Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros











Intervalo de año de publicación
1.
Int J Mol Sci ; 23(15)2022 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-35955542

RESUMEN

Systemic lupus erythematosus is a complex autoimmune disorder mostly mediated by B-cells in which costimulatory signals are involved. This immune dysregulation can cause tissue damage and inflammation of the kidney, resulting in lupus nephritis and chronic renal failure. Given the previous experience reported with CTLA4-Ig as well as recent understanding of the PD-1 pathway in this setting, our group was encouraged to evaluate, in the NZBWF1 model, a human fusion recombinant protein (Hybri) with two domains: CTLA4, blocking the CD28-CD80 costimulatory pathway, and PD-L2, exacerbating the PD-1-PD-L2 coinhibitory pathway. After achieving good results in this model, we decided to validate the therapeutic effect of Hybri in the more severe MRL/lpr model of lupus nephritis. The intraperitoneal administration of Hybri prevented the progression of proteinuria and anti-dsDNA antibodies to levels like those of cyclophosphamide and reduced the histological score, infiltration of B-cells, T-cells, and macrophages and immune deposition in both lupus-prone models. Additionally, Hybri treatment produced changes in both inflammatory-related circulating cytokines and kidney gene expression. To summarize, both in vivo studies revealed that the Hybri effect on costimulatory-coinhibitory pathways may effectively mitigate lupus nephritis, with potential for use as a maintenance therapy.


Asunto(s)
Lupus Eritematoso Sistémico , Nefritis Lúpica , Animales , Anticuerpos Antinucleares , Modelos Animales de Enfermedad , Humanos , Inmunomodulación , Riñón/metabolismo , Ratones , Ratones Endogámicos MRL lpr , Receptor de Muerte Celular Programada 1/metabolismo , Proteínas Recombinantes/metabolismo
2.
Biomolecules ; 12(7)2022 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-35883511

RESUMEN

A key aspect of the inflammatory phenomenon is the involvement of costimulatory molecules expressed by antigen-presenting cells (APCs) and their ability to secrete cytokines to set instructions for an adaptive immune response and to generate tolerance or inflammation. In a novel integrative approach, we aimed to evaluate the kinetic expression of the membrane and soluble B7 costimulatory molecules CD86, ICOS-L, PDL1, PDL2, the transcription factor Interferon Regulatory Factor 4 (IRF4), and the cytokines produced by monocyte-derived dendritic cells (Mo-DCs) after challenging them with different concentrations of stimulation with E. coli lipopolysaccharide (LPS) for different lengths of time. Our results showed that the stimuli concentration and time of exposure to an antigen are key factors in modulating the dynamic expression pattern of membrane and soluble B7 molecules and cytokines.


Asunto(s)
Antígeno B7-1 , Lipopolisacáridos , Antígenos B7/metabolismo , Antígeno B7-1/metabolismo , Citocinas/metabolismo , Células Dendríticas , Escherichia coli/metabolismo , Humanos , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Lipopolisacáridos/metabolismo , Lipopolisacáridos/farmacología
3.
Am J Cancer Res ; 12(4): 1621-1634, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35530287

RESUMEN

Subcutaneous implantation of a human cancer cell line in immune-deficient mice (CDX) is a commonly used tool in preclinical studies for the assessment of potential anti-cancer drugs. As immunotherapy is transforming cancer treatment, tumor models in immunocompetent mice are necessary for us to understand the immune aspects of tumor biology. However, the systemic immune response to the implantation of cancer cells at proteome level is unclear. In this study, we characterized the dynamic proteomic changes of subcutaneous tumors and 5 immune organs (draining lymph node, mesenteric lymph node, spleen, thymus and marrow) at six time points after implantation using a Hepa1-6 derived allograft mouse model. Our data suggest that interaction of the implanted tumor cells with mouse immune system followed the trajectory of "tumor rejection" to "immune evasion" in that the tumor gained the ability to evade the immune system for growth. Furthermore, anti-PDL2 antibody was validated here as an optional immunotherapy strategy to inhibit the growth of Hepa1-6 subcutaneous tumors. These findings from our study provided valuable information for the understanding of tumor and immune interaction and shed light on the rational design for clinical cancer treatment and other preclinical experiments.

4.
Cancers (Basel) ; 13(4)2021 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-33567641

RESUMEN

BACKGROUND: Hodgkin lymphoma (HL) is predominantly composed of reactive, non-neoplastic cells surrounding scarcely distributed tumor cells, that is, so-called Hodgkin and Reed-Sternberg (HRS) or lymphocyte predominant (LP) cells. This scarcity impeded the analysis of the tumor cell genomes for a long time, but recently developed methods (especially laser capture microdissection, flow cytometry/fluorescence-activated cell sorting) facilitated molecular investigation, elucidating the pathophysiological principles of "Hodgkin lymphomagenesis". METHODS: We reviewed the relevant literature of the last three decades focusing on the genomic landscape of classic and nodular lymphocyte predominant HL (NLPHL) and summarized molecular cornerstones. RESULTS: Firstly, the malignant cells of HL evade the immune system by altered expression of PDL1/2, B2M and MHC class I and II due to various genetic alterations. Secondly, tumor growth is promoted by permanently activated JAK/STAT signaling due to pervasive mutations of multiple genes involved in the pathway. Thirdly, apoptosis of neoplastic cells is prevented by alterations of NF-κB compounds and the PI3K/AKT/mTOR axis. Additionally, Epstein-Barr virus infection can simultaneously activate JAK/STAT and NF-κB, similarly leading to enhanced survival and evasion of apoptosis. Finally, epigenetic phenomena such as promoter hypermethylation lead to the downregulation of B-lineage-specific, tumor-suppressor and immune regulation genes. CONCLUSION: The blueprint of HL genomics has been laid, paving the way for future investigations into its complex pathophysiology.

5.
Cell Oncol (Dordr) ; 44(2): 423-432, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33469839

RESUMEN

BACKGROUND: Immune responses have long been an area of interest in cancer research. In this study, the effects of programmed cell death-1 (PD-1) and its ligand (PD-L2) on the prognosis of colorectal cancer (CRC) were investigated. METHODS: Primary tumour specimens of stage III CRC patients operated between 2002 and 2013 were assessed for PD-1 and PD-L2 expression and various clinicopathological and prognostic factors. RESULTS: We observed a significant relationship between poor prognostic factors and PD-1/PD-L2 expression. These biomarkers were also found to serve as independent risk factors for LIR and MSI. In univariate analysis, relapse-free survival (RFS) and overall survival (OS) rates were found to be poor in PD-1 and PD-L2 positive patients. In multivariate analysis, these biomarkers were found to serve as independent poor prognostic factors for RFS and OS. CONCLUSIONS: Our data indicate that PD-1 and PD-L2 may serve as independent prognostic survival parameters for CRC patients and may be employed for the design of targeted therapies.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Recurrencia Local de Neoplasia/metabolismo , Recurrencia Local de Neoplasia/patología , Proteína 2 Ligando de Muerte Celular Programada 1/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Análisis de Regresión , Reproducibilidad de los Resultados , Factores de Riesgo
6.
FEBS Open Bio ; 10(6): 1162-1170, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32324337

RESUMEN

CD279 is a cell surface protein predominantly expressed on T cells. Its ligands CD273 and CD274 are expressed on antigen-presenting cells and tumors. CD279 has been shown to act as an important immune check point by inhibiting CD8 T cell activation, and antibodies against CD279 enhance T cell-mediated cytotoxic function. However, whether CD279 has other functions in CD4 T cell homeostasis or in mediating T cell interactions with antigen-presenting cells remains unclear. In the present study, we show that antibody-mediated inhibition of CD279 reduces T cell survival in bone marrow in vivo. Unexpectedly, CD279 blockade also compromised regulatory T cell and macrophage interactions by reducing their contact time. The observation that the CD273 antagonist had little effect suggests that CD274 (the second ligand of CD279) plays a more central role in contact between conventional T cells (Tcon) and macrophages. The results of the present study suggest that both CD279 ligands contribute to the interaction length between T cells and macrophages as a mechanism of maintaining Treg homeostasis. Furthermore, CD273 and CD274 are not redundant ligands because CD274 may have unique effects on Tcon in this complex immune axis. Therefore, ligand selection for check point blockade as a tool for cancer immunotherapy has important implications with respect to anti-tumor T cell activation and the avoidance of side effects.


Asunto(s)
Comunicación Celular/inmunología , Inhibidores de Puntos de Control Inmunológico/farmacología , Macrófagos/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo , Linfocitos T Reguladores/metabolismo , Traslado Adoptivo , Animales , Antígeno B7-H1/metabolismo , Comunicación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/inmunología , Células Cultivadas , Humanos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Macrófagos/inmunología , Ratones , Ratones Noqueados , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Cultivo Primario de Células , Proteína 2 Ligando de Muerte Celular Programada 1/antagonistas & inhibidores , Proteína 2 Ligando de Muerte Celular Programada 1/metabolismo , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología
7.
Cell Rep ; 28(13): 3353-3366.e5, 2019 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-31553906

RESUMEN

Smad7, a negative regulator of TGF-ß signaling, has been implicated in the pathogenesis and treatment of inflammatory bowel diseases (IBDs), including Crohn's disease (CD) and ulcerative colitis (UC). Here, we found that Smad7 mediates intestinal inflammation by limiting the PDL2/1-PD1 axis in dendritic cells (DCs) and CD4+T cells. Smad7 deficiency in DCs promotes TGF-ß responsiveness and the co-inhibitory molecules PDL2/1 on DCs, and it further imprints T cell-PD1 signaling to promote Treg differentiation. DC-specific Smad7 deletion mitigates DSS-induced colitis by inducing CD103+PDL2/1+DCs and Tregs. In addition, Smad7 deficiency in CD4+T cells promotes PD1 and PD1-induced Tregs in vitro. The transfer of Smad7-deficient CD4+T cells enhances Tregs in vivo and protects against T cell-mediated colitis. Furthermore, Smad7 antisense ameliorates DSS-induced UC, increasing TGF-ß and PDL2/1-PD1 signaling. Enhancing PD1 signaling directly via Fc-fused PDL2/1 is also beneficial. Our results identify how Smad7 mediates intestinal inflammation and leverages these pathways therapeutically, providing additional strategies for IBD intervention.


Asunto(s)
Autoinmunidad/genética , Inflamación/genética , Intestinos/patología , Proteína smad7/genética , Humanos , Transducción de Señal
8.
Metab Brain Dis ; 34(6): 1705-1712, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31410774

RESUMEN

Inflammation involving migration of immune cells across the damaged blood-brain barrier (BBB), activation of resident innate microglia and production of inflammatory humoral mediators such as cytokines and chemokines play a critical role in the pathogenesis of ischemic stroke. Cell-cell signaling involved in the process also includes checkpoint interaction between programmed death receptor (PD1) and programmed death ligands, PDL1 and PDL2. Based on our previous studies showing reduced MCAO infarct volumes in PDL2 deficient mice, we evaluated the ability of anti-PDL2 mAb to treat MCAO in male and female C57BL/6 mice. We found that anti-PDL2 neutralizing antibody treatment of MCAO significantly reduced infarct volumes in male mice but had no protective effects in female mice even at a 5-fold increased dose of anti-PDL2 mAb. The protection in male mice was likely mediated by reduced percentages in the spleen of PDL2+CD19+ B cells, PDL1+CD4+ T cells and CD86+CD11b+ macrophages in concert with reduced expression of PDL1 and TNFα and continued expression of CD206, in the injured ipsilateral brain hemisphere. The lack of a therapeutic benefit of anti-PDL2 on stroke-induced infarct volumes in female mice was reflected by no detectable reduction in expressed PDL2 or PDL1 and an increased frequency of Th1 and Th17 pro-inflammatory T cell subsets in the spleen, an effect not seen in PDL2 mAb treated males. This result potentially limits the utility of anti-PDL2 mAb therapy in stroke to males but underscores the importance of meeting the STAIR requirements for development of new stroke therapies for both sexes.


Asunto(s)
Anticuerpos Neutralizantes/uso terapéutico , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Proteína 2 Ligando de Muerte Celular Programada 1/inmunología , Accidente Cerebrovascular/tratamiento farmacológico , Subgrupos de Linfocitos T/inmunología , Animales , Modelos Animales de Enfermedad , Femenino , Infarto de la Arteria Cerebral Media/inmunología , Masculino , Ratones , Factores Sexuales , Accidente Cerebrovascular/inmunología , Resultado del Tratamiento
9.
Oral Oncol ; 89: 34-39, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30732956

RESUMEN

OBJECTIVES: Circulating tumor cells (CTCs) are cells that have shed from tumor tissue into the bloodstream, and the detection and characterization of CTCs in head and neck squamous cell carcinoma (HNSCC) still remain a challenge. MATERIALS AND METHODS: CTCs were isolated from 30 patients with HNSCC with recurrent and/or distant metastasis, via the depletion of CD45-positive cells with magnetic beads and the expression of multiple epithelial markers (CK19, EpCAM, EGFR, and c-Met) was analyzed by RT-qPCR with a low concentration of RNA from the CTC population. We next investigated the expression of the immune-regulatory molecules, PD-L1, PD-L2, and CD47, in CTC-positive patients and the PD-L1 expression in CTCs was compared with that in tumor tissues. RESULTS: Twenty-four (80.0%) of the 30 patients were positive for at least one epithelial-related gene. Among the 24 CTC-positive patients, 19 (79.2%), 20 (83.3%), and 17 (70.8%) patients were positive for CD47, PD-L1, and PD-L2, respectively. Interestingly, the expression of these three immune-regulatory molecules was positively correlated to each other. As expected, PD-L1 expression in the tumor tissue did not correspond completely with that in the CTCs. CONCLUSION: Although clinical application and/or characterization of CTCs are still developing, our findings suggest that the CTCs are rapidly becoming a powerful tool in cancer treatments that involve the use of immune checkpoint inhibitors.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Células Neoplásicas Circulantes/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/inmunología , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Masculino , Persona de Mediana Edad
10.
Virchows Arch ; 474(4): 497-509, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30132131

RESUMEN

Chromosome 9 harbors several relevant oncogenes related to hematolymphoid malignancies and one specific region, 9p24, has come into the focus of attention in the last years as it contains recurrently mutant genes of therapeutic interest. The most prominent genes of this locus are programmed death ligands 1 and 2 (PDL1/PDL2), with the amplification of PDL1 being a hallmark of both classical Hodgkin and primary mediastinal B cell lymphoma, and Janus kinase 2 (JAK2), which is point-mutated in myeloproliferative neoplasms and other myeloid malignancies, and rearranged in PCM1-JAK2-positive myeloid/lymphoid neoplasms with eosinophila. Finally, this locus contains the lysine (K)-specific demethylase 4C (KDM4C/JMJD2C), which is also relevant for oncogenesis. Activation of these genes is effectuated, as exemplified, by multiple mechanisms, which is rather unique to oncogenes, since they are usually affected by just one type of mutation, and points towards the central role of these genes in tumor initiation and growth. Amplifications and, less frequently, translocations are the most common findings for PDL1/PDL2 and JAK2 in lymphomas. In this review, we describe the role of genes located on chromosome 9p24 and their derived proteins in diverse subtypes of lymphomas, with a special focus on PDL1 and PDL2, which are becoming a central target of immunotherapy, not only in classical Hodgkin lymphoma but also in various types of solid cancers. We also elucidate the role of the surgical pathologists in this setting - concerning what they can contribute - both diagnostically and predictively.


Asunto(s)
Cromosomas Humanos Par 9/genética , Inmunoterapia , Linfoma/genética , Linfoma/terapia , Antígeno B7-H1/genética , Humanos , Janus Quinasa 2/genética , Proteínas de la Membrana/genética , Mutación , Proteínas Supresoras de Tumor/genética
11.
Oncoimmunology ; 7(3): e1356144, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29399387

RESUMEN

Although immune checkpoint blockade have demonstrated promising results, their effects on gastric cancer (GC) are under investigation. Understanding the clinical significance of PD1 and its ligands' expression, together with T cell infiltration might provide clues for biomarkers screening in GC immunotherapy. Immunohistochemistry were performed on a tissue microarray including 1,014 GC specimens using PD1, PDL1 and PDL2 antibodies. T cell markers CD3 and CD8 were also stained and quantified by automated image analysis. Correlation with clinical features and outcome were analyzed after controlling for potential confounders including EBV infection, HER2, C-met and PCNA expression. 37.8% of the cases showed membranous PD-L1 expression in tumor cells and 74.9% in infiltrating immune cells. PDL1 expression rate was rather higher in patients without metastasis, in EBV positive group and those with C-met and PCNA expression. GC patients with high level PDL1 expression exhibited better survival. GC Patients with higher T cell infiltration also showed elevated PDL1, PDL2 and PD1 expression and predict favorable outcome, indicating an adaptive immune resistance mechanism may exist. The group of patients infiltrated with lower density CD3+ T cells also without PDL1 expression in tumor cells predict the worst outcome in the subgroup of different PTNM stage, which may suggest an inactive immune status. These results highlights the need to assess both PDL1 expression in all tumor context and the characterization of the GC immune microenvironment.

12.
Artículo en Chino | WPRIM (Pacífico Occidental) | ID: wpr-611566

RESUMEN

Objective To investigate the real-time regulatory effects of IFN-γ, programed death ligand 2(PDL2) and janus kinase/signal transducers and activators of transcription (JAK/STAT) signaling pathway on the adherence, proliferation and migration of human placenta-derived mesenchymal stem cells(hPMSCs) based on a finding that IFN-γ could enhance the expression of PDL2 in hPMSCs through JAK/STAT signaling pathway.Methods hPMSCs were isolated by using enzyme digestion method and then co-cultured with IFN-γ, anti-PDL2 monoclonal antibody (anti-PDL2 McAb) and JAK inhibitor, respectively.Real-time cell analysis (RTCA) was used to detect the dynamic changes in the adherence, proliferation and migration of hPMSCs following various interventions.Results IFN-γ remarkably suppressed hPMSCs proliferation during the period from 40 hours to 80 hours after intervention and also inhibited the non-targeted migration of hPMSCs.However, hPMSCs adherence was not affected by IFN-γ.Co-culturing hPMSCs with anti-PDL2 McAb significantly enhanced hPMSCs adhesion and inhibited their non-targeted migration, but had no significant effect on hPMSCs proliferation.Furthermore, the proliferation of hPMSCs co-cultured with IFN-γ and anti-PDL2 McAb was significantly inhibited as compared with that of anti-PDL2McAb treatment group.The adhesion, migration and proliferation of hPMSCs were significantly inhibited after co-culturing them with JAK inhibitor.Conclusion IFN-γ can remarkably suppress the proliferation and migration of hPMSCs.PDL2 can enhance the migration and inhibit the adhesion of hPMSCs.JAK/STAT signaling pathway is involved in regulating the adhesion, migration and proliferation of hPMSCs.

13.
Hum Vaccin Immunother ; 12(11): 2777-2789, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27398650

RESUMEN

The programmed cell death protein 1 (PD1) is one of the checkpoints that regulates the immune response. Ligation of PD1 with its ligands PDL1 and PDL2 results in transduction of negative signals to T-cells. PD1 expression is an important mechanism contributing to the exhausted effector T-cell phenotype. The expression of PD1 on effector T-cells and PDL1 on neoplastic cells enables tumor cells to evade anti-tumor immunity. Blockade of PD1 is an important immunotherapeutic strategy for cancers. Pembrolizumab (Keytruda) is a humanized monoclonal anti-PD1 antibody that has been extensively investigated in numerous malignancies. In melanoma refractory to targeted therapy, pembrolizumab induced overall response rates (ORRs) of 21-34%. It was superior to another immune checkpoint inhibitor ipilimumab (Yervoy) in stage III/IV unresectable melanoma. In refractory non-small cell lung cancer (NSCLC), pembrolizumab induced ORRs of 19-25%. Based on these results, pembrolizumab was approved by the USA FDA for the treatment of advanced melanoma and NSCLC. Tumor cell PDL1 expression may be a valid response predictor. Molecular analysis also showed that tumors with high gene mutation burdens, which might result in the formation of more tumor-related neo-antigens, had better responses to pembrolizumab. In malignancies including lymphomas and other solid tumors, preliminary data showed that ORRs of around 20-50 % could be achieved. Adverse events occurred in up to 60% of patients, but grade 3/4 toxicities were observed in <10% of cases. Immune-related adverse events including thyroid dysfunction, hepatitis and pneumonitis are more serious and may lead to cessation of treatment.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Anticuerpos Monoclonales Humanizados/efectos adversos , Antineoplásicos/efectos adversos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/epidemiología , Humanos , Resultado del Tratamiento , Estados Unidos
14.
Immune Netw ; 16(1): 1-12, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26937227

RESUMEN

Th2 cell immunity is required for host defense against helminths, but it is detrimental in allergic diseases in humans. Unlike Th1 cell and Th17 cell subsets, the mechanism by which dendritic cells modulate Th2 cell responses has been obscure, in part because of the inability of dendritic cells to provide IL-4, which is indispensable for Th2 cell lineage commitment. In this regard, immune cells other than dendritic cells, such as basophils and innate lymphoid cells, have been suggested as Th2 cell inducers. More recently, multiple independent researchers have shown that specialized subsets of dendritic cells mediate Th2 cell responses. This review will discuss the current understanding related to the regulation of Th2 cell responses by dendritic cells and other immune cells.

15.
Immune Network ; : 1-12, 2016.
Artículo en Inglés | WPRIM (Pacífico Occidental) | ID: wpr-211463

RESUMEN

Th2 cell immunity is required for host defense against helminths, but it is detrimental in allergic diseases in humans. Unlike Th1 cell and Th17 cell subsets, the mechanism by which dendritic cells modulate Th2 cell responses has been obscure, in part because of the inability of dendritic cells to provide IL-4, which is indispensable for Th2 cell lineage commitment. In this regard, immune cells other than dendritic cells, such as basophils and innate lymphoid cells, have been suggested as Th2 cell inducers. More recently, multiple independent researchers have shown that specialized subsets of dendritic cells mediate Th2 cell responses. This review will discuss the current understanding related to the regulation of Th2 cell responses by dendritic cells and other immune cells.


Asunto(s)
Humanos , Basófilos , Células Dendríticas , Helmintos , Interleucina-4 , Linfocitos , Células TH1 , Células Th17 , Células Th2
16.
Immunotherapy ; 7(11): 1145-58, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26599163

RESUMEN

AIM: We provided potent dendritic cells (DCs) for induction of stronger antitumor immune responses. MATERIALS & METHODS: Using siRNA and shRNA systems, PDL-1 and PDL-2 were knocked down and then DC in vitro and in vivo properties were evaluated. RESULTS: Mild suppression of PDL-1/PDL-2 molecules was accompanied by appropriate expression of DCs co-stimulatory molecules and release of proinflammatory cytokines. In vitro T-cell engagement induced the proliferation and secretion of Th1 cytokines. Injection of DCs to a 4T1 mice model induced intratumor CD8(+) infiltrating lymphocytes, splenocytes expansion, Th1 cytokine profile shift, and a mild drift to tumor growth inhibition and mice survival. CONCLUSION: Manipulated DCs induced significant antitumor immunity, but this subject needs further evaluation in different animals.


Asunto(s)
Antígeno B7-H1/metabolismo , Linfocitos T CD8-positivos/inmunología , Carcinoma/terapia , Células Dendríticas/fisiología , Inmunoterapia Adoptiva/métodos , Proteína 2 Ligando de Muerte Celular Programada 1/metabolismo , Células TH1/inmunología , Animales , Antígeno B7-H1/genética , Línea Celular Tumoral , Movimiento Celular , Células Dendríticas/trasplante , Femenino , Terapia Genética , Células HEK293 , Humanos , Activación de Linfocitos/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neoplasias Experimentales , Proteína 2 Ligando de Muerte Celular Programada 1/genética , ARN Interferente Pequeño/genética , Carga Tumoral , Escape del Tumor/genética
17.
Cytotherapy ; 17(11): 1560-71, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26432559

RESUMEN

BACKGROUND AIMS: Mesenchymal stromal cells (MSCs) and regulatory T cells (Treg) have been successfully used in treating autoimmune diseases accompanied by abundant inflammatory cytokines such as interferon (IFN)-γ and tumor necrosis factor (TNF)-α. Therefore, this work investigated the effects of IFN-γ and TNF-α on the ability of human placenta-derived mesenchymal stromal cells (hPMSCs) on inducing the differentiation of CD4(+)interleukin (IL)-10(+)and CD8(+)IL-10(+)Treg subsets. METHODS: Human PMSCs were co-cultured with T cells in the presence or absence of a trans-well system or anti- programmed death ligand-2 (PDL2) monoclonal antibody (mAb), respectively. CD4(+)IL-10(+)and CD8(+)IL-10(+)Treg subsets, as well as the levels of IL-10 in the supernatants, were detected on this basis. Examinations were conducted to explore the impact of IFN-γ and TNF-α on the expression of PDL2 in hPMSCs. In this process, flow cytometry, Western blot and reverse-transcriptase-polymerase chain reaction were used. RESULTS: CD4(+)IL-10(+)and CD8(+)IL-10(+)Treg subsets from T cells either non-activated or activated by use of phytohaemagglutinin (PHA) or CD3/CD28mAb significantly increased in the presence of hPMSCs. However, these levels markedly decreased after blocking the expression of PDL2 in hPMSCs. IL-10 followed the same pattern. Furthermore, the percentages of CD4(+)IL-10(+) and CD8(+)IL-10(+)T cells also sharply declined under the trans-well system, whereas the percentages as well as the expression of PDL2 in hPMSCs oppositely raised after hPMSCs pre-stimulated by IFN-γ and TNF-α. IFN-γ could promote the expression of PDL2 partly through the JAK/STAT signaling pathway. CONCLUSIONS: IFN-γ and TNF-α could promote the ability of hPMSCs in inducing the differentiation of CD4(+)IL-10(+)and CD8(+)IL-10(+)Treg subsets and enhance the expression of PDL2 in hPMSCs. These would benefit the application of hPMSCs in clinical trials.


Asunto(s)
Interferón gamma/metabolismo , Interleucina-10/metabolismo , Células Madre Mesenquimatosas/fisiología , Placenta/citología , Proteína 2 Ligando de Muerte Celular Programada 1/metabolismo , Linfocitos T Reguladores/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/metabolismo , Antígenos CD8/metabolismo , Diferenciación Celular/efectos de los fármacos , Técnicas de Cocultivo , Femenino , Humanos , Interferón gamma/inmunología , Interferón gamma/farmacología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Embarazo , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Factor de Necrosis Tumoral alfa/farmacología
18.
Cancer Biol Ther ; 16(5): 662-70, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25806780

RESUMEN

We report 3 cases of durable complete response (CR) in patients with BRAF-mutated metastatic melanoma who were initially treated unsuccessfully with sequential immunotherapies (high dose interleukin 2 followed by ipilimumab with or without concurrent radiation therapy). After progression during or post immunotherapy, these patients were given BRAF inhibitor therapy and developed rapid CRs. Based on the concomitant presence of autoimmune manifestations (including vitiligo and hypophysitis), we postulated that there was a synergistic effect between the prior immune therapy and the BRAF targeting agents. Accordingly, the inhibitors were gradually weaned off beginning at 3 months and were stopped completely at 9-12 months. The three patients remain well and in CR off of all therapy at up to 15 months radiographic follow-up. The institution of the BRAF therapy was associated with development of severe rheumatoid-like arthritis in 2 patients which persisted for months after discontinuation of therapy, suggesting it was not merely a known toxicity of BRAF inhibitors (arthralgias). On immunologic analysis, these patients had high levels of non-T-regulatory, CD4 positive effector phenotype T-cells, which persisted after completion of therapy. Of note, we had previously reported a similar phenomenon in patients with metastatic melanoma who failed high dose interleukin-2 and were then placed on a finite course of temozolomide with rapid complete responses that have remained durable for many years after discontinuation of temozolomide. We postulate that a finite course of cytotoxic or targeted therapy specific for melanoma given after apparent failure of prior immunotherapy can result in complete and durable remissions that may persist long after the specific cytotoxic or targeted agents have been discontinued suggesting the existence of sequence specific synergism between immunotherapy and these agents. Here, we discuss these cases in the context of the literature on synergy between conventional or targeted cytotoxic therapy and immunotherapy in cancer treatment.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Inmunoterapia/métodos , Interleucina-2/metabolismo , Melanoma/tratamiento farmacológico , Melanoma/inmunología , Proteínas Proto-Oncogénicas B-raf/genética , Anticuerpos Monoclonales/administración & dosificación , Femenino , Humanos , Ipilimumab , Masculino , Melanoma/patología , Persona de Mediana Edad , Metástasis de la Neoplasia , Proteínas Proto-Oncogénicas B-raf/metabolismo
19.
Front Immunol ; 5: 74, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24605114

RESUMEN

For the past 150 years cancer immunotherapy has been largely a theoretical hope that recently has begun to show potential as a highly impactful treatment for various cancers. In particular, the identification and targeting of immune checkpoints have given rise to exciting data suggesting that this strategy has the potential to activate sustained antitumor immunity. It is likely that this approach, like other anti-cancer strategies before it, will benefit from co-administration with an additional therapeutic and that it is this combination therapy that may generate the greatest clinical outcome for the patient. In this regard, oncolytic viruses are a therapeutic moiety that is well suited to deliver and augment these immune-modulating therapies in a highly targeted and economically advantageous way over current treatment. In this review, we discuss the blockade of immune checkpoints, how oncolytic viruses complement and extend these therapies, and speculate on how this combination will uniquely impact the future of cancer immunotherapy.

20.
Artículo en Chino | WPRIM (Pacífico Occidental) | ID: wpr-454448

RESUMEN

Objective To investigate the regulatory effects of IFN-γon the expression of pro-grammed death ligand 2 (PDL2) on human placenta mesenchymal stem cells (hPMSCs) and the hPMSCs-induced differentiation of peripheral blood CD 8+IL-10+T cell subsets .Methods hPMSCs were isolated from mature human placenta by enzyme digestion .The expression of PDL2 on hPMSCs and the regulatory effects of IFN-γon PDL2 expression were detected by RT-PCR and flow cytometry ( FCM ) , respectively . Peripheral blood mononuclear cells (PBMCs) were isolated from healthy subjects by density gradient centrif-ugation.T cells were purified with sheep red blood cells .FCM was used to detect the ratios of CD 8+IL-10+T cell subsets in PHA or CD3/CD28 beads activated T cells in the presence of hPMSCs treated with Anti-PDL2 McAb or IFN-γ.Results PDL2 molecules were highly expressed on hPMSCs that could be further enhanced by IFN-γ.The results of FCM demonstrated that hPMSCs could induce the differentiation of CD 8+IL-10+T cell subsets .The ratios of CD8+IL-10+T cell population in T cells activated by different stimulators including PHA and CD3/CD28 beads were significantly increased in the presence of hPMSCs as compared with those without hPMSCs (P<0.01).In addition, the antibody blocking experiments indicated that PDL 2 McAb down-regulated the percentages of CD 8+IL-10+T cell subsets in PHA or CD 3/CD28 beads stimulated T cells in the presence of hPMSCs as compared with those of unblocked groups .CD8+IL-10+T cell subsets were up-regulated in IFN-γtreated hPMSCs groups as compared with those of untreated groups .Conclusion hPMSCs could induce the differentiation of peripheral blood T cells into CD 8+IL-10+T cell subsets , which was enhanced by PDL 2 expressed on hPMSCs .IFN-γcould promote the differentiation of CD 8+IL-10+T cell subsets induced by hPMSCs through up-regulating the expression of PDL2 on hPMSCs.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA