Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
JCI Insight ; 2024 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-39298269

RESUMEN

Tumor cell-derived prostaglandin E2 (PGE2) is a tumor cell-intrinsic factor that supports immunosuppression in the tumor microenvironment (TME) by acting on the immune cells, but the impact of PGE2 signaling in tumor cells on immunosuppressive TME is unclear. We demonstrate that deleting the PGE2 synthesis enzyme or disrupting autocrine PGE2 signaling through EP4 receptors on tumor cells reverses the T cell-low, myeloid cell-rich TME, activates T cells, and suppresses tumor growth. Knockout (KO) of Ptges (the gene encoding PGE2 synthesis enzyme mPGES-1) or the EP4 receptor gene (Ptger4) in KPCY (KrasG12D/P53R172H/Yfp/CrePdx) pancreatic tumor cells abolished growth of implanted tumors in a T cell-dependent manner. Blockade of the EP4 receptor in combination with immunotherapy, but not immunotherapy alone, induced complete tumor regressions and immunological memory. Mechanistically, Ptges and Ptger4 KO tumor cells exhibited altered T and myeloid cell attractant chemokines, became more susceptible to TNF-α killing, and exhibited reduced adenosine synthesis. In hosts treated with an adenosine deaminase inhibitor, Ptger4 KO tumor cells accumulated adenosine and gave rise to tumors. These studies reveal an unexpected finding - a non-redundant role for the autocrine mPGES1-PGE2-EP4 signaling axis in pancreatic cancer cells - further nominating mPGES-1 inhibition and EP4 blockade as immune-sensitizing therapy in cancer.

2.
Front Pharmacol ; 14: 1224623, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37388454
3.
Children (Basel) ; 9(7)2022 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-35884020

RESUMEN

The sudden health and economic crisis brought about by the COVID-19 pandemic affords an opportunity to examine the impact of economic disruption to children and families. Any negative effects on the well-being of children are important to consider in relation to both short- and long-term outcomes. Using pre-pandemic and mid-pandemic waves of the longitudinal Growing Up in Ireland study, we examined whether the impact of economic disruption was equivalent for families who were (or were not) financially vulnerable pre-pandemic. We then investigated whether economic disruption was associated with a negative effect on the emotional well-being of 12-year-olds, and if there was evidence for such a negative effect being mediated through a lack of material resources or strain on family dynamics. Our results indicated that middle-income rather than lowest-income families experienced the most economic disruption, likely reflecting the sector-specific nature of business closures in the pandemic. Families who were financially vulnerable pre-pandemic were less likely to have had suitable resources for homeschooling. Both falls in income and strain in family relationships, such as arguing more with their parents, were associated with poorer scores on a measure of the child's emotional well-being. The emergency income support payment introduced at the start of the pandemic appeared to have a protective effect on the association between family income loss and child well-being, which has wider implications for policy on child poverty.

4.
Longit Life Course Stud ; 14(1): 48-72, 2022 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-36722293

RESUMEN

This article draws on the Growing Up in Ireland study and the Scottish sample of the Millennium Cohort Study to explore the factors influencing inequalities in children's cognitive skills on entry to primary education. It adopts a multidimensional comparative approach, which directly compares the effects of parental education and household income on several cognitive outcomes (vocabulary, language, reading and numbers) among five-year-old children and examines the extent to which inequalities in these outcomes are mediated by the home learning environment (HLE) and early childhood education (ECE). Home learning environment plays a stronger role in explaining actual vocabulary differences in Ireland while it plays a stronger role in school readiness (teacher-assessed skills) in Scotland. In both countries, use of centre-based care at 9 months and 3 years was markedly higher among the top income quintile. Centre-based care is found to play a mediating role in school readiness in Scotland. Nonetheless, the findings point to important direct effects of family background even when HLE and childcare are taken into account. The analyses point to differences in the trajectory of early skill development in the two countries, with the impact of early skill development being more marked in Scotland than Ireland. Comparative analyses of this kind thus yield important insights for policy development by highlighting potential domains (such as childcare) or timing (preschool or within-school) for intervention.


Asunto(s)
Cuidado del Niño , Instituciones Académicas , Preescolar , Niño , Humanos , Estudios de Cohortes , Irlanda , Escocia
5.
6.
Oncogene ; 37(34): 4735-4749, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29765153

RESUMEN

The pro-oncogenic kinase PKCε is overexpressed in human prostate cancer and cooperates with loss of the tumor suppressor Pten for the development of prostatic adenocarcinoma. However, the effectors driving PKCε-mediated phenotypes remain poorly defined. Here, using cellular and mouse models, we showed that PKCε overexpression acts synergistically with Pten loss to promote NF-κB activation and induce cyclooxygenase-2 (COX-2) expression, phenotypic traits which are also observed in human prostate tumors. Targeted disruption of PKCε from prostate cancer cells impaired COX-2 induction and PGE2 production. Notably, COX-2 inhibitors selectively killed prostate epithelial cells overexpressing PKCε, and this ability was greatly enhanced by Pten loss. Long-term COX-2 inhibition markedly reduced adenocarcinoma formation, as well as angiogenesis in a mouse model of prostate-specific PKCε expression and Pten loss. Overall, our results provide strong evidence for the involvement of the canonical NF-κB pathway and its target gene COX2 as PKCε effectors, and highlight the potential of PKCε as a useful biomarker for the use of COX inhibition for chemopreventive and/or chemotherapeutic purposes in prostate cancer.


Asunto(s)
Carcinogénesis/genética , Ciclooxigenasa 2/genética , Neoplasias de la Próstata/genética , Proteína Quinasa C-epsilon/genética , Adenocarcinoma/genética , Adenocarcinoma/patología , Animales , Línea Celular Tumoral , Humanos , Masculino , Ratones , FN-kappa B/genética , Próstata/patología , Neoplasias de la Próstata/patología , Transducción de Señal/genética
7.
Oncoimmunology ; 3: e29287, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25114833

RESUMEN

Three studies addressed the role of cyclooxygenase-2 (COX-2) in mammary tumorigenesis using epithelial and macrophage COX-2 knockout mice. Deletion of COX-2 in either cell restored, at least partially, tumor immunosurveillance either by changing macrophage function to offset pro-tumor effects, or by attracting more cytotoxic T lymphocytes and natural killer cells to the tumor. These studies suggest benefits from targeted COX-2 selective inhibition in combination with immunotherapies.

8.
Mol Pharmacol ; 86(1): 1-11, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24723492

RESUMEN

It is now well accepted that protease activated receptor (PAR) 1 and PAR4 have differential roles in platelet activation. PAR4, a low-affinity thrombin receptor in human platelets, participates in sustained platelet activation in a P2Y12-dependent manner; however, the mechanisms are not defined. Our previous studies demonstrated that thrombin induces the association of PAR4 with P2Y12, together with arrestin recruitment to the complex. Here we show that PAR4 and P2Y12 directly interact to coregulate Akt signaling after PAR4 activation. We observed direct and specific interaction of P2Y12 with PAR4 but not PAR1 by bioluminescent resonance energy transfer when the receptors were coexpressed in human embryonic kidney 293T cells. PAR4-P2Y12 dimerization was promoted by PAR4-AP and inhibited by P2Y12 antagonist. By using sequence comparison of the transmembrane domains of PAR1 and PAR4, we designed a mutant form of PAR4, "PAR4SFT," by replacing LGL194-196 at the base of transmembrane domain 4 with the corresponding aligned PAR1 residues SFT 220-222. PAR4SFT supported only 8.74% of PAR4-P2Y12 interaction, abolishing P2Y12-dependent arrestin recruitment to PAR4 and Akt activation. Nonetheless, PAR4SFT still supported homodimerization with PAR4. PAR4SFT failed to induce a calcium flux when expressed independently; however, coexpression of increasing concentrations of PAR4SFT, together with PAR4 potentiated PAR4-mediated calcium flux, suggested that PAR4 act as homodimers to signal to Gq-coupled calcium responses. In conclusion, PAR4 LGL (194-196) governs agonist-dependent association of PAR4 with P2Y12 and contributes to Gq-coupled calcium responses. PAR4-P2Y12 association supports arrestin-mediated sustained signaling to Akt. Hence, PAR4-P2Y12 dimerization is likely to be important for the PAR4-P2Y12 dependent stabilization of platelet thrombi.


Asunto(s)
Arrestinas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Purinérgicos P2Y12/metabolismo , Receptores de Trombina/metabolismo , Calcio/metabolismo , Células Cultivadas , Células HEK293 , Humanos , Proteínas de la Membrana/metabolismo , Activación Plaquetaria/fisiología , Estructura Terciaria de Proteína , Transducción de Señal/fisiología , Trombina/metabolismo
9.
Carcinogenesis ; 35(8): 1788-97, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24590894

RESUMEN

Cyclooxygenase-2 (COX-2) expression is associated with poor prognosis across a range of human cancers, including breast cancer. The contribution of tumor cell-derived COX-2 to tumorigenesis has been examined in numerous studies; however, the role of stromal-derived COX-2 is ill-defined. Here, we examined how COX-2 in myeloid cells, an immune cell subset that includes macrophages, influences mammary tumor progression. In mice engineered to selectively lack myeloid cell COX-2 [myeloid-COX-2 knockout (KO) mice], spontaneous neu oncogene-induced tumor onset was delayed, tumor burden reduced, and tumor growth slowed compared with wild-type (WT). Similarly, growth of neu-transformed mammary tumor cells as orthotopic tumors in immune competent syngeneic myeloid-COX-2 KO host mice was reduced compared with WT. By flow cytometric analysis, orthotopic myeloid-COX-2 KO tumors had lower tumor-associated macrophage (TAM) infiltration consistent with impaired colony stimulating factor-1-dependent chemotaxis by COX-2 deficient macrophages in vitro. Further, in both spontaneous and orthotopic tumors, COX-2-deficient TAM displayed lower immunosuppressive M2 markers and this was coincident with less suppression of CD8(+) cytotoxic T lymphocytes (CTLs) in myeloid-COX-2 KO tumors. These studies suggest that reduced tumor growth in myeloid-COX-2 KO mice resulted from disruption of M2-like TAM function, thereby enhancing T-cell survival and immune surveillance. Antibody-mediated depletion of CD8(+), but not CD4(+) cells, restored tumor growth in myeloid-COX-2 KO to WT levels, indicating that CD8(+) CTLs are dominant antitumor effectors in myeloid-COX-2 KO mice. Our studies suggest that inhibition of myeloid cell COX-2 can potentiate CTL-mediated tumor cytotoxicity and may provide a novel therapeutic approach in breast cancer therapy.


Asunto(s)
Ciclooxigenasa 2/fisiología , Macrófagos/inmunología , Neoplasias Mamarias Animales/inmunología , Neoplasias Mamarias Animales/prevención & control , Células Mieloides/inmunología , Linfocitos T Citotóxicos/inmunología , Microambiente Tumoral/inmunología , Animales , Western Blotting , Transformación Celular Neoplásica/inmunología , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Femenino , Citometría de Flujo , Humanos , Técnicas para Inmunoenzimas , Integrasas/metabolismo , Activación de Linfocitos , Macrófagos/metabolismo , Macrófagos/patología , Neoplasias Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Células Mieloides/metabolismo , Células Mieloides/patología , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor de Factor Estimulante de Colonias de Macrófagos/genética , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Linfocitos T Citotóxicos/metabolismo , Linfocitos T Citotóxicos/patología , Células Tumorales Cultivadas
10.
Breast Cancer Res ; 15(5): R75, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24004819

RESUMEN

INTRODUCTION: Systemic inhibition of the inflammatory enzyme cyclooxygenase (COX) 2 decreases the risk of breast cancer and its recurrence. However, the biology of COX-2 in the multicellular tumor microenvironment is poorly defined. METHODS: Mammary tumor onset and multiplicity were examined in ErbB2 transgenic mice that were deficient in mammary epithelial cell COX-2 (COX-2(MEC)KO) compared to wild type (WT) mice. Tumors were analyzed, by real time PCR, immune-staining and flow cytometry, for proliferation, apoptosis, angiogenesis and immune microenvironment. Lentiviral shRNA delivery was used to knock down (KD) COX-2 in ErbB2-transformed mouse breast cancer cells (COX-2KD), and growth as orthotopic tumors was examined in syngenic recipient mice, with or without depletion of CD8+ immune cells. RESULTS: Mammary tumor onset was delayed, and multiplicity halved, in COX-2(MEC)KO mice compared to WT. COX-2(MEC)KO tumors showed decreased expression of Ki67, a proliferation marker, as well as reduced VEGFA, its receptor VEGFR2, endothelial NOS and the vascular endothelial marker CD31, indicating reduced tumor vascularization. COX-2(MEC)KO tumors contained more CD4+ T helper (Th) cells and CD8+ cytotoxic immune cells (CTL) consistent with increased immune surveillance. The ratio of Th markers Tbet (Th1) to GATA3 (Th2) was higher, and levels of Retnla, a M2 macrophage marker, lower, in COX-2(MEC)KO tumor infiltrating leukocytes compared to WT, suggesting a prevalence of pro-immune Th1 over immune suppressive Th2 lymphocytes, and reduced macrophage polarization to the immune suppressive M2 phenotype. Enhanced immune surveillance in COX-2(MEC)KO tumors was coincident with increased intratumoral CXCL9, a T cell chemoattractant, and decreased expression of T lymphocyte co-inhibitory receptors CTLA4 and PD-1, as well as PD-L1, the ligand for PD-1. PD-L1 was also decreased in IFNγ-treated COX-2KD mouse mammary cancer cells in vitro and, compared to control cells, growth of COX-2KD cells as orthotopic tumors in immune competent mice was markedly suppressed. However, robust growth of COX-2KD tumor cells was evident when recipients were depleted of CD8+ cells. CONCLUSIONS: The data strongly support that, in addition to its angiogenic function, tumor cell COX-2 suppresses intratumoral cytotoxic CD8+ immune cell function, possibly through upregulation of immune checkpoints, thereby contributing to tumor immune escape. COX-2 inhibition may be clinically useful to augment breast cancer immunotherapy.


Asunto(s)
Neoplasias de la Mama/inmunología , Neoplasias de la Mama/metabolismo , Carcinoma/inmunología , Carcinoma/metabolismo , Ciclooxigenasa 2/metabolismo , Vigilancia Inmunológica , Animales , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/inmunología , Ciclooxigenasa 2/genética , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Técnicas de Silenciamiento del Gen , Activación de Linfocitos/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Neoplasias Mamarias Experimentales , Ratones , Ratones Noqueados , Neovascularización Patológica/genética , Neovascularización Patológica/inmunología , Fenotipo , Escape del Tumor/inmunología
11.
J Lipid Res ; 54(6): 1678-1690, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23493750

RESUMEN

Thromboxane A2 (TXA2) contributes to cardiovascular disease (CVD) by activating platelets and vascular constriction and proliferation. Despite their preclinical efficacy, pharmacological antagonists of the TXA2 receptor (TP), a G protein-coupled receptor, have not been clinically successful, raising interest in novel approaches to modifying TP function. We determined that disruption of a GxxxGxxxL helical interaction motif in the human TP's (α isoform) fifth transmembrane (TM) domain suppressed TP agonist-induced Gq signaling and TPα homodimerization, but not its cell surface expression, ligand affinity, or Gq association. Heterodimerization of TPα with the functionally opposing prostacyclin receptor (IP) shifts TPα to signal via the IP-Gs cascade contributing to prostacyclin's restraint of TXA2 function. Interestingly, disruption of the TPα-TM5 GxxxGxxxL motif did not modify either IP-TPα heterodimerization or its Gs-cAMP signaling. Our study indicates that distinct regions of the TPα receptor direct its homo- and heterodimerization and that homodimerization is necessary for normal TPα-Gq activation. Targeting the TPα-TM5 GxxxGxxxL domain may allow development of biased TPα homodimer antagonists that avoid suppression of IP-TPα heterodimer function. Such novel therapeutics may prove superior in CVD compared with nonselective suppression of all TP functions with TXA2 biosynthesis inhibitors or TP antagonists.


Asunto(s)
Multimerización de Proteína/fisiología , Receptores de Prostaglandina/metabolismo , Receptores de Tromboxanos/metabolismo , Sistemas de Mensajero Secundario/fisiología , Secuencias de Aminoácidos , AMP Cíclico/genética , AMP Cíclico/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Células HEK293 , Humanos , Estructura Terciaria de Proteína , Receptores de Epoprostenol , Receptores de Prostaglandina/genética , Receptores de Tromboxanos/genética
12.
Am J Physiol Heart Circ Physiol ; 304(3): H382-92, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23203967

RESUMEN

Arachidonic acid (AA) metabolites mediate endothelium-dependent relaxation in many vascular beds. Previously, we identified the major AA 12/15-lipoxygenase (12/15-LO) metabolite of mouse arteries as 12-hydroxyeicosatetraenoic acid (12-HETE). The goal was to determine the stereospecific configuration of mouse vascular 12-HETE and characterize the role of 12-HETE stereoisomers in the regulation of vascular tone. Using normal, reverse phase, and chiral HPLC, the stereospecific configuration was identified as 12(S)-HETE. 12(S)-HETE relaxed U46619-, carbocyclic thromboxane A(2)-, PGF(2α)-, and 8-iso PGF(2α)-preconstricted mesenteric arteries, but not phenylephrine-preconstricted arteries. 12(R)-HETE was more potent than 12(S)-HETE in relaxing U46619-preconstricted mouse arteries (maximum relaxations = 91.4 ± 2.7% and 71.8 ± 5.9%, respectively). Neither 12-HETE isomer caused constriction. Pretreatment with 12(S)- or 12(R)-HETE (1 µM) inhibited constrictions to U46619 but not phenylephrine. To investigate the role of thromboxane A(2) (TP) receptors in 12-HETE vascular actions, [(3)H]SQ29548 radioligand binding studies were performed in mouse platelets. U46619, 12(R)-HETE, and 12(S)-HETE displaced [(3)H]SQ29548 binding with IC(50)s of 0.07, 0.32, and 1.73 µM, respectively. Both 12(S)- and 12(R)-HETE inhibited intracellular calcium increases induced by U46619 (10 nM) in HEK293 cells overexpressing TP(α) receptor (65.5% and 45.1%, respectively) and coexpressing prostacyclin (IP) and TP(α) receptors (58.0% and 27.1%, respectively). The LO inhibitor NDGA (10 µM) reduced AA relaxations in arteries preconstricted with U46619 but not phenylephrine. These results indicate that exogenous and endogenous 12(S)-HETE relax mouse mesenteric arteries that are preconstricted with thromboxane agonists. These 12(S)-HETE relaxations are mediated by TP receptor competitive inhibition and inhibition of TP agonist-induced increases in intracellular calcium.


Asunto(s)
Ácido 12-Hidroxi-5,8,10,14-Eicosatetraenoico/farmacología , Endotelio Vascular/efectos de los fármacos , Arterias Mesentéricas/efectos de los fármacos , Receptores de Tromboxanos/antagonistas & inhibidores , Vasodilatadores/farmacología , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacología , Animales , Ácido Araquidónico/metabolismo , Plaquetas/efectos de los fármacos , Western Blotting , Compuestos Bicíclicos Heterocíclicos con Puentes , Señalización del Calcio/efectos de los fármacos , Cromatografía Líquida de Alta Presión , Ácidos Grasos Insaturados , Células HEK293 , Humanos , Hidrazinas/farmacología , Contracción Isométrica/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores Acoplados a Proteínas G/efectos de los fármacos , Receptores de Leucotrieno B4/efectos de los fármacos , Receptores de Tromboxano A2 y Prostaglandina H2/efectos de los fármacos , Vasoconstrictores/farmacología
13.
Arterioscler Thromb Vasc Biol ; 33(1): 60-6, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23162015

RESUMEN

OBJECTIVE: Prostacyclin and thromboxane mediate opposing cardiovascular actions through receptors termed IP and TP, respectively. When dimerized with IP, the TP shifts to IP-like function. IP localizes to cholesterol-enriched membrane rafts, but TP and IPTP heterodimer localization is not defined. We examined these receptors' membrane localization and the role of rafts in receptor function. METHODS AND RESULTS: Microdomain distribution of IP, TP, and IPTP heterodimers was examined in COS-7 cells by measuring energy transfer from renilla luciferase-fused receptors to fluorescently labeled rafts. IP raft association was confirmed. TP was raft excluded, but redistributed to rafts upon dimerization with IP. Signaling of the IP and IPTP heterodimer, but not TP alone, was suppressed after raft disruption by cholesterol depletion. Cholesterol enrichment also selectively suppressed IP and IPTP function. Native IP and IPTP signaling in smooth muscle cells and macrophages were similarly sensitive to cholesterol manipulation, whereas macrophages from hypercholesterolemic mice displayed suppressed IP and IPTP function. CONCLUSIONS: IP and TP function within distinct microdomains. Raft incorporation of TP in the IPTP heterodimer likely facilitates its signaling shift. We speculate that changes in IP and IPTP signaling after perturbation of membrane cholesterol may contribute to cardiovascular disease associated with hypercholesterolemia.


Asunto(s)
Microdominios de Membrana/metabolismo , Receptores de Prostaglandina/metabolismo , Receptores de Tromboxano A2 y Prostaglandina H2/metabolismo , Animales , Células COS , Chlorocebus aethiops , Colesterol/deficiencia , AMP Cíclico/metabolismo , Dimerización , Modelos Animales de Enfermedad , Femenino , Transferencia Resonante de Energía de Fluorescencia , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Humanos , Hipercolesterolemia/metabolismo , Luciferasas de Renilla/genética , Luciferasas de Renilla/metabolismo , Macrófagos Peritoneales/metabolismo , Ratones , Miocitos del Músculo Liso/metabolismo , Transporte de Proteínas , Receptores de Epoprostenol , Receptores de Prostaglandina/genética , Receptores de Tromboxano A2 y Prostaglandina H2/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Factores de Tiempo , Transfección
14.
Prostaglandins Other Lipid Mediat ; 96(1-4): 14-20, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21907301

RESUMEN

COX-derived prostanoids play multiple roles in inflammation and cancer. This review highlights research examining COX-2 and PGE(2)-dependent regulation of immune cell polarization and function within the tumor microenvironment, particularly as it pertains to breast cancer. Appreciating PGE(2)-mediated immunomodulation is important in understanding how tumors evade immune surveillance by re-educating infiltrating inflammatory and immune cells to support tumorigenesis. Elucidation of the multiple and complex influences exerted by tumor stromal components may lead to targeted therapies in breast and other cancers that restrain microenvironmental permissiveness and maintain natural defenses against malignancies.


Asunto(s)
Neoplasias de la Mama/inmunología , Carcinoma/inmunología , Transformación Celular Neoplásica/inmunología , Ciclooxigenasa 2/inmunología , Dinoprostona/inmunología , Inmunomodulación , Inmunoterapia/métodos , Microambiente Tumoral/inmunología , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Carcinoma/tratamiento farmacológico , Carcinoma/genética , Carcinoma/metabolismo , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Citocinas/genética , Citocinas/inmunología , Citocinas/metabolismo , Dinoprostona/genética , Dinoprostona/metabolismo , Dinoprostona/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Transducción de Señal/genética , Transducción de Señal/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo
15.
Carcinogenesis ; 32(10): 1441-9, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21771729

RESUMEN

Inhibition of cyclooxygenase (COX) 2, which is associated with >40% of breast cancers, decreases the risk of tumorigenesis and breast cancer recurrence. To study the role of COX-2 in breast cancer, we engineered mice that lack selectively mammary epithelial cell (MEC) COX-2 (COX-2 KO(MEC)). Compared with wild type (WT), MEC from COX-2 KO(MEC) mice expressed >90% less COX-2 messenger RNA (mRNA) and protein and produced 90% less of the dominant pro-oncogenic COX-2 product, prostaglandin (PG) E(2). We confirmed COX-2 as the principle source of PGE(2) in MEC treated with selective COX-2 and COX-1 inhibitors. Tumors were induced in mice using medroxyprogesterone acetate and 7,12-dimethylbenz[a]anthracene. Breast cancer onset was significantly delayed in COX-2 KO(MEC) compared with WT (P = 0.03), equivalent to the delay following systemic COX-2 inhibition with rofecoxib. Compared with WT, COX-2 KO(MEC) tumors showed increased mRNA for Caspase-3, Ki-67 and common markers for leukocytes (CD45) and macrophages (F4/80). Analysis of multiple markers/cytokines, namely CD86, inducible nitric oxide synthase (iNOS), interleukin-6, tumor necrosis factor α (TNFα) and Tim-3 indicated a shift toward antitumorigenic type 1 immune responses in COX-2 KO(MEC) tumors. Immunohistochemical analysis confirmed elevated expression of CD45, F4/80 and CD86 in COX-2 KO(MEC) tumors. Concordant with a role for COX-2 in restraining M1 macrophage polarization, CD86 and TNFα expression were offset by exogenous PGE(2) in bone marrow-derived macrophages polarized in vitro to the M1 phenotype. Our data reveal the importance of epithelial COX-2 in tumor promotion and indicate that deletion of epithelial COX-2 may skew tumor immunity toward type 1 responses, coincident with delayed tumor development.


Asunto(s)
Ciclooxigenasa 2/fisiología , Células Epiteliales/inmunología , Glándulas Mamarias Animales/inmunología , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/prevención & control , Células TH1/inmunología , 9,10-Dimetil-1,2-benzantraceno/toxicidad , Animales , Western Blotting , Carcinógenos/toxicidad , Células Cultivadas , Anticonceptivos Sintéticos Orales/toxicidad , Citocinas/metabolismo , Eicosanoides/metabolismo , Células Epiteliales/metabolismo , Femenino , Técnicas para Inmunoenzimas , Mediadores de Inflamación/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Macrófagos Peritoneales/citología , Macrófagos Peritoneales/metabolismo , Masculino , Glándulas Mamarias Animales/metabolismo , Neoplasias Mamarias Experimentales/inducido químicamente , Medroxiprogesterona/toxicidad , Ratones , Ratones Noqueados , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células TH1/metabolismo
16.
Arterioscler Thromb Vasc Biol ; 30(9): 1802-9, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20522800

RESUMEN

OBJECTIVE: Prostacyclin and thromboxane mediate opposing cardiovascular effects through their receptors, the prostacyclin receptor (IP) and thromboxane receptor (TP). Individuals heterozygous for an IP variant, IP(R212C), displayed exaggerated loss of platelet IP responsiveness and accelerated cardiovascular disease. We examined association of IP(R212C) into homo- and heterodimeric receptor complexes and the impact on prostacyclin and thromboxane biology. METHODS AND RESULTS: Dimerization of the IP, IP(R212C), and TPalpha was examined by bioluminesence resonance energy transfer in transfected HEK293 cells. We observed an equal propensity for formation of IPIP homodimers and IPTPalpha heterodimers. Compared with the IP alone, IP(R212C) displayed reduced cAMP generation and increased endoplasmic reticulum localization but underwent normal homo- and heterodimerization. When the IP(R212C) and IP were coexpressed, a dominant negative action of the variant was evident with enhanced wild-type IP localization to the endoplasmic reticulum and reduced agonist-dependent signaling. Further, the TPalpha activation response, which was shifted from inositol phosphate to cAMP generation following IPTPalpha heterodimerization, was normalized when the TPalpha instead dimerized with IP(R212C). CONCLUSIONS: IP(R212C) exerts a dominant action on the wild-type IP and TPalpha through dimerization. This likely contributes to accelerated cardiovascular disease in individuals carrying 1 copy of the variant allele.


Asunto(s)
Enfermedades Cardiovasculares/metabolismo , Mutación , Receptores de Prostaglandina/metabolismo , Receptores de Tromboxano A2 y Prostaglandina H2/metabolismo , Enfermedades Cardiovasculares/genética , Línea Celular , AMP Cíclico/metabolismo , Dimerización , Relación Dosis-Respuesta a Droga , Retículo Endoplásmico/metabolismo , Epoprostenol/análogos & derivados , Epoprostenol/farmacología , Transferencia Resonante de Energía de Fluorescencia , Genotipo , Heterocigoto , Homocigoto , Humanos , Fosfatos de Inositol/metabolismo , Fenotipo , Receptores de Epoprostenol , Receptores de Prostaglandina/agonistas , Receptores de Prostaglandina/genética , Proteínas Recombinantes de Fusión/metabolismo , Sistemas de Mensajero Secundario , Transfección
17.
Clin Lipidol ; 5(2): 209-219, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-20543887

RESUMEN

Thromboxane A(2) (TXA(2)), the primary product of COX-1-dependent metabolism of arachidonic acid, mediates its biological actions through the TXA(2) receptor, termed the TP. Irreversible inhibition of platelet COX-1-derived TXA(2) with low-dose aspirin affords protection against primary and secondary vascular thrombotic events, underscoring the central role of TXA(2) as a platelet agonist in cardiovascular disease. The limitations associated with aspirin use include significant gastrointestinal toxicity, bleeding complications, potential interindividual response variability and poor efficacy in some disease states. This, together with the broad role of TXA(2) in cardiovascular disease beyond the platelet, has refocused interest towards additional TXA(2)-associated drug targets, in particular TXA(2) synthase and the TP. The superiority of these agents over low-dose aspirin, in terms of clinical efficacy, tolerability and commercial viability, remain open questions that are the focus of ongoing research.

18.
Circ Res ; 106(2): 337-45, 2010 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-19940265

RESUMEN

RATIONALE: Cyclooxygenase (COX)-derived prostanoids (PGs) are involved in blood pressure homeostasis. Both traditional nonsteroidal antiinflammatory drugs (NSAIDs) that inhibit COX-1 and COX-2 and NSAIDs designed to be selective for inhibition of COX-2 cause sodium retention and elevate blood pressure. OBJECTIVE: To elucidate the role of COX-2 in blood pressure homeostasis using COX-1>COX-2 mice, in which the COX-1 expression is controlled by COX-2 regulatory elements. METHODS AND RESULTS: COX-1>COX-2 mice developed systolic hypertension relative to wild types (WTs) on a high-salt diet (HSD); this was attenuated by a PGI(2) receptor agonist. HSD increased expression of COX-2 in WT mice and of COX-1 in COX-1>COX-2 mice in the inner renal medulla. The HSD augmented in all strains urinary prostanoid metabolite excretion, with the exception of the major PGI(2) metabolite that was suppressed on regular chow and unaltered by the HSD in both mutants. Furthermore, inner renal medullary expression of the receptor for PGI(2), but not for other prostanoids, was depressed by HSD in WT and even more so in both mutant strains. Increasing osmolarity augmented expression of COX-2 in WT renal medullary interstitial cells and again the increase in formation of PGI(2) observed in WTs was suppressed in cells derived from both mutants. Intramedullary infusion of the PGI(2) receptor agonist increased urine volume and sodium excretion in mice. CONCLUSIONS: These studies suggest that dysregulated expression of the COX-2 dependent, PGI(2) biosynthesis/response pathway in the renal inner renal medulla undermines the homeostatic response to a HSD. Inhibition of this pathway may contribute directly to the hypertensive response to NSAIDs.


Asunto(s)
Presión Sanguínea/fisiología , Ciclooxigenasa 2/metabolismo , Epoprostenol/biosíntesis , Homeostasis/fisiología , Animales , Western Blotting , Células Cultivadas , Ciclooxigenasa 2/genética , Epoprostenol/análogos & derivados , Epoprostenol/farmacología , Femenino , Hipertensión/etiología , Hipertensión/metabolismo , Médula Renal/citología , Médula Renal/efectos de los fármacos , Médula Renal/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados , Natriuresis/efectos de los fármacos , Receptores de Prostaglandina/metabolismo , Renina/genética , Renina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Cloruro de Sodio Dietético/administración & dosificación
19.
J Biol Chem ; 284(46): 31936-44, 2009 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-19776010

RESUMEN

Recent reports demonstrate T-cell infiltration of adipose tissue in early obesity. We hypothesized that interferon (IFN) gamma, a major T-cell inflammatory cytokine, would attenuate human adipocyte functions and sought to establish signaling mechanisms. Differentiated human adipocytes were treated with IFNgamma +/- pharmacological inhibitors prior to insulin stimulation. [(3)H]Glucose uptake and AKT phosphorylation were assessed as markers of insulin sensitivity. IFNgamma induced sustained loss of insulin-stimulated glucose uptake in human adipocytes, coincident with reduced Akt phosphorylation and down-regulation of the insulin receptor, insulin receptor substrate-1, and GLUT4. Loss of adipocyte triglyceride storage was observed with IFNgamma co-incident with reduced expression of peroxisome proliferator-activated receptor gamma, adiponectin, perilipin, fatty acid synthase, and lipoprotein lipase. Treatment with IFNgamma also blocked differentiation of pre-adipocytes to the mature phenotype. IFNgamma-induced robust STAT1 phosphorylation and SOCS1 mRNA expression, with modest, transient STAT3 phosphorylation and SOCS3 induction. Preincubation with a non-selective JAK inhibitor restored glucose uptake and Akt phosphorylation while completely reversing IFNgamma suppression of adipogenic mRNAs and adipocyte differentiation. Specific inhibition of JAK2 or JAK3 failed to block IFNgamma effects suggesting a predominant role for JAK1-STAT1. We demonstrate that IFNgamma attenuates insulin sensitivity and suppresses differentiation in human adipocytes, an effect most likely mediated via sustained JAK-STAT1 pathway activation.


Asunto(s)
Adipocitos/metabolismo , Diferenciación Celular , Insulina/metabolismo , Interferón gamma/farmacología , Janus Quinasa 1/metabolismo , Factor de Transcripción STAT1/metabolismo , Triglicéridos/metabolismo , Western Blotting , Glucosa/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Humanos , Hipoglucemiantes/metabolismo , Proteínas Sustrato del Receptor de Insulina/metabolismo , Resistencia a la Insulina , Metabolismo de los Lípidos , PPAR gamma/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
20.
Proc Natl Acad Sci U S A ; 106(19): 7985-90, 2009 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-19416858

RESUMEN

Little is known about prostaglandin F(2alpha) in cardiovascular homeostasis. Prostaglandin F(2alpha) dose-dependently elevates blood pressure in WT mice via activation of the F prostanoid (FP) receptor. The FP is expressed in preglomerular arterioles, renal collecting ducts, and the hypothalamus. Deletion of the FP reduces blood pressure, coincident with a reduction in plasma renin concentration, angiotensin, and aldosterone, despite a compensatory up-regulation of AT1 receptors and an augmented hypertensive response to infused angiotensin II. Plasma and urinary osmolality are decreased in FP KOs that exhibit mild polyuria and polydipsia. Atherogenesis is retarded by deletion of the FP, despite the absence of detectable receptor expression in aorta or in atherosclerotic lesions in Ldlr KOs. Although vascular TNF(alpha), inducible nitric oxide enzyme and TGF(beta) are reduced and lesional macrophages are depleted in the FP/Ldlr double KOs, this result reflects the reduction in lesion burden, as the FP is not expressed on macrophages and its deletion does not alter macrophage cytokine generation. Blockade of the FP offers an approach to the treatment of hypertension and its attendant systemic vascular disease.


Asunto(s)
Aterosclerosis/etiología , Presión Sanguínea , Dinoprost/fisiología , Hipertensión/etiología , Animales , Femenino , Macrófagos/metabolismo , Masculino , Ratones , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo II/metabolismo , Receptores de Prostaglandina/metabolismo , Renina/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA