Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
1.
Biochem Pharmacol ; 188: 114541, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33812857

RESUMEN

The acute phase response, as a component of the innate immune system, is part of the first line of defense against invading pathogens. The Stimulator of Interferon Genes (STING) pathway initiates innate immune responses upon recognition of exogenous bacterial and viral DNA. However, whether STING signaling pathway plays any roles in regulating acute phase response during bacterial infection remains unknown. In this study, we used STING-deficient (Tmem173gt) and wildtype mice to investigate acute phase responses to bacterial infection (Escherichia coli, E. coli) and test the effect of exogenous cyclic GMP-AMP (cGAMP, a STING agonist) treatment. Bacterial infection of STING-deficient mice resulted in an increase in mortality and bacterial dissemination. Also, inflammation-induced acute phase response was drastically reduced in STING-deficient mice, showing significant reduction in expression of cytokine TNF-α and acute phase proteins. In contrast, exogenous cGAMP treatment enhanced inflammation-induced acute phase response by increasing the expression of TNF-α and acute phase proteins. Also, cGAMP accelerated bacterial clearance and improved survival rate of wildtype mice, but not STING-deficient mice. Interestingly, cGAMP treatment mitigated bacterial infection induced liver injury in both wildtype and STING-deficient mice. Further in vitro evidence showed that cGAMP treatment retarded TNF-α-mediated hepatocyte apoptosis, potentially accelerating autophagy. Taken together, our results indicated that cGAMP/STING signaling pathway is critical for organism to initiate blood-borne innate immune-responses to defend bacterial infection, and cGAMP is envisaged as a drug candidate for further clinical trial.


Asunto(s)
Reacción de Fase Aguda/metabolismo , Reacción de Fase Aguda/prevención & control , AMP Cíclico/administración & dosificación , GMP Cíclico/administración & dosificación , Infecciones por Escherichia coli/metabolismo , Infecciones por Escherichia coli/prevención & control , Proteínas de la Membrana/deficiencia , Reacción de Fase Aguda/genética , Animales , Escherichia coli , Infecciones por Escherichia coli/genética , Hepatocitos/metabolismo , Hepatocitos/microbiología , Masculino , Proteínas de la Membrana/agonistas , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos
2.
Biomed Res Int ; 2021: 8852233, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33575350

RESUMEN

BACKGROUND/AIM: Irreversible electroporation (IRE) showed promising results for small-size tumors and very early cancers. However, further development is needed to evolve this procedure into a more efficient ablation technique for long-term control of tumor growth. In this work, we show that it is possible to increase the antitumor efficiency of IRE by simmultaneously injecting c-di-GMP, a STING agonist, intratumorally. MATERIALS AND METHODS: Intratumoral administration of c-di-GMP simultaneously to IRE was evaluated in murine models of melanona (B16.OVA) and hepatocellular carcinoma (PM299L). RESULTS: The combined therapy increased the number of tumor-infiltrating IFN-γ/TNF-α-producing CD4 and CD8 T cells and delayed tumor growth, as compared to the effect observed in groups treated with c-di-GMP or IRE alone. CONCLUSION: These results can lead to the development of a new therapeutic strategy for the treatment of cancer patients refractory to other therapies.


Asunto(s)
Técnicas de Ablación/métodos , Carcinoma Hepatocelular/terapia , GMP Cíclico/análogos & derivados , Electroporación/métodos , Neoplasias Hepáticas/terapia , Proteínas de la Membrana/agonistas , Animales , Línea Celular , Terapia Combinada/métodos , GMP Cíclico/administración & dosificación , Femenino , Neoplasias Hepáticas Experimentales/terapia , Ratones Endogámicos C57BL
3.
Front Immunol ; 11: 1674, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32849581

RESUMEN

Cyclic dinucleotides (CDNs) are promising vaccine adjuvants inducing balanced, potent humoral, and cellular immune responses. How aging influences CDN efficacy is unclear. We examined the vaccine efficacy of 3',5'-cyclic diguanylic acid (cyclic di-GMP, CDG), the founding member of CDNs, in 1-year-old (middle-aged) and 2-year-old (aged) C57BL/6J mice. We found that 1- and 2-year-old C57BL/6J mice are defective in CDG-induced memory T helper (Th)1 and Th17 responses and high-affinity serum immunoglobulin (Ig)G, mucosal IgA production. Next, we generated two novel tumor necrosis factor (TNF) fusion proteins that target soluble TNF (solTNF) and transmembrane TNF (tmTNF) to monocyte-derived dendritic cells (moDCs) to enhance CDG vaccine efficacy in 1- and 2-year-old mice. The moDC-targeting TNF fusion proteins restored CDG-induced memory Th1, Th17, and high-affinity IgG, IgA responses in the 1- and 2-year-old mice. Together, the data suggested that aging negatively impacts CDG vaccine adjuvanticity. MoDC-targeting TNF fusion proteins enhanced CDG adjuvanticity in the aging mice.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , GMP Cíclico/análogos & derivados , Células Dendríticas/efectos de los fármacos , Fragmentos Fc de Inmunoglobulinas/farmacología , Pulmón/efectos de los fármacos , Proteínas Recombinantes de Fusión/farmacología , Factor de Necrosis Tumoral alfa/farmacología , Adyuvantes Inmunológicos/administración & dosificación , Administración Intranasal , Factores de Edad , Envejecimiento/inmunología , Envejecimiento/metabolismo , Animales , Células Cultivadas , GMP Cíclico/administración & dosificación , GMP Cíclico/farmacología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Inmunidad Mucosa/efectos de los fármacos , Inmunización , Inmunogenicidad Vacunal , Inmunoglobulina A/inmunología , Inmunoglobulina A/metabolismo , Fragmentos Fc de Inmunoglobulinas/administración & dosificación , Pulmón/inmunología , Pulmón/metabolismo , Ratones Endogámicos C57BL , Proteínas Recombinantes de Fusión/administración & dosificación , Células TH1/efectos de los fármacos , Células TH1/inmunología , Células TH1/metabolismo , Células Th17/efectos de los fármacos , Células Th17/inmunología , Células Th17/metabolismo , Factor de Necrosis Tumoral alfa/administración & dosificación
4.
Cancer Res ; 79(20): 5394-5406, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31431457

RESUMEN

Effective cancer immunotherapy depends on the robust activation of tumor-specific antigen-presenting cells (APC). Immune agonists encapsulated within nanoparticles (NP) can be delivered to tumor sites to generate powerful antitumor immune responses with minimal off-target dissemination. Systemic delivery enables widespread access to the microvasculature and draining to the APC-rich perivasculature. We developed an immuno-nanoparticle (immuno-NP) coloaded with cyclic diguanylate monophosphate, an agonist of the stimulator of interferon genes pathway, and monophosphoryl lipid A, and a Toll-like receptor 4 agonist, which synergize to produce high levels of type I IFNß. Using a murine model of metastatic triple-negative breast cancer, systemic delivery of these immuno-NPs resulted in significant therapeutic outcomes due to extensive upregulation of APCs and natural killer cells in the blood and tumor compared with control treatments. These results indicate that NPs can facilitate systemic delivery of multiple immune-potentiating cargoes for effective APC-driven local and systemic antitumor immunity. SIGNIFICANCE: Systemic administration of an immuno-nanoparticle in a murine breast tumor model drives a robust tumor site-specific APC response by delivering two synergistic immune-potentiating molecules, highlighting the potential of nanoparticles for immunotherapy.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , GMP Cíclico/análogos & derivados , Sistemas de Liberación de Medicamentos/métodos , Interferón beta/fisiología , Lípido A/análogos & derivados , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Melanoma Experimental/tratamiento farmacológico , Nanocápsulas/administración & dosificación , Receptor Toll-Like 4/agonistas , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Animales , Células Presentadoras de Antígenos/efectos de los fármacos , GMP Cíclico/administración & dosificación , GMP Cíclico/uso terapéutico , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Femenino , Células Asesinas Naturales/inmunología , Lípido A/administración & dosificación , Lípido A/uso terapéutico , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/patología , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Microcirculación , Neoplasias de la Mama Triple Negativas/inmunología , Neoplasias de la Mama Triple Negativas/patología
5.
Mol Pharm ; 15(11): 5437-5443, 2018 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-30299105

RESUMEN

Microneedle-based skin allergen-specific immunotherapy (AIT) can benefit from adjuvants that can stimulate a stronger Th1 response against the allergen. We evaluated two stimulator of interferon genes (STING) agonists, namely, cyclic diguanylate monophosphate (c-di-GMP) and cyclic diadenylate monophosphate (c-di-AMP), as skin adjuvants using coated microneedles (MNs). For comparison, the approved subcutaneous (SC) hypodermic injection containing alum was used. Ovalbumin (Ova) was used as a model allergen. Ova-specific IgG2a antibody in serum, which is a surrogate marker for Th1 type immune response was significantly higher when STING agonists were used with coated MNs as compared to SC injection of Ova+alum in mice. In contrast, IgG1 antibody, a surrogate marker for Th2 type immune response, was at comparable levels in the MN and SC groups. Restimulation of splenocytes with Ova produced higher levels of Th1 cytokines (IFN-γ and IL-2) in the STING agonists MN groups as compared to the SC group. In conclusion, delivery of STING agonists into the skin using coated MNs activated the Th1 pathway better than SC- and MN-based delivery of alum. Thus, STING agonists could fulfill the role of adjuvants for skin AIT and even for infectious disease vaccines, where stimulation of the Th1 pathway is of interest.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Alérgenos/administración & dosificación , Desensibilización Inmunológica/métodos , Células TH1/inmunología , Células Th2/inmunología , Administración Cutánea , Compuestos de Alumbre/administración & dosificación , Animales , GMP Cíclico/administración & dosificación , GMP Cíclico/análogos & derivados , Fosfatos de Dinucleósidos/administración & dosificación , Femenino , Proteínas de la Membrana/agonistas , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos BALB C , Modelos Animales , Agujas , Ovalbúmina/administración & dosificación , Células TH1/efectos de los fármacos , Células Th2/efectos de los fármacos
6.
Nanoscale ; 10(19): 9311-9319, 2018 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-29737353

RESUMEN

Nanotechnology has demonstrated tremendous clinical utility, with potential applications in cancer immunotherapy. Although nanoparticles with intrinsic cytotoxicity are often considered unsuitable for clinical applications, such toxicity may be harnessed in the fight against cancer. Nanoparticle-associated toxicity can induce acute necrotic cell death, releasing tumor-associated antigens which may be captured by antigen-presenting cells to initiate or amplify tumor immunity. To test this hypothesis, cytotoxic cationic silica nanoparticles (CSiNPs) were directly administered into B16F10 melanoma implanted in C57BL/6 mice. CSiNPs caused plasma membrane rupture and oxidative stress of tumor cells, inducing local inflammation, tumor cell death and the release of tumor-associated antigens. The CSiNPs were further complexed with bis-(3'-5')-cyclic dimeric guanosine monophosphate (c-di-GMP), a molecular adjuvant which activates the stimulator of interferon genes (STING) in antigen-presenting cells. Compared with unformulated c-di-GMP, the delivery of c-di-GMP with CSiNPs markedly prolonged its local retention within the tumor microenvironment and activated tumor-infiltrating antigen-presenting cells. The combination of CSiNPs and a STING agonist showed dramatically increased expansion of antigen-specific CD8+ T cells, and potent tumor growth inhibition in murine melanoma. These results demonstrate that cationic nanoparticles can be used as an effective in situ vaccine platform which simultaneously causes tumor destruction and immune activation.


Asunto(s)
Muerte Celular , Melanoma Experimental/inmunología , Proteínas de la Membrana/metabolismo , Nanopartículas , Dióxido de Silicio , Microambiente Tumoral , Animales , Linfocitos T CD8-positivos , GMP Cíclico/administración & dosificación , GMP Cíclico/análogos & derivados , Femenino , Melanoma Experimental/terapia , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo
7.
Proc Natl Acad Sci U S A ; 115(13): E2997-E3006, 2018 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-29531030

RESUMEN

Inherited retinal degeneration (RD) is a devastating and currently untreatable neurodegenerative condition that leads to loss of photoreceptor cells and blindness. The vast genetic heterogeneity of RD, the lack of "druggable" targets, and the access-limiting blood-retinal barrier (BRB) present major hurdles toward effective therapy development. Here, we address these challenges (i) by targeting cGMP (cyclic guanosine- 3',5'-monophosphate) signaling, a disease driver common to different types of RD, and (ii) by combining inhibitory cGMP analogs with a nanosized liposomal drug delivery system designed to facilitate transport across the BRB. Based on a screen of several cGMP analogs we identified an inhibitory cGMP analog that interferes with activation of photoreceptor cell death pathways. Moreover, we found liposomal encapsulation of the analog to achieve efficient drug targeting to the neuroretina. This pharmacological treatment markedly preserved in vivo retinal function and counteracted photoreceptor degeneration in three different in vivo RD models. Taken together, we show that a defined class of compounds for RD treatment in combination with an innovative drug delivery method may enable a single type of treatment to address genetically divergent RD-type diseases.


Asunto(s)
Barrera Hematorretinal/metabolismo , GMP Cíclico/análogos & derivados , GMP Cíclico/administración & dosificación , Modelos Animales de Enfermedad , Sistemas de Liberación de Medicamentos , Degeneración Retiniana/tratamiento farmacológico , Animales , Barrera Hematorretinal/efectos de los fármacos , GMP Cíclico/farmacología , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Liposomas , Ratones , Células Fotorreceptoras/metabolismo , Retina/efectos de los fármacos , Retina/metabolismo , Degeneración Retiniana/metabolismo , Transducción de Señal/efectos de los fármacos
8.
Braz J Med Biol Res ; 51(5): e6693, 2018 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-29561954

RESUMEN

Testosterone synthesis within Leydig cells is a calcium-dependent process. Intracellular calcium levels are regulated by different processes including ATP-activated P2X purinergic receptors, T-type Ca2+ channels modulated by the luteinizing hormone, and intracellular calcium storages recruited by a calcium-induced calcium release mechanism. On the other hand, nitric oxide (NO) is reported to have an inhibitory role in testosterone production. Based on these observations, we investigated the interaction between the purinergic and nitrergic systems in Leydig cells of adult mice. For this purpose, we recorded ATP-evoked currents in isolated Leydig cells using the whole cell patch clamp technique after treatment with L-NAME (300 µM and 1 mM), L-arginine (10, 100, 300, and 500 µM), ODQ (300 µM), and 8-Br-cGMP (100 µM). Our results show that NO produced by Leydig cells in basal conditions is insufficient to change the ATP-evoked currents and that extra NO provided by adding 300 µM L-arginine positively modulates the current through a mechanism involving the NO/cGMP signaling pathway. Thus, we report an interaction between the nitrergic and purinergic systems in Leydig cells and suggest that Ca2+ entry via the purinergic receptors can be regulated by NO.


Asunto(s)
Adenosina Trifosfato/fisiología , Células Intersticiales del Testículo/fisiología , Óxido Nítrico/fisiología , Receptores Purinérgicos/metabolismo , Potenciales de Acción , Animales , Arginina/administración & dosificación , Arginina/metabolismo , Células Cultivadas , GMP Cíclico/administración & dosificación , GMP Cíclico/análogos & derivados , GMP Cíclico/metabolismo , Masculino , Ratones , NG-Nitroarginina Metil Éster/administración & dosificación , NG-Nitroarginina Metil Éster/metabolismo , Óxido Nítrico/biosíntesis , Técnicas de Placa-Clamp , Tionucleótidos/administración & dosificación , Tionucleótidos/metabolismo
9.
Appl Environ Microbiol ; 84(7)2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29427430

RESUMEN

Pseudomonas aeruginosa exhibits flagellum-mediated swimming in liquid and swarming on hydrated surfaces under diverse nutrient conditions. Prior studies have implicated a phosphodiesterase, DipA, in regulating these flagellum-mediated motilities, but collectively, the necessity for DipA was unclear. In this study, we find that the medium composition conditionally constrains the influence of DipA on flagellar motility. We show that DipA exhibits more influence on minimal medium supplemented with glutamate or glucose, where flagellar motility was negated for the dipA mutant. Conversely, a dipA-deficient mutant exhibits flagellar motility when growing with LB Lennox broth and minimal medium supplemented with Casamino Acids. Swarming under these rich medium conditions occurs under elevated levels of c-di-GMP. We also demonstrate that the influence of DipA upon swimming often differs from that upon swarming, and we conclude that a direct comparison of the motility phenotypes is generally valid only when characterizing motility assay results from the same medium composition. Our results are consistent with the explanation that DipA is one of several phosphodiesterases responding to the nutrient environment sensed by P. aeruginosa On minimal medium with glutamate or glucose, DipA is dominant; however, on rich medium, the necessity of DipA is fully negated.IMPORTANCE Motile and ubiquitous bacteria such as Pseudomonas aeruginosa can quickly colonize surfaces and form biofilms in numerous environments such as water distribution systems, soil, and the human lung. To effectively disrupt bacterial colonization, it is imperative to understand how bacteria regulate motility in these different growth environments. Here, we show that the phosphodiesterase DipA is not required for flagellar motility under all nutrient conditions. Thus, the maintenance of intracellular c-di-GMP levels to promote flagellar motility or biofilm development must be conditionally regulated by differing phosphodiesterases in variation with select nutrient cues.


Asunto(s)
Proteínas Bacterianas/genética , GMP Cíclico/análogos & derivados , Regulación Bacteriana de la Expresión Génica/fisiología , Hidrolasas Diéster Fosfóricas/genética , Pseudomonas aeruginosa/genética , Proteínas Bacterianas/metabolismo , GMP Cíclico/administración & dosificación , Hidrolasas Diéster Fosfóricas/metabolismo , Pseudomonas aeruginosa/metabolismo
10.
Braz. j. med. biol. res ; 51(5): e6693, 2018. graf
Artículo en Inglés | LILACS | ID: biblio-889091

RESUMEN

Testosterone synthesis within Leydig cells is a calcium-dependent process. Intracellular calcium levels are regulated by different processes including ATP-activated P2X purinergic receptors, T-type Ca2+ channels modulated by the luteinizing hormone, and intracellular calcium storages recruited by a calcium-induced calcium release mechanism. On the other hand, nitric oxide (NO) is reported to have an inhibitory role in testosterone production. Based on these observations, we investigated the interaction between the purinergic and nitrergic systems in Leydig cells of adult mice. For this purpose, we recorded ATP-evoked currents in isolated Leydig cells using the whole cell patch clamp technique after treatment with L-NAME (300 μM and 1 mM), L-arginine (10, 100, 300, and 500 μM), ODQ (300 μM), and 8-Br-cGMP (100 μM). Our results show that NO produced by Leydig cells in basal conditions is insufficient to change the ATP-evoked currents and that extra NO provided by adding 300 μM L-arginine positively modulates the current through a mechanism involving the NO/cGMP signaling pathway. Thus, we report an interaction between the nitrergic and purinergic systems in Leydig cells and suggest that Ca2+ entry via the purinergic receptors can be regulated by NO.


Asunto(s)
Animales , Masculino , Ratones , Adenosina Trifosfato/fisiología , Receptores Purinérgicos/metabolismo , Células Intersticiales del Testículo/fisiología , Óxido Nítrico/fisiología , Arginina/administración & dosificación , Arginina/metabolismo , Tionucleótidos/administración & dosificación , Tionucleótidos/metabolismo , Potenciales de Acción , Células Cultivadas , GMP Cíclico/administración & dosificación , GMP Cíclico/análogos & derivados , GMP Cíclico/metabolismo , Técnicas de Placa-Clamp , NG-Nitroarginina Metil Éster/administración & dosificación , NG-Nitroarginina Metil Éster/metabolismo , Óxido Nítrico/biosíntesis
11.
J Clin Invest ; 127(6): 2176-2191, 2017 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-28436934

RESUMEN

Therapies using T cells that are programmed to express chimeric antigen receptors (CAR T cells) consistently produce positive results in patients with hematologic malignancies. However, CAR T cell treatments are less effective in solid tumors for several reasons. First, lymphocytes do not efficiently target CAR T cells; second, solid tumors create an immunosuppressive microenvironment that inactivates T cell responses; and third, solid cancers are typified by phenotypic diversity and thus include cells that do not express proteins targeted by the engineered receptors, enabling the formation of escape variants that elude CAR T cell targeting. Here, we have tested implantable biopolymer devices that deliver CAR T cells directly to the surfaces of solid tumors, thereby exposing them to high concentrations of immune cells for a substantial time period. In immunocompetent orthotopic mouse models of pancreatic cancer and melanoma, we found that CAR T cells can migrate from biopolymer scaffolds and eradicate tumors more effectively than does systemic delivery of the same cells. We have also demonstrated that codelivery of stimulator of IFN genes (STING) agonists stimulates immune responses to eliminate tumor cells that are not recognized by the adoptively transferred lymphocytes. Thus, these devices may improve the effectiveness of CAR T cell therapy in solid tumors and help protect against the emergence of escape variants.


Asunto(s)
Biopolímeros/administración & dosificación , Carcinoma Ductal Pancreático/terapia , Melanoma Experimental/terapia , Neoplasias Pancreáticas/terapia , Traslado Adoptivo , Animales , Células Presentadoras de Antígenos/fisiología , Antineoplásicos/administración & dosificación , Carcinoma Ductal Pancreático/inmunología , Línea Celular Tumoral , GMP Cíclico/administración & dosificación , GMP Cíclico/análogos & derivados , Portadores de Fármacos/administración & dosificación , Femenino , Implantes Experimentales , Melanoma Experimental/inmunología , Proteínas de la Membrana/agonistas , Ratones Endogámicos C57BL , Ratones Transgénicos , Trasplante de Neoplasias , Neoplasias Pancreáticas/inmunología , Linfocitos T/fisiología
12.
Brain Behav Immun ; 57: 360-370, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27189036

RESUMEN

Patients with hepatic encephalopathy (HE) show working memory and visuo-spatial orientation deficits. Hyperammonemia is a main contributor to cognitive impairment in HE. Hyperammonemic rats show impaired spatial learning and learning ability in the Y maze. Intracerebral administration of extracellular cGMP restores learning in the Y-maze. The underlying mechanisms remain unknown. It also remains unknown whether extracellular cGMP improves neuroinflammation or restores spatial learning in hyperammonemic rats and if it affects differently reference and working memory. The aims of this work were: Spatial working and reference memory were assessed using the radial and Morris water mazes and neuroinflammation by immunohistochemistry and Western blot. Membrane expression of NMDA and AMPA receptor subunits was analyzed using the BS3 crosslinker. Extracellular cGMP was administered intracerebrally using osmotic minipumps. Chronic hyperammonemia induces neuroinflammation in hippocampus, with astrocytes activation and increased IL-1ß, which are associated with increased NMDA receptors membrane expression and impaired working memory. This process is not affected by extracellular cGMP. Hyperammonemia also activates microglia and increases TNF-α, alters membrane expression of AMPA receptor subunits (increased GluA1 and reduced GluA2) and impairs reference memory. All these changes are reversed by extracellular cGMP. These results show that extracellular cGMP modulates spatial reference memory but not working memory. This would be mediated by modulation of TNF-α levels and of membrane expression of GluA1 and GluA2 subunits of AMPA receptors.


Asunto(s)
Disfunción Cognitiva/metabolismo , GMP Cíclico/farmacología , Hipocampo/metabolismo , Hiperamonemia/metabolismo , Inflamación/metabolismo , Interleucina-1beta/metabolismo , Memoria a Corto Plazo , Receptores AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Memoria Espacial , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/etiología , GMP Cíclico/administración & dosificación , Modelos Animales de Enfermedad , Hipocampo/efectos de los fármacos , Hiperamonemia/complicaciones , Hiperamonemia/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Inflamación/etiología , Interleucina-1beta/efectos de los fármacos , Masculino , Memoria a Corto Plazo/efectos de los fármacos , Ratas , Ratas Wistar , Receptores AMPA/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Memoria Espacial/efectos de los fármacos , Factor de Necrosis Tumoral alfa/efectos de los fármacos
13.
Cancer Res ; 76(8): 2137-52, 2016 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-26951929

RESUMEN

Endoplasmic reticulum (ER) stress responses through the IRE-1/XBP-1 pathway are required for the function of STING (TMEM173), an ER-resident transmembrane protein critical for cytoplasmic DNA sensing, IFN production, and cancer control. Here we show that the IRE-1/XBP-1 pathway functions downstream of STING and that STING agonists selectively trigger mitochondria-mediated apoptosis in normal and malignant B cells. Upon stimulation, STING was degraded less efficiently in B cells, implying that prolonged activation of STING can lead to apoptosis. Transient activation of the IRE-1/XBP-1 pathway partially protected agonist-stimulated malignant B cells from undergoing apoptosis. In Eµ-TCL1 mice with chronic lymphocytic leukemia, injection of the STING agonist 3'3'-cGAMP induced apoptosis and tumor regression. Similarly efficacious effects were elicited by 3'3'-cGAMP injection in syngeneic or immunodeficient mice grafted with multiple myeloma. Thus, in addition to their established ability to boost antitumoral immune responses, STING agonists can also directly eradicate malignant B cells. Cancer Res; 76(8); 2137-52. ©2016 AACR.


Asunto(s)
Apoptosis/fisiología , Linfocitos B/metabolismo , Proteínas de la Membrana/agonistas , Animales , Linfocitos B/citología , Línea Celular , GMP Cíclico/administración & dosificación , GMP Cíclico/farmacología , Inyecciones Intraperitoneales , Proteínas de la Membrana/fisiología , Ratones
14.
Stem Cells ; 33(10): 2973-84, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26269433

RESUMEN

Cardiac hypertrophy is an abnormal enlargement of heart muscle. It frequently results in congestive heart failure, which is a leading cause of human death. Previous studies demonstrated that the nitric oxide (NO), cyclic GMP (cGMP), and protein kinase G (PKG) signaling pathway can inhibit cardiac hypertrophy and thus improve cardiac function. However, the underlying mechanisms are not fully understood. Here, based on the human embryonic stem cell-derived cardiomyocyte (hESC-CM) model system, we showed that Orai1, the pore-forming subunit of store-operated Ca(2+) entry (SOCE), is the downstream effector of PKG. Treatment of hESC-CMs with an α-adrenoceptor agonist phenylephrine (PE) caused a marked hypertrophy, which was accompanied by an upregulation of Orai1. Moreover, suppression of Orai1 expression/activity using Orai1-siRNAs or a dominant-negative construct Orai1(G98A) inhibited the hypertrophy, suggesting that Orai1-mediated SOCE is indispensable for the PE-induced hypertrophy of hESC-CMs. In addition, the hypertrophy was inhibited by NO and cGMP via activating PKG. Importantly, substitution of Ala for Ser(34) in Orai1 abolished the antihypertrophic effects of NO, cGMP, and PKG. Furthermore, PKG could directly phosphorylate Orai1 at Ser(34) and thus prevent Orai1-mediated SOCE. Together, we conclude that NO, cGMP, and PKG inhibit the hypertrophy of hESC-CMs via PKG-mediated phosphorylation on Orai1-Ser-34. These results provide novel mechanistic insights into the action of cGMP-PKG-related antihypertrophic agents, such as NO donors and sildenafil.


Asunto(s)
Canales de Calcio/genética , Cardiomegalia/genética , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Insuficiencia Cardíaca/genética , Calcio/metabolismo , Canales de Calcio/biosíntesis , Canales de Calcio/efectos de los fármacos , Cardiomegalia/patología , Diferenciación Celular/genética , GMP Cíclico/administración & dosificación , GMP Cíclico/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Insuficiencia Cardíaca/patología , Células Madre Embrionarias Humanas/efectos de los fármacos , Células Madre Embrionarias Humanas/metabolismo , Humanos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Óxido Nítrico/administración & dosificación , Óxido Nítrico/metabolismo , Proteína ORAI1 , Fenilefrina/administración & dosificación , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
15.
J Mol Cell Cardiol ; 87: 74-8, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26219954

RESUMEN

The Exchange Protein directly Activated by cAMP (EPAC) participates to the pathological signaling of cardiac hypertrophy and heart failure, in which the role of Ca(2+) entry through the Transient Receptor Potential Canonical (TRPC) channels begin to be appreciated. Here we studied whether EPAC activation could influence the activity and/or expression of TRPC channels in cardiac myocytes. In adult rat ventricular myocytes treated for 4 to 6h with the selective EPAC activator, 8-pCPT (10µM), we observed by Fluo-3 confocal fluorescence a Store-Operated Ca(2+) Entry (SOCE) like-activity, which was blunted by co-incubation with EPAC inhibitors (ESI-05 and CE3F4 at 10 µM). This SOCE-like activity, which was very small in control incubated cells, was sensitive to 30-µM SKF-96365. Molecular screening showed a specific upregulation of TRPC3 and C4 protein isoforms after 8-pCPT treatment. Moreover, sustained EPAC activation favored proarrhythmic Ca(2+) waves, which were reduced either by co-incubation with EPAC inhibitors or bath perfusion with TRPC inhibitors. Our study provides the first evidence that sustained selective EPAC activation leads to an increase in TRPC3 and C4 protein expression and induces a proarrhythmic SOCE-like activity in adult rat ventricular cardiomyocytes, which might be of importance during the development of cardiac diseases.


Asunto(s)
Cardiomegalia/genética , Complemento C4/biosíntesis , Factores de Intercambio de Guanina Nucleótido/biosíntesis , Miocitos Cardíacos/metabolismo , Canales Catiónicos TRPC/genética , Animales , Derivados del Benceno/administración & dosificación , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Cardiomegalia/tratamiento farmacológico , Cardiomegalia/patología , Complemento C4/genética , AMP Cíclico/metabolismo , GMP Cíclico/administración & dosificación , GMP Cíclico/análogos & derivados , Factores de Intercambio de Guanina Nucleótido/genética , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/patología , Humanos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Quinolinas/administración & dosificación , Ratas , Sulfonas/administración & dosificación , Canales Catiónicos TRPC/antagonistas & inhibidores , Tionucleótidos/administración & dosificación
16.
Cancer Immunol Immunother ; 64(8): 1057-66, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25986168

RESUMEN

Therapeutic vaccines to induce anti-tumor CD8 T cells have been used in clinical trials for advanced melanoma patients, but the clinical response rate and overall survival time have not improved much. We believe that these dismal outcomes are caused by inadequate number of antigen-specific CD8 T cells generated by most vaccines. In contrast, huge CD8 T cell responses readily occur during acute viral infections. High levels of type-I interferon (IFN-I) are produced during these infections, and this cytokine not only exhibits anti-viral activity but also promotes CD8 T cell responses. The studies described here were performed to determine whether promoting the production of IFN-I could enhance the potency of a peptide vaccine. We report that cyclic diguanylate monophosphate (c-di-GMP), which activates the stimulator of interferon genes, potentiated the immunogenicity and anti-tumor effects of a peptide vaccine against mouse B16 melanoma. The synergistic effects of c-di-GMP required co-administration of costimulatory anti-CD40 antibody, the adjuvant poly-IC, and were mediated in part by IFN-I. These findings demonstrate that peptides representing CD8 T cell epitopes can be effective inducers of large CD8 T cell responses in vaccination strategies that mimic acute viral infections.


Asunto(s)
Linfocitos T CD8-positivos/efectos de los fármacos , Vacunas contra el Cáncer/uso terapéutico , GMP Cíclico/análogos & derivados , Inmunoterapia/métodos , Melanoma Experimental/terapia , Neoplasias Cutáneas/terapia , Animales , Anticuerpos Monoclonales/administración & dosificación , Antígenos CD40/inmunología , Linfocitos T CD8-positivos/inmunología , GMP Cíclico/administración & dosificación , GMP Cíclico/efectos adversos , Epítopos de Linfocito T/administración & dosificación , Humanos , Interferón Tipo I/inmunología , Activación de Linfocitos/efectos de los fármacos , Melanoma Experimental/inmunología , Proteínas de la Membrana/agonistas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Poli I-C/administración & dosificación , Receptores de Interferón/genética , Neoplasias Cutáneas/inmunología , Vacunas de Subunidad/uso terapéutico
17.
Cell Rep ; 11(1): 71-84, 2015 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-25843711

RESUMEN

Upon systemic bacterial infection, hematopoietic stem and progenitor cells (HSPCs) migrate to the periphery in order to supply a sufficient number of immune cells. Although pathogen-associated molecular patterns reportedly mediate HSPC activation, how HSPCs detect pathogen invasion in vivo remains elusive. Bacteria use the second messenger bis-(3'-5')-cyclic dimeric guanosine monophosphate (c-di-GMP) for a variety of activities. Here, we report that c-di-GMP comprehensively regulated both HSPCs and their niche cells through an innate immune sensor, STING, thereby inducing entry into the cell cycle and mobilization of HSPCs while decreasing the number and repopulation capacity of long-term hematopoietic stem cells. Furthermore, we show that type I interferon acted as a downstream target of c-di-GMP to inhibit HSPC expansion in the spleen, while transforming growth factor-ß was required for c-di-GMP-dependent splenic HSPC expansion. Our results define machinery underlying the dynamic regulation of HSPCs and their niches during bacterial infection through c-di-GMP/STING signaling.


Asunto(s)
GMP Cíclico/análogos & derivados , Células Madre Hematopoyéticas/metabolismo , Inmunidad Innata , Factor 3 Regulador del Interferón/genética , Proteínas de la Membrana/genética , Animales , Bacterias/metabolismo , Bacterias/patogenicidad , GMP Cíclico/administración & dosificación , GMP Cíclico/inmunología , GMP Cíclico/metabolismo , Regulación de la Expresión Génica , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/microbiología , Factor 3 Regulador del Interferón/inmunología , Proteínas de la Membrana/inmunología , Ratones , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Nicho de Células Madre/inmunología , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/inmunología
18.
Hum Vaccin Immunother ; 11(5): 1235-43, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25714901

RESUMEN

Highly pathogenic avian influenza H5N1 infection remains a public health threat and vaccination is the best measure of limiting the impact of a potential pandemic. Mucosal vaccines have the advantage of eliciting immune responses at the site of viral entry, thereby preventing infection as well as further viral transmission. In this study, we assessed the protective efficacy of hemagglutinin (HA) from the A/Indonesia/05/05 (H5N1) strain of influenza virus that was produced by transient expression in plants. The plant-derived vaccine, in combination with the mucosal adjuvant (3',5')-cyclic dimeric guanylic acid (c-di-GMP) was used for intranasal immunization of mice and ferrets, before challenge with a lethal dose of the A/Indonesia/05/05 (H5N1) virus. Mice vaccinated with 15 µg or 5 µg of adjuvanted HA survived the viral challenge, while all control mice died within 10 d of challenge. Vaccinated animals elicited serum hemagglutination inhibition, IgG and IgA antibody titers. In the ferret challenge study, all animals vaccinated with the adjuvanted plant vaccine survived the lethal viral challenge, while 50% of the control animals died. In both the mouse and ferret models, the vaccinated animals were better protected from weight loss and body temperature changes associated with H5N1 infection compared with the non-vaccinated controls. Furthermore, the systemic spread of the virus was lower in the vaccinated animals compared with the controls. Results presented here suggest that the plant-produced HA-based influenza vaccine adjuvanted with c-di-GMP is a promising vaccine/adjuvant combination for the development of new mucosal influenza vaccines.


Asunto(s)
Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Subtipo H5N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Administración Intranasal , Animales , GMP Cíclico/administración & dosificación , GMP Cíclico/análogos & derivados , Modelos Animales de Enfermedad , Femenino , Hurones , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Subtipo H5N1 del Virus de la Influenza A/genética , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/genética , Vacunas contra la Influenza/aislamiento & purificación , Masculino , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/prevención & control , Plantas Modificadas Genéticamente , Análisis de Supervivencia , Resultado del Tratamiento , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Vacunas Sintéticas/aislamiento & purificación
19.
Cancer Immunol Res ; 2(9): 901-10, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24913717

RESUMEN

Cancer vaccination may be our best and most benign option for preventing or treating metastatic cancer. However, breakthroughs are hampered by immune suppression in the tumor microenvironment. In this study, we analyzed whether cyclic diguanylate (c-di-GMP), a ligand for stimulator of interferon genes (STING), could overcome immune suppression and improve vaccination against metastatic breast cancer. Mice with metastatic breast cancer (4T1 model) were therapeutically immunized with an attenuated Listeria monocytogenes (LM)-based vaccine, expressing tumor-associated antigen Mage-b (LM-Mb), followed by multiple low doses of c-di-GMP (0.2 µmol/L). This treatment resulted in a striking and near elimination of all metastases. Experiments revealed that c-di-GMP targets myeloid-derived suppressor cells (MDSC) and tumor cells. Low doses of c-di-GMP significantly increased the production of IL12 by MDSCs, in correlation with improved T-cell responses to Mage-b, whereas a high dose of c-di-GMP (range, 0.3-3 mmol/L) activated caspase-3 in the 4T1 tumor cells and killed the tumor cells directly. On the basis of these results, we tested one administration of high-dose c-di-GMP (3 mmol/L) followed by repeated administrations of low-dose c-di-GMP (0.2 µmol/L) in the 4T1 model, and found equal efficacy compared with the combination of LM-Mb and c-di-GMP. This finding correlated with a mechanism of improved CD8 T-cell responses to tumor-associated antigens (TAA) Mage-b and Survivin, most likely through cross-presentation of these TAAs from c-di-GMP-killed 4T1 tumor cells, and through c-di-GMP-activated TAA-specific T cells. Our results demonstrate that activation of STING-dependent pathways by c-di-GMP is highly attractive for cancer immunotherapy.


Asunto(s)
Neoplasias de la Mama/secundario , Neoplasias de la Mama/terapia , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/uso terapéutico , GMP Cíclico/análogos & derivados , Proteínas de la Membrana/genética , Animales , Antígenos de Neoplasias/genética , Línea Celular Tumoral , Reactividad Cruzada , GMP Cíclico/administración & dosificación , GMP Cíclico/genética , Femenino , Células HEK293 , Humanos , Inmunoterapia , Listeria monocytogenes/genética , Neoplasias Mamarias Experimentales , Ratones , Ratones Endogámicos BALB C , Microambiente Tumoral , Vacunas Atenuadas/uso terapéutico
20.
J Cell Physiol ; 229(12): 2142-52, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24911927

RESUMEN

Published data provide strong evidence that heparin treatment of proliferating vascular smooth muscle cells results in decreased signaling through the ERK pathway and decreases in cell proliferation. In addition, these changes have been shown to be mimicked by antibodies that block heparin binding to the cell surface. Here, we provide evidence that the activity of protein kinase G is required for these heparin effects. Specifically, a chemical inhibitor of protein kinase G, Rp-8-pCPT-cGMS, eliminates heparin and anti-heparin receptor antibody effects on bromodeoxyuridine incorporation into growth factor-stimulated cells. In addition, protein kinase G inhibitors decrease heparin effects on ERK activity, phosphorylation of the transcription factor Elk-1, and heparin-induced MKP-1 synthesis. Although transient, the levels of cGMP increase in heparin treated cells. Finally, knock down of protein kinase G also significantly decreases heparin effects in growth factor-activated vascular smooth muscle cells. Together, these data indicate that heparin effects on vascular smooth muscle cell proliferation depend, at least in part, on signaling through protein kinase G.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Heparina/metabolismo , Músculo Liso Vascular/metabolismo , Animales , GMP Cíclico/administración & dosificación , GMP Cíclico/análogos & derivados , GMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/genética , Regulación de la Expresión Génica/efectos de los fármacos , Heparina/administración & dosificación , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Fosforilación/efectos de los fármacos , Ratas , Receptores de Superficie Celular/metabolismo , Tionucleótidos/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...