-1beta, TNF-alpha, or MIF. Expression of prostacyclin synthase (PGIS) and cyclooxygenase-2 (COX-2) mRNAs, and PGI2 synthesis, were measured. RESULTS: We found that PGIS mRNA expression was suppressed by high concentrations of TNF-alpha and MIF, while COX-2 mRNA was induced by all three cytokines tested. IL-1beta increased PGI2 production in a dose-dependent manner. TNF-alpha and MIF also increased PGI2 production, but to a far lesser degree at high concentrations. TNF-alpha paradoxically decreased PGI2 production at a low concentration. CONCLUSIONS: These results suggest that TNF-alpha and MIF are potentially antagonistic to the action of PGI2 in rat PASMCs via down-regulation of PGIS mRNA. Simultaneous induction of COX-2 mRNA may further counteract the action of PGI2 by increasing the levels of eicosanoids other than PGI2.


Assuntos
Epoprostenol/biossíntese , Interleucina-1/metabolismo , Fatores Inibidores da Migração de Macrófagos/metabolismo , Músculo Liso Vascular/imunologia , Miócitos de Músculo Liso/imunologia , Fator de Necrose Tumoral alfa/metabolismo , 6-Cetoprostaglandina F1 alfa/metabolismo , Análise de Variância , Animais , Células Cultivadas , Ciclo-Oxigenase 2 , Sistema Enzimático do Citocromo P-450/metabolismo , Epoprostenol/imunologia , Oxirredutases Intramoleculares/metabolismo , Isoenzimas/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Prostaglandina-Endoperóxido Sintases/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley
14.
Nurs Times ; 99(16): 30-2, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12739281

RESUMO

Non-steroidal anti-inflammatory drugs (NSAIDs) are among the most widely used of all therapeutic agents. Although they are effective analgesics they do have significant adverse side-effects. Before treatment is started the prescriber should always weigh efficacy against possible side-effects. NSAIDs vary in their selectivity for inhibiting the two types of cyclo-oxygenase. An NSAID should therefore be chosen on the basis of the incidence of associated side-effects.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Seleção de Pacientes , Anti-Inflamatórios não Esteroides/efeitos adversos , Transtornos Plaquetários/induzido quimicamente , Ciclo-Oxigenase 1 , Ciclo-Oxigenase 2 , Dinoprostona/imunologia , Epoprostenol/imunologia , Hemorragia Gastrointestinal/induzido quimicamente , Humanos , Mediadores da Inflamação/imunologia , Isoenzimas/efeitos dos fármacos , Isoenzimas/imunologia , Nefropatias/induzido quimicamente , Proteínas de Membrana , Prostaglandina-Endoperóxido Sintases/efeitos dos fármacos , Prostaglandina-Endoperóxido Sintases/imunologia , Fatores de Risco , Tromboxano A2/imunologia
15.
Anal Chem ; 74(15): 3892-8, 2002 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-12175181

RESUMO

This work describes a solid-phase immunoassay for 6-keto-prostaglandin F1alpha, the stable hydrolysis product of prostacyclin (prostaglandin I2). Prostacyclin, a potent vasodilator with antiplatelet and antiproliferative properties is an effective treatment for primary pulmonary hypertension and pulmonary arterial hypertension associated with scleroderma and scleroderma-like syndrome. Levels of 6-keto-prostaglandin F1alpha can be directly correlated with levels of prostacyclin. Therefore, 6-keto-prostaglandin F1alpha, has become the indicator of choice to measure prostacyclin levels. The single-step immunoassay for 6-keto-prostaglandin F1alpha reported here was developed using the bioluminescent protein aequorin as a label. Analyte-label conjugates were constructed by linking the carboxyl group of 6-keto-prostaglandin F1alpha and lysine residues of aequorin by chemical conjugation methods. The binding properties of 6-keto-prostaglandin F1alpha toward its antibody and the bioluminescent properties of aequorin were retained in the conjugate, which was then used to generate a dose-response curve for the analyte in a convenient microtiter plate format. The concentration of 6-keto-prostaglandin F1alpha after extraction from plasma showed good correlation with the concentration of 6-ketoprostaglandin F1alpha obtained without prior extraction of the same plasma sample. This measurement demonstrated that the assay allows the measurement of 6-keto-prostaglandin F1alpha directly in plasma without any pretreatment of the samples, which results in a much simpler method with a faster assay time.


Assuntos
Anti-Hipertensivos/sangue , Monitoramento de Medicamentos/métodos , Epoprostenol/sangue , 6-Cetoprostaglandina F1 alfa/sangue , 6-Cetoprostaglandina F1 alfa/imunologia , Equorina , Anti-Hipertensivos/farmacocinética , Estudos de Casos e Controles , Reações Cruzadas , Monitoramento de Medicamentos/normas , Epoprostenol/imunologia , Epoprostenol/farmacocinética , Humanos , Hipertensão Pulmonar/tratamento farmacológico , Imunoensaio/métodos , Proteínas Luminescentes , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
16.
Crit Care Med ; 29(7): 1445-51, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11445706

RESUMO

OBJECTIVE: This study evaluated whether prostacyclin is a necessary mediator of inflammation in graded bacteremia or is sufficient alone in pathophysiologic concentrations to cause the pulmonary derangement of bacteremic shock. DESIGN: Experimental. SETTING: Laboratory. SUBJECTS: Twenty-three anesthetized adult swine. INTERVENSIONS: Swine were studied in four groups for 4 hrs: a) an anesthesia control group (n = 6); b) a septic control group (n = 6), in which 1010/mL Aeromonas hydrophila was infused intravenously at 0.2 mL.kg-1.hr-1 and increased to 4.0 mL.kg-1.hr-1 over 3 hrs; c) a prostacyclin infusion group (n = 6), which received prostacyclin infusion to match septic control plasma concentrationsclm without bacteremia; and d) an antiprostacyclin antibody group (n = 5), which received continuous Aeromonas hydrophila infusion plus antiprostacyclin antibody infusion. MEASUREMENTS AND MAIN RESULTS: Pulmonary hemodynamics, arterial blood gases, and plasma concentrations of arachidonate metabolites were measured hourly over a 4-hr period. In the septic control group and antiprostacyclin antibody group, elevated pulmonary vascular resistance index and pulmonary artery pressure with decreased Pao2, as well as lower pH, were documented after 1 and 3 hrs of graded bacteremia compared with the anesthesia control group and prostacyclin infusion group (p <.05). Thromboxane B2 concentration increased significantly in all groups during septic shock. In the antiprostacyclin antibody group, leukotriene B4 increased immediately after starting antiprostacyclin antibody infusion and reached significance at 3 hrs compared with the septic control group (p <.05). The prostacyclin infusion group had consistently lower concentrations of leukotrienes C4, D4, and E4 than all other groups. CONCLUSIONS: Prostacyclin does not mediate blood gas changes, alterations of pulmonary hemodynamics, or platelet abnormalities in porcine septic shock, because antiprostacyclin antibody infusion did not change the pulmonary hypertension and hypoxemia, and infusion of prostacyclin to pathophysiologic blood concentrations did not reproduce such changes. Antiprostacyclin blockade during bacteremia significantly increased concentrations of leukotrienes C4, D4, and E4 and leukotriene B4, whereas prostacyclin infusion suppressed concentrations of leukotrienes C4, D4, and E4, suggesting that endogenous prostacyclin may blunt leukotriene release.


Assuntos
Anti-Hipertensivos/imunologia , Bacteriemia/fisiopatologia , Epoprostenol/imunologia , Pneumopatias/imunologia , Choque Séptico/fisiopatologia , 6-Cetoprostaglandina F1 alfa/sangue , Análise de Variância , Animais , Anti-Hipertensivos/farmacologia , Epoprostenol/farmacologia , Infecções por Bactérias Gram-Negativas/fisiopatologia , Hemodinâmica , Hipertensão Pulmonar/imunologia , Leucotrieno B4/sangue , Análise por Pareamento , Troca Gasosa Pulmonar/imunologia , Síndrome do Desconforto Respiratório/imunologia , SRS-A/sangue , Suínos , Tromboxano B2/sangue
17.
Shock ; 13(6): 478-84, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10847636

RESUMO

This study evaluated whether or not prostacyclin (PGI2) was necessary or sufficient by itself in a pathophysiologic concentration to mediate the cardiovascular dysfunction of septic shock. Anesthetized adult swine received anesthesia only (ANESTHESIA CONTROL, n = 6); graded Aeromonas hydrophila, 10(10)/mL, infusion at 0.2 mL/kg/h that increased to 4.0 mL/kg/h over 3 h (SEPTIC SHOCK CONTROL, n = 6); pathophysiologic prostacyclin infusion to match septic shock control plasma levels without bacteremia (PGI2 INFUSION, n = 6), or graded Aeromonas hydrophila plus anti-prostacyclin antibody infusion (ANTI-PGI2-Ab INFUSION, n = 5). This graded porcine bacteremia model was 100% lethal after 4 h. Cardiovascular hemodynamics, arterial blood gases, and plasma levels of arachidonate metabolites were measured at baseline and hourly over a 4-h period. The results showed that PGI2 was not a necessary mediator of impaired cardiovascular hemodynamics in graded bacteremia, as anti-PGI2 antibody infusion did not improve the cardiac index, systemic vascular resistance, or peripheral oxygen balance in septic animals. Also, PGI2 was not sufficient alone to cause the cardiovascular dysfunction of sepsis, as pathophysiologic infusion of PGI2 did not reproduce such changes in normal animals. PGI2 blockade during bacteremia significantly increased LTC4D4E4, and LTB4 whereas PGI2 infusion suppressed LTC4D4E4 concentration, suggesting that endogenous PGI2 may blunt leukotriene release during septic shock. These results indicate a complex dynamic equilibrium among prostacyclin and leukotrienes in septic shock.


Assuntos
Epoprostenol/toxicidade , Leucotrieno C4/metabolismo , Leucotrieno D4/metabolismo , Leucotrieno E4/metabolismo , Choque Séptico/fisiopatologia , 6-Cetoprostaglandina F1 alfa/sangue , Aeromonas hydrophila , Animais , Anticorpos/imunologia , Anticorpos/farmacologia , Anticorpos/uso terapêutico , Bacteriemia/etiologia , Epoprostenol/administração & dosagem , Epoprostenol/imunologia , Infecções por Bactérias Gram-Negativas/complicações , Choque Séptico/etiologia , Choque Séptico/metabolismo , Suínos , Tromboxano B2/sangue
18.
Clin Exp Hypertens ; 22(2): 181-92, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10744358

RESUMO

This study was designed to examine the contribution of lipoxygenase products to mechanisms of vascular contraction and elevated blood pressure in rats with aortic coarctation-induced hypertension. In cytosolic fractions of aortae taken from hypertensive rats, 12-lipoxygenase protein was increased as compared to normotensive controls. Aortic rings from hypertensive, but not from normotensive rats, exhibited a basal tone which was reduced 74+/-12 and 71+/-22%, respectively, by the lipoxygenase inhibitors cinnamyl-3,4-dihydroxy-alpha-cyanocinnamate (CDC, 10(-5) mol/L) and 5,8,11-eicosatriynoic acid (ETI, 10(-5) mol/L). CDC (8 mg/kg s.c.) did not affect the blood pressure of normotensive rats but decreased that of hypertensive rats from 182+/-6 to 151+/-10 mm Hg. The blood pressure lowering effect of CDC was blunted in hypertensive rats pretreated with indomethacin or antibodies against 5,6-dihydro-prostaglandin I2. These data suggest contribution of lipoxygenase-derived products to mechanisms underlying aortic smooth muscle basal tone and elevated blood pressure in rats with aortic coarctation-induced hypertension. The vasodepressor effect of CDC depends on a mechanism involving vasodilatory prostaglandins.


Assuntos
Hipertensão/enzimologia , Lipoxigenase/metabolismo , Ácido 12-Hidroxi-5,8,10,14-Eicosatetraenoico/farmacologia , Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Ácido 8,11,14-Eicosatrienoico/farmacologia , Animais , Aorta Torácica/efeitos dos fármacos , Aorta Torácica/enzimologia , Aorta Torácica/fisiopatologia , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/fisiologia , Ácidos Cafeicos/farmacologia , Inibidores de Ciclo-Oxigenase/farmacologia , Modelos Animais de Doenças , Epoprostenol/análogos & derivados , Epoprostenol/imunologia , Epoprostenol/farmacologia , Hipertensão/etiologia , Hipertensão/fisiopatologia , Hipertensão/prevenção & controle , Indometacina/farmacologia , Leucotrienos/farmacologia , Lipoxigenase/efeitos dos fármacos , Inibidores de Lipoxigenase/farmacologia , Masculino , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/enzimologia , Músculo Liso Vascular/fisiopatologia , Prostaglandinas Sintéticas/imunologia , Prostaglandinas Sintéticas/farmacologia , Ratos , Ratos Sprague-Dawley , Vasoconstrição/efeitos dos fármacos , Vasoconstrição/fisiologia , Vasoconstritores/farmacologia
19.
Am J Physiol Heart Circ Physiol ; 278(5): H1717-24, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10775153

RESUMO

Potential long-term cardioprotection was investigated in an extensive experimental study. Lactobacillus cultivation components (LCC) were administered intravenously in anesthetized rats 1, 7, and 21 days before global ischemia (GI). GI was produced by full stop flow in isolated Langendorff-perfused hearts for 20 min and was followed by reperfusion. Control animals were injected with saline. LCC reduced reperfusion tachyarrhythmia significantly and improved functional recovery of the ischemized rat heart. These beneficial effects were associated with reduction of release of norepinephrine (NE) and prostacyclin at the first minute of reperfusion, activation of myocardial catalase, and overexpression of 70-kDa heat stress protein (HSP-70) at ischemia and reperfusion (P < 0.05). This cardioprotection was documented up to 21 days after a single injection of LCC. Thus Lactobacillus cultivation components are new nontoxic materials that produce marked long-term cardioprotection against ischemia-reperfusion damage. This effect is attributed to an activation of the cellular defense system, manifested by activation of the antioxidant pathway and by expression of protective proteins. NE is involved in this process, and the data also suggest a role for prostacyclin in this model of cardioprotection. The potential of LCC and related compounds working through similar mechanisms in the prevention and therapy of various ischemic heart syndromes should be explored.


Assuntos
Fármacos Cardiovasculares/administração & dosagem , Lactobacillus , Preparações Farmacêuticas/administração & dosagem , Traumatismo por Reperfusão/prevenção & controle , Trifosfato de Adenosina/metabolismo , Animais , Temperatura Corporal/efeitos dos fármacos , Fármacos Cardiovasculares/análise , Catalase/metabolismo , Circulação Coronária/efeitos dos fármacos , Epoprostenol/imunologia , Epoprostenol/metabolismo , Proteínas de Choque Térmico HSP70/biossíntese , Hemodinâmica/efeitos dos fármacos , Ácido Láctico/metabolismo , Lactobacillus/química , Lactobacillus/imunologia , Masculino , Isquemia Miocárdica/enzimologia , Isquemia Miocárdica/fisiopatologia , Norepinefrina/metabolismo , Preparações Farmacêuticas/análise , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica/efeitos dos fármacos , Traumatismo por Reperfusão/fisiopatologia , Taquicardia/tratamento farmacológico , Taquicardia/prevenção & controle
20.
Blood ; 84(11): 3724-33, 1994 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-7949128

RESUMO

Platelet adhesion to purified surface-immobilized fibronectin under flow conditions was investigated. Fibronectin was found to support attachment and spreading of platelets. The extent of platelet spreading depended on the amount of immobilized fibronectin. An antiglycoprotein (anti-GP) IIb/IIIa antibody and an Arg-Gly-Asp (RGD)-containing peptide inhibited adhesion almost completely, whereas antibodies directed against platelet GP Ic/IIa (very late antigen 5) inhibited by 50%. Similar results with the antibodies and the peptide were found in a static system. A comparison of different anticoagulants showed no difference in adhesion using citrate or hirudin. However, unfractionated heparin (UFH) or low-molecular-weight heparin (LMWH) as the only anticoagulant or in combination with citrate maximally inhibited adhesion by 80% and 60%, respectively. Preincubation of the immobilized fibronectin with UFH resulted in a maximal inhibition of 90%, whereas preincubation with LMWH had no effect. When we preincubated the surface with heparins of different size, we observed 40% inhibition of adhesion with heparins with an average MW of up to 18 kD, whereas a heparin with an average MW of 21 kD almost completely blocked adhesion. These results indicate that platelet adhesion to fibronectin in flow involves several receptors, is highly RGD-mediated, does not require physiologic levels of divalent cations, and can be inhibited by direct binding of heparin to the fibronectin surface.


Assuntos
Fibronectinas/metabolismo , Heparina/farmacologia , Adesividade Plaquetária , Glicoproteínas da Membrana de Plaquetas/fisiologia , Receptores de Fibronectina/fisiologia , Sequência de Aminoácidos , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Citratos/farmacologia , Ácido Cítrico , Endotélio Vascular/citologia , Epoprostenol/imunologia , Epoprostenol/fisiologia , Matriz Extracelular/metabolismo , Vidro , Heparina de Baixo Peso Molecular/farmacologia , Hirudinas/farmacologia , Humanos , Dados de Sequência Molecular , Oligopeptídeos/farmacologia , Glicoproteínas da Membrana de Plaquetas/imunologia , Receptores de Fibronectina/imunologia , Estresse Mecânico , Propriedades de Superfície , Trombastenia/sangue , Veias Umbilicais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...