Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Crit Care ; 23(1): 279, 2019 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-31412949

RESUMEN

BACKGROUND: The rapid development in big data analytics and the data-rich environment of intensive care units together provide unprecedented opportunities for medical breakthroughs in the field of critical care. We developed and validated a machine learning-based model, the Pediatric Risk of Mortality Prediction Tool (PROMPT), for real-time prediction of all-cause mortality in pediatric intensive care units. METHODS: Utilizing two separate retrospective observational cohorts, we conducted model development and validation using a machine learning algorithm with a convolutional neural network. The development cohort comprised 1445 pediatric patients with 1977 medical encounters admitted to intensive care units from January 2011 to December 2017 at Severance Hospital (Seoul, Korea). The validation cohort included 278 patients with 364 medical encounters admitted to the pediatric intensive care unit from January 2016 to November 2017 at Samsung Medical Center. RESULTS: Using seven vital signs, along with patient age and body weight on intensive care unit admission, PROMPT achieved an area under the receiver operating characteristic curve in the range of 0.89-0.97 for mortality prediction 6 to 60 h prior to death. Our results demonstrated that PROMPT provided high sensitivity with specificity and outperformed the conventional severity scoring system, the Pediatric Index of Mortality, in predictive ability. Model performance was indistinguishable between the development and validation cohorts. CONCLUSIONS: PROMPT is a deep model-based, data-driven early warning score tool that can predict mortality in critically ill children and may be useful for the timely identification of deteriorating patients.


Asunto(s)
Enfermedad Crítica/mortalidad , Aprendizaje Profundo , Mortalidad/tendencias , Pediatría/instrumentación , Medición de Riesgo/métodos , Adolescente , Área Bajo la Curva , Macrodatos , Niño , Preescolar , Estudios de Cohortes , Femenino , Humanos , Lactante , Masculino , Pediatría/métodos , Pediatría/normas , Curva ROC , República de Corea , Estudios Retrospectivos
2.
Dis Model Mech ; 11(7)2018 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-29967068

RESUMEN

Inflammatory bowel disease (IBD) is a chronic inflammatory disease, in which the intestinal epithelium loses its barrier function. Given the existence of the oxygen gradient in the intestinal epithelium and that inflammation further contributes to the tissue hypoxia, we investigated the role of hypoxia-inducible factor (HIF), a transcription factor activated under hypoxic conditions in myeloid cells, in the progression of IBD. To do this, we utilized myeloid-specific knockout (KO) mice targeting HIF pathways, created by a Cre-loxP system with human MRP8 (hMRP8), an intracellular calcium-binding protein, as the myeloid promoter. By feeding 5% dextran sodium sulfate (DSS) to hMRP8 von Hippel Lindau (Vhl) KO mice, in which HIF-1α and HIF-2α are constitutively activated in myeloid cells, we found that these mice were highly susceptible to DSS-induced colitis, demonstrating greater body weight loss, increased mortality, faster onset of rectal bleeding, shortened colon length, and increased CD11b- or Gr-1-positive myeloid cells in the colon compared with wild-type (WT) mice. These parameters were restored to, if not better than, the WT levels when we examined hMRP8 Hif-1a KO mice upon 5% DSS feeding. hMRP8 Hif-2a KO mice, on the other hand, exhibited a similar degree of DSS-induced colitis to that of WT mice. Lastly, when DSS was given together with azoxymethane to induce tumorigenesis in the colon, we found that hMRP8 Hif-1a KO mice exhibited comparable levels of colorectal tumors to those of WT mice, indicating that HIF-1α in myeloid cells is dispensable for tumorigenesis. Collectively, our results suggest that HIF-1α activation in myeloid cells critically regulates IBD progression.


Asunto(s)
Colitis/inducido químicamente , Colitis/patología , Progresión de la Enfermedad , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Células Mieloides/metabolismo , Células Mieloides/patología , Transportadoras de Casetes de Unión a ATP/genética , Animales , Antígenos Ly/metabolismo , Azoximetano , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Antígeno CD11b/metabolismo , Carcinogénesis/metabolismo , Carcinogénesis/patología , Colitis/metabolismo , Colitis/prevención & control , Colon/metabolismo , Colon/patología , Sulfato de Dextran , Susceptibilidad a Enfermedades , Humanos , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas/genética , Factor de Necrosis Tumoral alfa/metabolismo , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo
3.
Immunol Lett ; 176: 36-43, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27229912

RESUMEN

IL-12p40 homodimer, a natural antagonist of IL-12 and IL-23, performs an important role in the expression of proinflammatory cytokines that is essential for Th1 and Th17 immune responses. Here, we reveal the therapeutic and immunosuppressive effect of the IL-12p40 subunit ((p40)2-Fc) in an experimental autoimmune arthritis model. We hypothesized that (p40)2-Fc may reduce the inflammatory response and the activation of T cells. In this study, we intraperitoneally injected (p40)2-Fc into collagen induced arthritis (CIA) mice to identify whether (p40)2-Fc attenuates CIA severity. (p40)2-Fc reduced the development of CIA, joint inflammation and cartilage destruction. (p40)2-Fc also significantly decreased the concentration of serum immunoglobulin as well as the number of T cells and C II specific T cells. In addition, osteoclastogenesis in (p40)2-Fc treated mice was down-regulated compared to the mice treated with (p40)2-Fc control. We observed that (p40)2-Fc treatment alleviates arthritis in mice with CIA, reducing inflammation and osteoclast differentiation. These findings suggest that (p40)2-Fc can be a potential therapeutic approach for autoimmune arthritis.


Asunto(s)
Antiinflamatorios/uso terapéutico , Artritis Experimental/terapia , Fragmentos Fc de Inmunoglobulinas/uso terapéutico , Inmunoterapia/métodos , Subunidad p40 de la Interleucina-12/inmunología , Linfocitos T/inmunología , Animales , Formación de Anticuerpos/efectos de los fármacos , Artritis Experimental/inmunología , Células Cultivadas , Colágeno Tipo II/inmunología , Terapia de Inmunosupresión , Inyecciones Intraperitoneales , Masculino , Ratones , Ratones Endogámicos DBA , Osteogénesis/efectos de los fármacos
4.
J Allergy Clin Immunol ; 137(5): 1466-1476.e3, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26824999

RESUMEN

BACKGROUND: Psoriasis is one of the most common chronic inflammatory diseases of the skin. Recently, IL-17-producing T cells have been shown to play a critical role in psoriatic inflammation. Programmed cell death 1 (PD-1) is a coinhibitory receptor expressed on T cells in various chronic inflammatory diseases; however, the expression and function of PD-1 during psoriatic inflammation have not previously been characterized. OBJECTIVE: We examined PD-1 expression on IL-17A-producing T cells from imiquimod-treated mice and patients with psoriasis. Additionally, we investigated the therapeutic effect of recombinant programmed cell death ligand 1 (PD-L1) protein on imiquimod-induced psoriatic inflammation. METHODS: PD-1 expression on IL-17A-producing γδ T cells from imiquimod-treated mice was examined by means of multicolor flow cytometric analysis. In the psoriatic skin of patients, PD-1 and IL-17A expression was analyzed by using immunofluorescence. The therapeutic effect of PD-L1-Fc fusion protein (PD-L1-Fc) was assessed in imiquimod-treated mice ex vivo and in vivo. RESULTS: During imiquimod-induced psoriatic inflammation, PD-1 is overexpressed on CD27(-)Vγ1(-) γδ T cells. Furthermore, PD-1 expression on IL-17A(+) T cells was confirmed in psoriatic skin tissues from patients and imiquimod-treated mice. In the CD27(-)Vγ1(-) γδ T-cell population, Vγ4(-) γδ T cells with Vγ6 mRNA expression showed a high level of PD-1 expression. Furthermore, these PD-1(hi)Vγ4(-) (Vγ6(+)) γδ T cells were specialized for anti-CD3-induced IL-17A production, which was inhibited by PD-L1-Fc treatment. In imiquimod-treated mice PD-L1-Fc reduced psoriatic inflammation when given alone and enhanced the therapeutic effect of anti-p40 when given in combination. CONCLUSION: PD-1 is overexpressed in IL-17A-producing T cells in both imiquimod-treated mice and patients with psoriasis. Moreover, recombinant PD-L1-Fc alleviates psoriatic inflammation in imiquimod-treated mice.


Asunto(s)
Interleucina-17/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo , Psoriasis/metabolismo , Subgrupos de Linfocitos T/metabolismo , Adyuvantes Inmunológicos , Aminoquinolinas , Animales , Antígeno B7-H1/farmacología , Antígeno B7-H1/uso terapéutico , Humanos , Imiquimod , Inflamación/metabolismo , Ratones Endogámicos C57BL , Psoriasis/inducido químicamente , Psoriasis/tratamiento farmacológico , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Piel/efectos de los fármacos , Piel/metabolismo
5.
Gut ; 64(2): 260-71, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24902766

RESUMEN

OBJECTIVE: Programmed death-ligand 1 (PD-L1) has been shown to negatively regulate immune responses via its interaction with PD-1 receptor. In this study, we investigated the effects of PD-L1-Fc treatment on intestinal inflammation using two murine models of inflammatory colitis induced by dextran sulfate sodium (DSS) and T-cell transfer. DESIGN: The anti-colitis effect of adenovirus expressing Fc-conjugated PD-L1 (Ad/PD-L1-Fc) and recombinant PD-L1-Fc protein was evaluated in DSS-treated wild-type and Rag-1 knockout (KO) mice. We examined differentiation of T-helper cells, frequency of innate immune cells, and cytokine production by dendritic cells (DCs) in the colon from DSS-treated mice after PD-L1-Fc administration. In Rag-1 KO mice reconstituted with CD4 CD45RB(high) T cells, we assessed the treatment effect of PD-L1-Fc protein on the development of colitis. RESULTS: Administration of Ad/PD-L1-Fc significantly ameliorated DSS-induced colitis, which was accompanied by diminished frequency of interleukin (IL)-17A-producing CD4 T cells and increased interferon-γ-producing CD4 T cells in the colon of DSS-fed mice. The anti-colitic effect of PD-L1-Fc treatment was also observed in DSS-treated Rag-1 KO mice, indicating lymphoid cell independency. PD-L1-Fc modulated cytokine production by colonic DCs and the effect was dependent on PD-1 expression. Furthermore, PD-L1-Fc protein could significantly reduce the severity of colitis in CD4 CD45RB(high) T-cell-transferred Rag-1 KO mice. CONCLUSIONS: Based on the protective effect of PD-L1-Fc against DSS-induced and T-cell-induced colitis, our results suggest that PD-1-mediated inhibitory signals have a crucial role in limiting the development of colonic inflammation. This implicates that PD-L1-Fc may provide a novel therapeutic approach to treat inflammatory bowel disease.


Asunto(s)
Antígeno B7-H1/uso terapéutico , Colitis Ulcerosa/prevención & control , Factores Inmunológicos/uso terapéutico , Enfermedad Aguda , Adenoviridae/genética , Animales , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Antígeno B7-H1/farmacología , Diferenciación Celular/efectos de los fármacos , Colitis Ulcerosa/etiología , Colitis Ulcerosa/inmunología , Colitis Ulcerosa/patología , Colon/inmunología , Citocinas/biosíntesis , Células Dendríticas/inmunología , Sulfato de Dextran , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos/métodos , Vectores Genéticos , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Inmunidad Innata , Inmunidad Mucosa , Fragmentos Fc de Inmunoglobulinas/genética , Fragmentos Fc de Inmunoglobulinas/metabolismo , Fragmentos Fc de Inmunoglobulinas/farmacología , Fragmentos Fc de Inmunoglobulinas/uso terapéutico , Factores Inmunológicos/genética , Factores Inmunológicos/metabolismo , Factores Inmunológicos/farmacología , Mucosa Intestinal/inmunología , Transfusión de Linfocitos , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Recombinantes de Fusión/farmacología , Proteínas Recombinantes de Fusión/uso terapéutico , Linfocitos T Colaboradores-Inductores/inmunología , Células Th17/inmunología
6.
Exp Mol Med ; 45: e46, 2013 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-24091748

RESUMEN

Interleukin (IL)-27 is a novel cytokine of the IL-6/IL-12 family that has been reported to be involved in the pathogenesis of autoimmune diseases and has a pivotal role as both a pro- and anti-inflammatory cytokine. We investigated the in vivo effects of IL-27 on arthritis severity in a murine collagen-induced arthritis (CIA) model and its mechanism of action regarding control of regulatory T (Tregs) and IL-17-producing T helper 17 (Th17) cells. IL-27-Fc-treated CIA mice showed a lower severity of arthritis. IL-17 expression in the spleens was significantly decreased in IL-27-Fc-treated CIA mice compared with that in the CIA model. The Th17 population was decreased in the spleens of IL-27-Fc-treated CIA mice, whereas the CD4(+)CD25(+)Foxp3(+) Treg population increased. In vitro studies revealed that IL-27 inhibited IL-17 production in murine CD4(+) T cells, and the effect was associated with retinoic acid-related orphan receptor γT and signal transducer and activator of transcription 3 inhibition. In contrast, fluorescein isothiocyanate-labeled forkhead box P3 (Foxp3) and IL-10 were profoundly augmented by IL-27 treatment. Regarding the suppressive capacity of Treg cells, the proportions of CTLA-4(+) (cytotoxic T-lymphocyte antigen 4), PD-1(+) (programmed cell death protein 1) and GITR(+) (glucocorticoid-induced tumor necrosis factor receptor) Tregs increased in the spleens of IL-27-Fc-treated CIA mice. Furthermore, in vitro differentiated Treg cells with IL-27 exerted a more suppressive capacity on T-cell proliferation. We found that IL-27 acts as a reciprocal regulator of the Th17 and Treg populations in CD4(+) cells isolated from healthy human peripheral blood mononuclear cells (PBMCs), as well as from humans with rheumatoid arthritis (RA) PBMCs. Our study suggests that IL-27 has the potential to ameliorate overwhelming inflammation in patients with RA through a reciprocal regulation of Th17 and Treg cells.


Asunto(s)
Artritis Experimental/tratamiento farmacológico , Interleucinas/uso terapéutico , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Animales , Artritis Experimental/inmunología , Células Cultivadas , Humanos , Interleucinas/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA
7.
Blood ; 121(22): 4484-92, 2013 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-23610371

RESUMEN

Interleukin-7 (IL-7) is essential to T-cell survival as well as homeostatic proliferation, and clinical trials that exploit the mitogenic effects of IL-7 have achieved success in treating human diseases. In mice, the in vivo potency of IL-7 improves dramatically when it is administered as a complex with the anti-IL-7 neutralizing monoclonal antibody clone M25. However, the mechanism whereby M25 augments IL-7 potency is unknown. We have analyzed the discrete contributions of the antibody constant (Fc) and IL-7-binding (Fab) domains to the mechanism. By engaging the neonatal Fc receptor the Fc domain extends the in vivo lifespan of IL-7/M25 complexes and accounts for the majority of their activity. Unexpectedly, the IL-7-neutralizing Fab domain provides an additional, albeit smaller, contribution, possibly by serving as a cytokine depot. This study is the first to demonstrate that the neutralizing aspect of the monoclonal antibody is directly involved in enhancing the potency of a cytokine with a single form of receptor. Lessons from the mechanism of IL-7/M25 complexes inform the design of next-generation cytokine therapeutics.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Fragmentos Fc de Inmunoglobulinas/inmunología , Inmunoglobulina G/inmunología , Interleucina-7/inmunología , Receptores de Interleucina-7/inmunología , Traslado Adoptivo , Animales , Anticuerpos Monoclonales/metabolismo , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/metabolismo , Benzofuranos , Unión Competitiva/inmunología , Femenino , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Fragmentos Fc de Inmunoglobulinas/metabolismo , Inmunoglobulina G/metabolismo , Interleucina-7/metabolismo , Masculino , Ratones , Ratones Transgénicos , Quinolinas , Receptores Fc/inmunología , Receptores Fc/metabolismo , Receptores de Interleucina-7/metabolismo , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
8.
Vaccine ; 31(27): 2884-90, 2013 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-23624092

RESUMEN

Improvement to the immunogenicity of DNA vaccines was evaluated in a Mycobacterium tuberculosis (MTB) infection mouse model examining the combined effects of nonlytic Fc-fused IL-7 DNA (IL-7-nFc) and Flt3-ligand fused Mtb32 (F-Mtb32) DNA. Mice were treated with conventional chemotherapy for 6 weeks from 4 weeks after aerosol infection of MTB. Following the start of chemotherapy, DNA immunizations were administered five times with 2-week intervals. Coadministration of IL-7-nFc and F-Mtb32 DNA given during chemotherapy synergistically enhanced the magnitude of Mtb32-specific T cell responses and sustained for one-year after the last immunization assessed by IFN-γ ELISPOT assay. After dexamethasone treatment, a significantly reduced MTB reactivation was observed in mice received both IL-7-nFc and F-Mtb32 DNA, compared with F-MTb32 DNA alone or with control mice. In addition, mice treated with IL-7-nFc and F-Mtb32 DNA together showed improved lung pathology and reduced pulmonary inflammation values relative to F-Mtb32 DNA or saline injected mice. Intracellular cytokine staining revealed that the protection levels induced by combination therapy with IL-7-nFc and F-Mtb32 DNA was associated with enhanced Mtb32-specific IFN-γ secreting CD4(+) T cell responses and CD8(+) T cell responses stimulated with CTL epitope peptide in the lungs and spleens. These data suggest that IL-7-nFc as a novel TB adjuvant may facilitate therapeutic TB DNA vaccine to the clinics through significant enhancement of codelivered DNA vaccine-induced T cell immunity.


Asunto(s)
Antígenos Bacterianos/inmunología , Fragmentos Fc de Inmunoglobulinas/inmunología , Interleucina-7/inmunología , Proteínas de la Membrana/inmunología , Vacunas contra la Tuberculosis/inmunología , Tuberculosis Pulmonar/prevención & control , Vacunas de ADN/inmunología , Animales , Antígenos Bacterianos/genética , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Sinergismo Farmacológico , Femenino , Fragmentos Fc de Inmunoglobulinas/genética , Interferón gamma/biosíntesis , Interleucina-7/genética , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Mycobacterium tuberculosis/inmunología , Tuberculosis Pulmonar/inmunología , Vacunación
9.
Immunology ; 137(4): 326-35, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22812379

RESUMEN

Interleukin (IL)-27 is a heterodimeric cytokine that is known to have both stimulatory and inhibitory functions during immune responses. We investigated the effects of IL-27 on arthritis and bone erosion in the murine collagen-induced arthritis (CIA) model. We demonstrate that the inhibitory effect of IL-27 on osteoclastogenesis is associated with interferon-γ (IFN-γ) production by using an IFN-γ knockout mouse model. The IL-27-Fc was injected into both CIA and IFN-γ-deficient mice. The effects of IL-27-Fc on osteoclast differentiation were evaluated both in vitro and in vivo. The IL-27-Fc-injected mice showed significantly lower arthritis indices and fewer tartrate-resistant acid-phosphatase-positive osteoclasts in their joint tissues than untreated mice. Interleukin-27 inhibited osteoclastogenesis from bone marrow-derived mononuclear cells in vitro, which was counteracted by the addition of anti-IFN-γ antibody. The IL-27-Fc did not affect arthritis in IFN-γ knockout mice. Interleukin-27 also suppressed osteoclast differentiation in human and intriguingly, it could promote the expression of IFN-γ on priming osteoclasts. These results imply that IL-27 suppressed the generation of CIA and osteoclastogenesis, which were mediated by the induction of IFN-γ.


Asunto(s)
Interferón gamma/biosíntesis , Interleucina-17/farmacología , Osteoclastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Animales , Artritis Experimental/etiología , Diferenciación Celular/efectos de los fármacos , Humanos , Interferón gamma/inmunología , Interleucina-17/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Osteoclastos/fisiología
10.
Clin Immunol ; 144(3): 190-9, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22836084

RESUMEN

IL-12p40 homodimer is a natural antagonist of IL-12 and IL-23, which are potent pro-inflammatory cytokines required for Th1 and Th17 immune responses, respectively. It has been reported that Th17 response is involved in inflammatory bowel disease (IBD), a chronic disorder of the digestive system with steadily increasing incidence. Here, we investigated the effects of IL-12p40 delivered via recombinant adenovirus (rAd/IL-12p40) or mesenchymal stem cells (MSC/IL-12p40) in a dextran sulfate sodium salt (DSS)-induced colitis model. Injection of rAd/IL-12p40 or MSC/IL-12p40 efficiently attenuated colitis symptoms and tissue damage, leading to an increased survival rate. Moreover, IL-12p40 delivery suppressed IL-17A, but enhanced IFN-γ production from mesenteric lymph node cells, supporting the preferential suppression of IL-23 by IL-12p40 homodimer in vitro and the suppression of Th17 responses in vivo. Our results demonstrate that IL-12p40 delivery ameliorates DSS-induced colitis by suppressing IL-17A production and inflammation in the intestinal mucosa, providing an effective new therapeutic strategy for IBDs.


Asunto(s)
Colitis/inmunología , Inflamación/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Subunidad p40 de la Interleucina-12/inmunología , Interleucina-17/inmunología , Mucosa Intestinal/inmunología , Adenoviridae/inmunología , Animales , Colitis/inducido químicamente , Sulfato de Dextran , Modelos Animales de Enfermedad , Femenino , Interferón gamma/inmunología , Interleucina-12/inmunología , Interleucina-23/inmunología , Ganglios Linfáticos/inmunología , Células Madre Mesenquimatosas/inmunología , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley , Células Th17/inmunología
11.
Yonsei Med J ; 52(6): 1008-15, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22028167

RESUMEN

PURPOSE: Bacillus Calmette-Guérin (BCG) vaccine has widely been used to immunize against tuberculosis, but its protective efficacy is variable in adult pulmonary tuberculosis, while it is not efficiently protective against progressive infection of virulent Mycobacterium tuberculosis strains. In this study, the protective effects of plasmid DNA vaccine constructs encoding IL-12 or IL-18 with the BCG vaccine were evaluated against progressive infection of M. tuberculosis, using mouse aerosol challenge model. MATERIALS AND METHODS: Plasmid DNA vaccine constructs encoding IL-12 or IL-18 were constructed and mice were immunized with the BCG vaccine or with IL-12 DNA or IL-18 DNA vaccine constructs together with the BCG vaccine. RESULTS: The BCG vaccine induced high level of interferon gamma (IFN-γ) but co-immunization of IL-12 or IL-18 DNA vaccine constructs with the BCG vaccine induced significantly higher level of IFN-γ than a single BCG vaccine. The BCG vaccine was highly protective at early stage of M. tuberculosis infection, but its protective efficacy was reduced at later stage of infection. The co-immunization of IL-12 DNA vaccine constructs with the BCG vaccine was slightly more protective at early stage of infection and was significantly more protective at later stage infection than a single BCG vaccine. CONCLUSION: Co-immunization of IL-12 DNA vaccine with the BCG vaccine induced more protective immunity and was more effective for protection against progressive infection of M. tuberculosis.


Asunto(s)
Vacuna BCG/inmunología , Interleucina-12/genética , Interleucina-18/genética , Tuberculosis/inmunología , Vacunas de ADN/genética , Vacunas de ADN/inmunología , Animales , Femenino , Técnicas para Inmunoenzimas , Interferón gamma/sangre , Ratones , Ratones Endogámicos C57BL , Plásmidos/genética , Tuberculosis/sangre , Tuberculosis/prevención & control
12.
Cytokine ; 55(3): 420-8, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21700476

RESUMEN

4-1BB (CD137) is a powerful T-cell costimulatory molecule in the treatment of virus infections and tumors, but recent studies have also uncovered regulatory functions of 4-1BB signaling. Since 4-1BB triggering suppresses autoimmunity by accumulating indoleamine 2,3-dioxygenase (IDO) in dendritic cells (DCs) in an interferon (IFN)-γ-dependent manner, we asked whether similar molecular and cellular changes were induced by 4-1BB triggering in virus-infected mice. 4-1BB triggering increased IFN-γ and IDO, and suppressed CD4(+) T cells, in C57BL/6 mice infected with the type 1 KOS strain of Herpes simplex virus (HSV-1), as it does in an autoimmune disease model. Detailed analysis of the CD4(+) T suppression showed that freshly activated CD62L(high) T cells underwent apoptosis in the early phase of suppression, and CD62L(low) effector/memory T cells in the later phase. Although 4-1BB triggering resulted in similar cellular changes - increased CD8(+) T and decreased CD4(+) T cells, it had different effects on mortality in mice infected with HSV-1 RE, influenza, and Japanese encephalitis virus (JEV); it increased mortality in influenza-infected mice but decreased it in JEV-infected mice. Since the dominant type of immune cell generated to protect the host was different for each virus - CD4(+) T cells and neutrophils in HSV-1 RE infection, both CD4(+) T and CD8(+) T cells in influenza infection, and a crucial role for B cells in JEV infection, 4-1BB triggering resulted in different therapeutic outcomes. We conclude that the therapeutic outcome of 4-1BB triggering is determined by whether the protective immunity generated against the virus was beneficially altered by the 4-1BB triggering.


Asunto(s)
Ligando 4-1BB/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Animales , Apoptosis/inmunología , Autoinmunidad , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/virología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/virología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Progresión de la Enfermedad , Virus de la Encefalitis Japonesa (Especie)/patogenicidad , Indolamina-Pirrol 2,3,-Dioxigenasa/biosíntesis , Indolamina-Pirrol 2,3,-Dioxigenasa/inmunología , Virus de la Influenza A/patogenicidad , Interferón gamma/biosíntesis , Selectina L , Recuento de Linfocitos , Ratones , Ratones Endogámicos BALB C/virología , Ratones Endogámicos C57BL/virología , Ratones Noqueados , Transducción de Señal/inmunología , Simplexvirus/patogenicidad , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología
13.
J Immunother ; 34(3): 297-306, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21389868

RESUMEN

Programmed death 1 (PD-1) signaling through its ligands, PD-L1 and PD-L2, has been known to negatively regulate T-cell responses. In addition, PD-L1 has been shown to interact with B7-1 costimulatory molecule to inhibit T-cell responses. Extensive studies have shown that PD-1/PD-L blockade restores exhausted T cells during chronic viral infections and tumors. In this study, we evaluated the effects of soluble PD-1 (sPD-1) as a blockade of PD-1 and PD-L1 on vaccine-elicited antigen-specific T-cell responses in mice. Coadministration of sPD-1 DNA with human papilloma virus-16 E7 DNA vaccine significantly enhanced E7-specific CD8(+) T-cell responses, resulting in potent antitumor effects against E7-expressing tumors. We also found that sPD-1, codelivered with adenovirus-based vaccine, could increase antigen-specific CD8(+) T-cell responses, indicating vaccine type-independent adjuvant effect of sPD-1. In addition, the frequency and functional activity of adoptively transferred OT-I cells, particularly memory CD8(+) T cells, were augmented by coadministration of sPD-1 DNA, which was closely associated with increased T-cell proliferation and reduced T-cell apoptosis through upregulation of Bcl-xL expression during T-cell activation. Codelivery of sPD-1 DNA also enhanced maturation of dendritic cells (DCs) in vivo which was accompanied by upregulation of DC maturation markers such as major histocompatibility complex class II. Taken together, our findings show that sPD-1 potently enhances codelivered antigen-specific CD8(+) T-cell responses and in vivo maturation of DCs during activation of naive CD8(+) T cells, suggesting that an immunization strategy with sPD-1 as an adjuvant can be used to increase antigen-specific T-cell immunity elicited by vaccination.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Traslado Adoptivo , Antígenos CD/inmunología , Antígenos CD/farmacología , Proteínas Reguladoras de la Apoptosis/inmunología , Proteínas Reguladoras de la Apoptosis/farmacología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer , Vacunas de ADN , Adenoviridae/genética , Adenoviridae/inmunología , Adenoviridae/metabolismo , Animales , Antígenos CD/metabolismo , Antígenos CD/uso terapéutico , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/uso terapéutico , Antígeno B7-H1 , Linfocitos T CD8-positivos/metabolismo , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Línea Celular , Proliferación Celular/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/inmunología , Papillomavirus Humano 16/metabolismo , Humanos , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Papiloma/inmunología , Papiloma/metabolismo , Papiloma/terapia , Proteínas E7 de Papillomavirus/inmunología , Proteínas E7 de Papillomavirus/metabolismo , Receptor de Muerte Celular Programada 1 , Vacunas de ADN/genética , Vacunas de ADN/inmunología , Vacunas de ADN/metabolismo
14.
Immune Netw ; 10(6): 198-205, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21286380

RESUMEN

BACKGROUND: A crucial limitation of DNA vaccines is its weak immunogenicity, especially in terms of eliciting antibody responses in non-human primates or humans; therefore, it is essential to enhance immune responses to vaccination for the development of successful DNA vaccines for humans. METHODS: Here, we approached this issue by evaluating interleukin-7 (IL-7) as a genetic adjuvant in cynomolgus monkeys immunized with multigenic HCV DNA vaccine. RESULTS: Codelivery of human IL-7 (hIL-7)-encoding DNA appeared to increase DNA vaccine-induced antibody responses specific for HCV E2 protein, which plays a critical role in protecting from HCV infection. HCV-specific T cell responses were also significantly enhanced by codelivery of hIL-7 DNA. Interestingly, the augmentation of T cell responses by codelivery of hIL-7 DNA was shown to be due to the enhancement of both the breadth and magnitude of immune responses against dominant and subdominant epitopes. CONCLUSION: Taken together, these findings suggest that the hIL-7-expressing plasmid serves as a promising vaccine adjuvant capable of eliciting enhanced vaccine-induced antibody and broad T cell responses.

15.
Eur J Immunol ; 40(2): 351-8, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19950168

RESUMEN

IL-7 plays a crucial role in the homeostatic proliferation, differentiation and survival of T cells, as well as in the survival and proliferation of precursor B cells. Here, we demonstrated that utilizing nonlytic Fc-fused IL-7 (IL-7-Fc(m)) as a genetic adjuvant significantly enhanced not only CD4(+) but also CD8(+) T-cell responses by E7 DNA immunization, in addition to improving protection against TC-1-induced tumors in comparison to IL-7 alone. Similar results were obtained in OT-1 adoptive transfer experiments with OVA DNA injection, suggesting independence from antigenic nature and experimental conditions. In particular, the increased frequency of CD8(+) T cells was mainly due to enhanced T-cell proliferation in T-cell priming, and not to decreased cellular apoptosis. Interestingly, the enhanced adjuvant effect was not seen in the co-delivery of lytic Fc-fused IL-7 (IL-7-Fc) which increases T-cell apoptosis as well as T-cell proliferation, suggesting that the T-cell proliferative effect may be neutralized by T-cell apoptosis. Thus, our findings suggest that nonlytic Fc, in contrast to lytic Fc, fusion to cytokines may provide an insight in designing a potent genetic adjuvant for inducing CD4(+) and CD8(+) T-cell responses.


Asunto(s)
Antígenos/inmunología , Fragmentos Fc de Inmunoglobulinas/inmunología , Interleucina-7/inmunología , Linfocitos T/inmunología , Adyuvantes Inmunológicos , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Femenino , Citometría de Flujo , Humanos , Fragmentos Fc de Inmunoglobulinas/genética , Fragmentos Fc de Inmunoglobulinas/metabolismo , Inmunoterapia Adoptiva , Interleucina-7/genética , Interleucina-7/metabolismo , Ratones , Ratones Endogámicos C57BL , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología , Neoplasias Experimentales/terapia , Proteínas Oncogénicas Virales/genética , Proteínas Oncogénicas Virales/inmunología , Proteínas Oncogénicas Virales/metabolismo , Ovalbúmina/inmunología , Proteínas E7 de Papillomavirus , Linfocitos T/citología , Linfocitos T/metabolismo , Vacunación/métodos , Vacunas de ADN/inmunología
16.
Vaccine ; 27(42): 5906-12, 2009 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-19651174

RESUMEN

Since human papillomavirus (HPV) E6 and E7 are promising tumor antigens, we engineered E6 and E7 antigens to generate an optimal HPV DNA vaccine by codon optimization (Co), fusion of E6 and E7, addition of a tissue plasminogen activator (tpa) signal sequence, addition of CD40 ligand (CD40L) or Fms-like tyrosine kinase-3 ligand (Flt3L). The resulting constructs were investigated in terms of their antitumor activity as well as induction of HPV-specific CD8(+) T cell responses. When E6(Co) and E7(Co) were fused (E67(Co)), CD8(+) T cell responses specific for E6 or E7 antigen decreased, but the preventive antitumor effect rather improved, demonstrating the importance of broad immunity. Interestingly, Flt3L-fused HPV DNA vaccine exhibited stronger E6- and E7-specific CD8(+) T cell responses as well as therapeutic antitumor effect than that of CD40L linked HPV DNA vaccine. Finally, the optimal construct, tFE67(Co), was generated by including tpa signal sequence, Flt3L, fusion of E6 and E7, and codon optimization, which induces 23 and 25 times stronger E6- and E7-specific CD8(+) T cell responses than those of initial E67 fusion construct. In particular, inclusion of electroporation in intramuscular immunization of tFE67(Co) further enhances HPV-specific CD8(+) T cell responses, leading to complete tumor regression in a therapeutic setting. Thus, our results provide valuable insight on effective HPV DNA vaccine design and suggest that tFE67(Co) delivered with electroporation may be a promising therapeutic HPV DNA vaccine against cervical cancer.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Infecciones por Papillomavirus/prevención & control , Vacunas contra Papillomavirus/inmunología , Neoplasias del Cuello Uterino/prevención & control , Animales , Ligando de CD40/inmunología , Vacunas contra el Cáncer/genética , Línea Celular , Electroporación , Femenino , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/inmunología , Inmunidad Celular , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Proteínas Oncogénicas Virales/genética , Proteínas Oncogénicas Virales/inmunología , Proteínas E7 de Papillomavirus , Infecciones por Papillomavirus/inmunología , Vacunas contra Papillomavirus/genética , Ingeniería de Proteínas , Proteínas Represoras/genética , Proteínas Represoras/inmunología , Neoplasias del Cuello Uterino/inmunología , Neoplasias del Cuello Uterino/virología , Vacunas de ADN/genética , Vacunas de ADN/inmunología
17.
Immune Netw ; 9(1): 1-3, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20107530
18.
Cancer Res ; 68(23): 9614-23, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19047138

RESUMEN

Adenovirus-mediated gene therapies against brain tumors have been limited by the difficulty in tracking glioma cells infiltrating the brain parenchyma. Human umbilical cord blood-derived mesenchymal stem cells (UCB-MSC) are particularly attractive cells for clinical use in cell-based therapies. In the present study, we evaluated the tumor targeting properties and antitumor effects of UCB-MSCs as gene delivery vehicles for glioma therapy. We efficiently engineered UCB-MSCs to deliver a secretable trimeric form of tumor necrosis factor-related apoptosis-inducing ligand (stTRAIL) via adenoviral transduction mediated by cell-permeable peptides. We then confirmed the migratory capacity of engineered UCB-MSCs toward tumor cells by an in vitro migration assay and by in vivo injection of UCB-MSCs into the tumor mass or the opposite hemisphere of established human glioma in nude mice. Moreover, in vitro coculture, experiments on Transwell plates, and in vivo survival experiments showed that MSC-based stTRAIL gene delivery has more therapeutic efficacy compared with direct injection of adenovirus encoding the stTRAIL gene into a tumor mass. In vivo efficacy experiments showed that intratumoral injection of engineered UCB-MSCs (MSCs-stTRAIL) significantly inhibited tumor growth and prolonged the survival of glioma-bearing mice compared with controls. These results suggest that human UCB-MSCs have potential use as effective delivery vehicles for therapeutic genes in the treatment of intracranial glioma.


Asunto(s)
Neoplasias Encefálicas/terapia , Sangre Fetal/citología , Terapia Genética/métodos , Glioma/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/fisiología , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Adenoviridae/genética , Animales , Apoptosis/fisiología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/fisiología , Glioma/genética , Glioma/metabolismo , Glioma/patología , Humanos , Masculino , Ratones , Ratones Desnudos , Ligando Inductor de Apoptosis Relacionado con TNF/administración & dosificación , Ligando Inductor de Apoptosis Relacionado con TNF/biosíntesis , Transducción Genética , Ensayos Antitumor por Modelo de Xenoinjerto
19.
J Immunol ; 181(2): 1507-18, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18606706

RESUMEN

IL-23, a clinically novel cytokine, targets CD4(+) T cells. Recent IL-1Ra(-/-) mouse studies have demonstrated that IL-23 indirectly stimulates the differentiation of osteoclast precursors by enhancing IL-17 release from CD4(+) T cells. IL-17, in turn, stimulates osteoclastogenesis in osteoclast precursor cells. In this study, we found that IL-23 up-regulates receptor activator of NF-kappaB ligand expression by CD4(+) T cells, and thus contributes to osteoclastogenesis. This indirect pathway is mediated by NF-kappaB and STAT3. We have also demonstrated that IL-23 can influence osteoclastogenesis positively under the special conditions in the IL-1-dominant milieu of IL-1Ra(-/-) mice. We propose that IL-23-enhanced osteoclastogenesis is mediated mainly by CD4(+) T cells. The results of this study show that IL-23 is a promising therapeutic target for the treatment of arthritis-associated bone destruction.


Asunto(s)
Artritis Experimental/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Interleucina-23/metabolismo , Osteoclastos/metabolismo , Ligando RANK/metabolismo , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Animales , Artritis Experimental/inmunología , Artritis Experimental/fisiopatología , Células Cultivadas , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Interleucina-23/inmunología , Articulaciones/inmunología , Articulaciones/metabolismo , Ratones , Ratones Noqueados , FN-kappa B/inmunología , FN-kappa B/metabolismo , Osteoclastos/citología , Osteoclastos/inmunología , Ligando RANK/inmunología , Receptor Activador del Factor Nuclear kappa-B/inmunología , Factor de Transcripción STAT3/inmunología , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Regulación hacia Arriba
20.
J Immunol ; 180(11): 7167-74, 2008 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-18490715

RESUMEN

IL-12p40 is a natural antagonist which inhibits IL-12- and IL-23-mediated biological activity by blocking the binding of IL-12/23 to their receptors. Recently, IL-12p40 was also shown to have immune-enhancing activity through the activation of macrophages or dendritic cells. In this study, we investigated the effects of IL-12p40 as a genetic adjuvant on immune modulation using recombinant adenoviruses expressing IL-12p40 (rAd/IL-12p40) and OVA (rAd/OVA). Coimmunization of rAd/IL-12p40 at a low dose (1 x 10(4) PFU) with rAd/OVA resulted in OVA-specific immune enhancement, while a high dose of rAd/IL-12p40 (1 x 10(8) PFU) caused significant suppression of CD8(+) T cell responses. In addition, the enhancement and suppression of OVA-specific CD8(+) T cell responses correlated with antitumor activity against E.G7-OVA tumor challenge, which subsequently affected the survival rate. Moreover, the differential CD8(+) T cell response by IL-12p40 was still observed in IL-12Rbeta2 knockout (IL-12Rbeta2KO), but not in IL-12Rbeta1 knockout (IL-12Rbeta1KO) mice, indicating that IL-12p40 is a cytokine which can modulate Ag-specific T cell responses depending on IL-12Rbeta1. Our findings provide a novel insight on the physiological role of IL-12p40, which can be informative in the design of vaccine strategies and therapeutic regimens.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Subunidad p40 de la Interleucina-12/inmunología , Animales , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Relación Dosis-Respuesta Inmunológica , Femenino , Interferón gamma/sangre , Interferón gamma/inmunología , Interleucina-12/inmunología , Interleucina-12/metabolismo , Subunidad p40 de la Interleucina-12/genética , Subunidad p40 de la Interleucina-12/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Neoplasias Experimentales/inmunología , Óxido Nítrico/sangre , Ovalbúmina/inmunología , Receptores de Interleucina-12/genética , Receptores de Interleucina-12/metabolismo , Proteínas Recombinantes/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...