Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Cancer Sci ; 114(4): 1256-1269, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36529525

RESUMEN

We previously reported that regulatory T (Treg) cells expressing CTLA-4 on the cell surface are abundant in head and neck squamous cell carcinoma (HNSCC). The role of expanded Treg cells in the tumor microenvironment of HNSCC remains unclear. In this study, we reveal that the tumor microenvironment of HNSCC is characterized by the high expression of genes related to Treg cells, dendritic cells (DCs), and interleukin (IL)-17-related molecules. Increased expression of IL17A, IL17F, or IL23A contributes to a favorable prognosis of HNSCC. In the tumor microenvironment of HNSCC, IL23A and IL12B are expressed in mature dendritic cells enriched in regulatory molecules (mregDCs). The mregDCs in HNSCC are a migratory and mature phenotype; their signature genes strongly correlate with Treg signature genes in HNSCC. We also observed that IL17A was highly expressed in Th17 cells and exhausted CD8+ T cells in HNSCC. These data suggest that mregDCs in HNSCC may contribute to the prognosis by balancing Treg cells and effector T cells that produce IL-17. Targeting mregDCs may be a novel strategy for developing new immune therapies against HNSCC.


Asunto(s)
Neoplasias de Cabeza y Cuello , Linfocitos T Reguladores , Humanos , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Linfocitos T CD8-positivos , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/metabolismo , Pronóstico , Células Dendríticas , Microambiente Tumoral
2.
PLoS Pathog ; 17(12): e1010085, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34882757

RESUMEN

Regulatory T (Treg) cells, which constitute about 5-10% of CD4+T cells expressing Foxp3 transcription factor and CD25(IL-2 receptor α chain), are key regulators in controlling immunological self-tolerance and various immune responses. However, how Treg cells control antigen-specific immunity to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) remains unclear. In this study, we examined the effect of transient breakdown of the immunological tolerance induced by Treg-cell depletion on adaptive immune responses against administered SARS-CoV-2 antigen, spike protein 1 (S1). Notably, without the use of adjuvants, transient Treg-cell depletion in mice induced anti-S1 antibodies that neutralized authentic SARS-CoV-2, follicular helper T cell formation and S1-binding germinal center B cell responses, but prevented the onset of developing autoimmune diseases. To further clarify the mechanisms, we investigated maturation of dendritic cells (DCs), which is essential to initiate antigen-specific immunity. We found that the transient Treg-cell depletion resulted in maturation of both migratory and resident DCs in draining lymph nodes that captured S1-antigen. Moreover, we observed S1-specific CD4+ T cells and CD8+ T cells with interferon-γ production. Thus, captured S1 was successfully presented by DCs, including cross-presentation to CD8+ T cells. These data indicate that transient Treg-cell depletion in the absence of adjuvants induces maturation of antigen-presenting DCs and succeeds in generating antigen-specific humoral and cellular immunity against emerging SARS-CoV-2 antigens. Finally, we showed that SARS-CoV-2 antigen-specific immune responses induced by transient Treg-cell depletion in the absence of adjuvants were compatible with those induced with an effective adjuvant, polyriboinosinic:polyribocytidyl acid (poly IC) and that the combination of transient Treg-cell depletion with poly IC induced potent responses. These findings highlight the capacity for manipulating Treg cells to induce protective adaptive immunity to SARS-CoV-2 with activating antigen-presenting DCs, which may improve the efficacy of ongoing vaccine therapies and help enhance responses to emerging SARS-CoV-2 variants.


Asunto(s)
Inmunidad Adaptativa/inmunología , Antígenos Virales/inmunología , COVID-19/inmunología , Factores de Transcripción Forkhead/inmunología , SARS-CoV-2/inmunología , Animales , Presentación de Antígeno/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , COVID-19/virología , Chlorocebus aethiops , Células Dendríticas/inmunología , Femenino , Centro Germinal/inmunología , Humanos , Tolerancia Inmunológica , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos MRL lpr , Linfocitos T Reguladores/inmunología , Células Vero
3.
Proc Natl Acad Sci U S A ; 117(34): 20696-20705, 2020 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-32769209

RESUMEN

Regulatory T (Treg) cells, expressing CD25 (interleukin-2 receptor α chain) and Foxp3 transcription factor, maintain immunological self-tolerance and suppress various immune responses. Here we report a feature of skin Treg cells expanded by ultraviolet B (UVB) exposure. We found that skin Treg cells possessing a healing function are expanded by UVB exposure with the expression of an endogenous opioid precursor, proenkephalin (PENK). Upon UVB exposure, skin Treg cells were expanded with a unique TCR repertoire. Also, they highly expressed a distinctive set of genes enriched in "wound healing involved in inflammatory responses" and the "neuropeptide signaling pathway," as indicated by the high expression of Penk. We found that not only was PENK expression at the protein level detected in the UVB-expanded skin Treg (UVB-skin Treg) cells, but that a PENK-derived neuropeptide, methionine enkephalin (Met-ENK), from Treg cells promoted the outgrowth of epidermal keratinocytes in an ex vivo skin explant assay. Notably, UVB-skin Treg cells also promoted wound healing in an in vivo wound closure assay. In addition, UVB-skin Treg cells produced amphiregulin (AREG), which plays a key role in Treg-mediated tissue repair. Identification of a unique function of PENK+ UVB-skin Treg cells provides a mechanism for maintaining skin homeostasis.


Asunto(s)
Encefalinas/metabolismo , Precursores de Proteínas/metabolismo , Linfocitos T Reguladores/metabolismo , Cicatrización de Heridas/fisiología , Anfirregulina/metabolismo , Animales , Células Cultivadas , Encefalina Metionina/metabolismo , Encefalinas/efectos de la radiación , Femenino , Homeostasis/fisiología , Humanos , Tolerancia Inmunológica/inmunología , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Precursores de Proteínas/efectos de la radiación , Autotolerancia/inmunología , Piel/metabolismo , Rayos Ultravioleta , Cicatrización de Heridas/inmunología
4.
J Immunol ; 200(1): 119-129, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29158419

RESUMEN

Skin dendritic cells (DCs) are divided into several subsets with distinctive functions. This study shows a previously unappreciated role of dermal CD11b-type Langerin- DCs in maintaining immunological self-tolerance after UVB exposure. After UVB exposure, dermal CD11b-type Langerin- DCs upregulated surface CD86 expression, induced proliferation of Foxp3+ regulatory T (Treg) cells without exogenous Ags, and upregulated a set of genes associated with immunological tolerance. This Treg-expansion activity was significantly hampered by CD80/CD86 blockade in vivo. These results indicate that CD11b-type Langerin- DCs from the UVB-exposed skin are specialized to expand Treg cells in the skin, which suppress autoimmunity.


Asunto(s)
Células Dendríticas/inmunología , Piel/patología , Linfocitos T Reguladores/inmunología , Animales , Antígenos CD/metabolismo , Antígeno CD11b/genética , Antígeno CD11b/metabolismo , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Tolerancia Inmunológica/genética , Lectinas Tipo C/metabolismo , Activación de Linfocitos , Lectinas de Unión a Manosa/metabolismo , Ratones , Ratones Endogámicos C57BL , Transcriptoma , Rayos Ultravioleta/efectos adversos
6.
Cancer Sci ; 109(7): 2119-2129, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29791768

RESUMEN

Immunological checkpoint blockade therapies benefit a limited population of cancer patients. We have previously shown that vaccine immunotherapy with Toll-like receptor (TLR)3-adjuvant and tumor antigen overcomes anti-programmed death ligand-1 (PD-L1) resistance in mouse tumor models. In the present study, 4 different ovalbumin (OVA)-expressing tumor cell lines were implanted into syngeneic mice and subjected to anti-tumor immunotherapy using ARNAX and whole OVA protein. ARNAX is a TLR3-specific agonist that does not activate the mitochondrial antiviral-signaling protein (MAVS) pathway, and thus does not induce systemic inflammation. Dendritic cell priming and proliferative CTL were induced by ARNAX + OVA, but complete remission was achieved only in a PD-L1-low cell line of EG7. Addition of anti-PD-L1 antibody to the ARNAX + OVA therapy brought complete remission to another PD-L1-high subline of EG7. Tumor shrinkage but not remission was observed in MO5 in that regimen. We analyzed tumor cells and tumor-infiltrating immune cells to identify factors associated with successful ARNAX vaccine therapy. Tumors that responded to ARNAX therapy expressed high levels of MHC class I and low levels of PD-L1. The tumor-infiltrating immune cells in ARNAX-susceptible tumors contained fewer immunosuppressive myeloid cells with low PD-L1 expression. Combination with anti-PD-L1 antibody functioned not only within tumor sites but also within lymphoid tissues, augmenting the therapeutic efficacy of the ARNAX vaccine. Notably, ARNAX therapy induced memory CD8+ T cells and rejection of reimplanted tumors. Thus, ARNAX vaccine + anti-PD-L1 therapy enabled permanent remission against some tumors that stably present antigens.


Asunto(s)
Inmunoterapia/métodos , Linfocitos T/inmunología , Receptor Toll-Like 3/agonistas , Animales , Antígenos de Neoplasias/inmunología , Modelos Animales de Enfermedad , Memoria Inmunológica/inmunología , Ratones , Ratones Endogámicos C57BL , Microambiente Tumoral/inmunología
7.
Cancer Sci ; 109(4): 956-965, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29465830

RESUMEN

Radiotherapy induces anti-tumor immunity by induction of tumor antigens and damage-associated molecular patterns (DAMP). DNA, a representative DAMP in radiotherapy, activates the stimulator of interferon genes (STING) pathway which enhances the immune response. However, the immune response does not always parallel the inflammation associated with radiotherapy. This lack of correspondence may, in part, explain the radiation-resistance of tumors. Additive immunotherapy is expected to revive tumor-specific CTL facilitating radiation-resistant tumor shrinkage. Herein pre-administration of the double-stranded RNA, polyinosinic-polycytidylic acid (polyI:C), in conjunction with radiotherapy, was shown to foster tumor suppression in mice bearing radioresistant, ovalbumin-expressing Lewis lung carcinoma (LLC). Extrinsic injection of tumor antigen was not required for tumor suppression. No STING- and CTL-response was induced by radiation in the implant tumor. PolyI:C was more effective for induction of tumor growth retardation at 1 day before radiation than at post-treatment. PolyI:C targeted Toll-like receptor 3 with minimal effect on the mitochondrial antiviral-signaling protein pathway. Likewise, the STING pathway barely contributed to LLC tumor suppression. PolyI:C primed antigen-presenting dendritic cells in draining lymph nodes to induce proliferation of antigen-specific CTL. By combination therapy, CTL efficiently infiltrated into tumors with upregulation of relevant chemokine transcripts. Batf3-positive DC and CD8+ T cells were essential for therapeutic efficacy. Furthermore, polyI:C was shown to stimulate tumor-associated macrophages and release tumor necrosis factor alpha, which acted on tumor cells and increased sensitivity to radiation. Hence, polyI:C treatment prior to radiotherapy potentially induces tumor suppression by boosting CTL-dependent and macrophage-mediated anti-tumor responses. Eventually, polyI:C and radiotherapy in combination would be a promising therapeutic strategy for radiation-resistant tumors.


Asunto(s)
Carcinoma Pulmonar de Lewis/radioterapia , Proliferación Celular/efectos de la radiación , Receptor Toll-Like 3/metabolismo , Animales , Antígenos de Neoplasias/metabolismo , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/efectos de la radiación , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Carcinoma Pulmonar de Lewis/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Terapia Combinada/métodos , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Células Dendríticas/efectos de la radiación , Modelos Animales de Enfermedad , Inmunoterapia Adoptiva/métodos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , Poli I-C/farmacología , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/metabolismo , Linfocitos T Citotóxicos/efectos de la radiación
8.
J Biomed Sci ; 24(1): 79, 2017 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-29041928

RESUMEN

BACKGROUND: Intestinal tumorigenesis is promoted by myeloid differentiation primary response gene 88 (MyD88) activation in response to the components of microbiota in Apc Min/+ mice. Microbiota also contains double-stranded RNA (dsRNA), a ligand for TLR3, which activates the toll-like receptor adaptor molecule 1 (TICAM-1, also known as TRIF) pathway. METHODS: We established Apc Min/+ Ticam1 -/- mice and their survival was compared to survival of Apc Min/+ Myd88 -/- and wild-type (WT) mice. The properties of polyps were investigated using immunofluorescence staining and RT-PCR analysis. RESULTS: We demonstrate that TICAM-1 is essential for suppression of polyp formation in Apc Min/+ mice. TICAM-1 knockout resulted in shorter survival of mice compared to WT mice or mice with knockout of MyD88 in the Apc Min/+ background. Polyps were more frequently formed in the distal intestine of Apc Min/+ Ticam1 -/- mice than in Apc Min/+ mice. Infiltration of immune cells such as CD11b+ and CD8α+ cells into the polyps was detected histologically. CD11b and CD8α mRNAs were increased in polyps of Apc Min/+ Ticam1 -/- mice compared to Apc Min/+ mice. Gene expression of inducible nitric oxide synthase (iNOS), interferon (IFN)-γ, CXCL9 and IL-12p40 was increased in polyps of Apc Min/+ Ticam1 -/- mice. mRNA and protein expression of c-Myc, a critical transcription factor for inflammation-associated polyposis, were increased in polyps of Apc Min/+ Ticam1 -/- mice. A Lactobacillus strain producing dsRNA was detected in feces of Apc Min/+ mice. CONCLUSION: These results imply that the TLR3/TICAM-1 pathway inhibits polyposis through suppression of c-Myc expression and supports long survival in Apc Min/+ mice.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/genética , Pólipos del Colon/genética , Neoplasias Colorrectales/genética , Pólipos Intestinales/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Transducción de Señal , Receptor Toll-Like 3/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor Toll-Like 3/metabolismo
9.
Cancer Sci ; 107(5): 644-52, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26931406

RESUMEN

Transforming growth factor-ß activated kinase 1 (TAK1) has been shown to play a crucial role in cell death, differentiation, and inflammation. Here, we live-imaged robust TAK1 activation in Lewis lung carcinoma 3LL cells implanted into the s.c. tissue of syngeneic C57BL/6 mice and treated with polyinosinic:polycytidylic acid (PolyI:C). First, we developed and characterized a Förster resonance energy transfer-based biosensor for TAK1 activity. The TAK1 biosensor, named Eevee-TAK1, responded to stress-inducing reagents such as anisomycin, tumor necrosis factor-α, and interleukin1-ß. The anisomycin-induced increase in Förster resonance energy transfer was abolished by the TAK1 inhibitor (5z)-7-oxozeaenol. Activity of TAK1 in 3LL cells was markedly increased by PolyI:C in the presence of macrophages. 3LL cells expressing Eevee-TAK1 were implanted into mice and observed through imaging window by two-photon excitation microscopy. During the growth of tumor, the 3LL cells at the periphery of the tumor showed higher TAK1 activity than the 3LL cells located at the center of the tumor, suggesting that cells at the periphery of the tumor mass were under stronger stress. Injection of PolyI:C, which is known to induce regression of the implanted tumors, induced marked and homogenous TAK1 activation within the tumor tissues. The effect of PolyI:C faded within 4 days. These observations suggest that Eevee-TAK1 is a versatile tool to monitor cellular stress in cancer tissues.


Asunto(s)
Técnicas Biosensibles/métodos , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Carcinoma Pulmonar de Lewis/enzimología , Quinasas Quinasa Quinasa PAM/metabolismo , Imagen Molecular/métodos , Poli I-C/uso terapéutico , Animales , Anisomicina/farmacología , Carcinoma Pulmonar de Lewis/patología , Supervivencia Celular , Activación Enzimática/efectos de los fármacos , Humanos , Interleucina-1beta/farmacología , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Microscopía , Poli I-C/farmacología , Estrés Fisiológico/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Zearalenona/análogos & derivados , Zearalenona/farmacología
10.
BMC Immunol ; 17(1): 9, 2016 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-27141827

RESUMEN

BACKGROUND: Triggering receptors expressed on myeloid cells (Trem) proteins are a family of cell surface receptors used to control innate immune responses such as proinflammatory cytokine production in mice. Trem genes belong to a rapidly expanding family of receptors that include activating and inhibitory paired-isoforms. RESULTS: By comparative genomic analysis, we found that Trem4, Trem5 and Trem-like transcript-6 (Treml6) genes typically paired receptors. These paired Trem genes were murine-specific and originated from an immunoreceptor tyrosine-based inhibition motif (ITIM)-containing gene. Treml6 encoded ITIM, whereas Trem4 and Trem5 lacked the ITIM but possessed positively-charged residues to associate with DNAX activating protein of 12 kDa (DAP12). DAP12 was directly associated with Trem4 and Trem5, and DAP12 coupling was mandatory for their expression on the cell surface. In bone marrow-derived dendritic cells (BMDCs) and macrophages (BMDMs), and splenic DC subsets, polyinosinic-polycytidylic acid (polyI:C) followed by type I interferon (IFN) production induced Trem4 and Treml6 whereas polyI:C or other TLR agonists failed to induce the expression of Trem5. PolyI:C induced Treml6 and Trem4 more efficiently in BMDMs than BMDCs. Treml6 was more potentially up-regulated in conventional DC (cDCs) and plasmacytoid DC (pDCs) than Trem4 in mice upon in vivo stimulation with polyI:C. DISCUSSION: Treml6-dependent inhibitory signal would be dominant in viral infection compared to resting state. Though no direct ligands of these Trem receptors have been determined, the results infer that a set of Trem receptors are up-regulated in response to viral RNA to regulate myeloid cell activation through modulation of DAP12-associated Trem4 and ITIM-containing Treml6.


Asunto(s)
Células Dendríticas/inmunología , Macrófagos/inmunología , Receptores Inmunológicos/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/genética , Animales , Células Cultivadas , Femenino , Regulación de la Expresión Génica , Inmunidad Innata , Interferón Tipo I/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Dominios Proteicos/genética , ARN Bicatenario/inmunología , Receptor de Interferón alfa y beta/genética , Receptores Inmunológicos/genética
11.
Biochem Biophys Res Commun ; 478(4): 1764-71, 2016 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-27608599

RESUMEN

An interferon-inducing DNA sensor STING participates in tumor rejection in mouse models. Here we examined what mechanisms contribute to STING-dependent growth retardation of B16 melanoma sublines by NK cells in vivo. The studies were designed using WT and STING KO black mice, and B16D8 (an NK-sensitive melanoma line having STING) and STING KO B16D8 sublines established for this study. The results from tumor-implant studies suggested that STING in host immune cells and tumor cells induced distinct profiles of chemokines including CXCL10, CCL5 and IL-33, and both participated in NK cell infiltration and activation in B16D8 tumor. Spontaneous activation of STING occurs in host-immune and tumor cells of this NK-sensitive tumor, thereby B16D8 tumor growth being suppressed in this model. Our data show that STING induces tumor cytotoxicity by NK cells through tumor and host immune cell network to contribute to innate surveillance and suppression of tumors in vivo.


Asunto(s)
Proliferación Celular/genética , Células Asesinas Naturales/metabolismo , Melanoma Experimental/genética , Proteínas de la Membrana/genética , Carga Tumoral/genética , Animales , Western Blotting , Línea Celular Tumoral , Quimiocina CCL5/genética , Quimiocina CCL5/metabolismo , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Interferón beta/genética , Interferón beta/metabolismo , Interleucina-33/genética , Interleucina-33/metabolismo , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Matriz Nuclear/genética , Proteínas Asociadas a Matriz Nuclear/metabolismo , Proteínas de Transporte Nucleocitoplasmático/genética , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
Cancer Sci ; 106(12): 1659-68, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26395101

RESUMEN

Immune-enhancing adjuvants usually targets antigen (Ag)-presenting cells to tune up cellular and humoral immunity. CD141(+) dendritic cells (DC) represent the professional Ag-presenting cells in humans. In response to microbial pattern molecules, these DCs upgrade the maturation stage sufficient to improve cross-presentation of exogenous Ag, and upregulation of MHC and costimulators, allowing CD4/CD8 T cells to proliferate and liberating cytokines/chemokines that support lymphocyte attraction and survival. These DCs also facilitate natural killer-mediated cell damage. Toll-like receptors (TLRs) and their signaling pathways in DCs play a pivotal role in DC maturation. Therefore, providing adjuvants in addition to Ag is indispensable for successful vaccine immunotherapy for cancer, which has been approved in comparison with antimicrobial vaccines. Mouse CD8α(+) DCs express TLR7 and TLR9 in addition to the TLR2 family (TLR1, 2, and 6) and TLR3, whereas human CD141(+) DCs exclusively express the TLR2 family and TLR3. Although human and mouse plasmacytoid DCs commonly express TLR7/9 to respond to their agonists, the results on mouse adjuvant studies using TLR7/9 agonists cannot be simply extrapolated to human adjuvant immunotherapy. In contrast, TLR2 and TLR3 are similarly expressed in both human and mouse Ag-presenting DCs. Bacillus Calmette-Guerin peptidoglycan and polyinosinic-polycytidylic acid are representative agonists for TLR2 and TLR3, respectively, although they additionally stimulate cytoplasmic sensors: their functional specificities may not be limited to the relevant TLRs. These adjuvants have been posted up to a certain achievement in immunotherapy in some cancers. We herein summarize the history and perspectives of TLR2 and TLR3 agonists in vaccine-adjuvant immunotherapy for cancer.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Vacunas contra el Cáncer/inmunología , Inmunoterapia/métodos , Receptor Toll-Like 2/agonistas , Receptor Toll-Like 3/agonistas , Animales , Humanos
13.
Biochem Biophys Res Commun ; 457(3): 445-50, 2015 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-25596131

RESUMEN

Myeloid-derived suppressor cells (MDSCs) are immature myeloid cells that exhibit potent immunosuppressive activity. They are increased in tumor-bearing hosts and contribute to tumor development. Toll-like receptors (TLRs) on MDSCs may modulate the tumor-supporting properties of MDSCs through pattern-recognition. Pam2 lipopeptides represented by Pam2CSK4 serve as a TLR2 agonist to exert anti-tumor function by dendritic cell (DC)-priming that leads to NK cell activation and cytotoxic T cell proliferation. On the other hand, TLR2 enhances tumor cell progression/invasion by activating tumor-infiltrating macrophages. How MDSCs respond to TLR2 agonists has not yet been determined. In this study, we found intravenous administration of Pam2CSK4 systemically up-regulated the frequency of MDSCs in EG7 tumor-bearing mice. The frequency of tumor-infiltrating MDSCs was accordingly increased in response to Pam2CSK4. MDSCs were not increased by Pam2CSK4 stimuli in TLR2 knockout (KO) mice. Adoptive transfer experiments using CFSE-labeled MDSCs revealed that the TLR2-positive MDSCs survived long in tumor-bearing mice in response to Pam2CSK4 treatment. Since the increased MDSC population sustained immune-suppressive properties, our study suggests that Pam2CSK4-triggered TLR2 activation enhances the MDSC potential and suppress antitumor immune response in tumor microenvironment.


Asunto(s)
Lipopéptidos/farmacología , Células Mieloides/efectos de los fármacos , Células Mieloides/metabolismo , Receptor Toll-Like 2/metabolismo , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Femenino , Inmunoterapia , Ligandos , Lipopéptidos/inmunología , Lipopéptidos/metabolismo , Linfoma/inmunología , Linfoma/metabolismo , Linfoma/terapia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Células Mieloides/inmunología , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 2/deficiencia , Receptor Toll-Like 2/genética , Microambiente Tumoral
14.
Proc Natl Acad Sci U S A ; 109(6): 2066-71, 2012 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-22308357

RESUMEN

Smoldering inflammation often increases the risk of progression for malignant tumors and simultaneously matures myeloid dendritic cells (mDCs) for cell-mediated immunity. PolyI:C, a dsRNA analog, is reported to induce inflammation and potent antitumor immune responses via the Toll-like receptor 3/Toll-IL-1 receptor domain-containing adaptor molecule 1 (TICAM-1) and melanoma differentiation-associated protein 5/IFN-ß promoter stimulator 1 (IPS-1) pathways in mDCs to drive activation of natural killer cells and cytotoxic T lymphocytes. Here, we found that i.p. or s.c. injection of polyI:C to Lewis lung carcinoma tumor-implant mice resulted in tumor regression by converting tumor-supporting macrophages (Mfs) to tumor suppressors. F4/80(+)/Gr1(-) Mfs infiltrating the tumor respond to polyI:C to rapidly produce inflammatory cytokines and thereafter accelerate M1 polarization. TNF-α was increased within 1 h in both tumor and serum upon polyI:C injection into tumor-bearing mice, followed by tumor hemorrhagic necrosis and growth suppression. These tumor responses were abolished in TNF-α(-/-) mice. Furthermore, F4/80(+) Mfs in tumors extracted from polyI:C-injected mice sustained Lewis lung carcinoma cytotoxic activity, and this activity was partly abrogated by anti-TNF-α Ab. Genes for supporting M1 polarization were subsequently up-regulated in the tumor-infiltrating Mfs. These responses were completely abrogated in TICAM-1(-/-) mice, and unaffected in myeloid differentiation factor 88(-/-) and IPS-1(-/-) mice. Thus, the TICAM-1 pathway is not only important to mature mDCs for cross-priming and natural killer cell activation in the induction of tumor immunity, but also critically engaged in tumor suppression by converting tumor-supporting Mfs to those with tumoricidal properties.


Asunto(s)
Carcinoma Pulmonar de Lewis/inmunología , Carcinoma Pulmonar de Lewis/patología , Células Mieloides/inmunología , Células Mieloides/patología , Transducción de Señal/inmunología , Receptor Toll-Like 3/inmunología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Antineoplásicos/farmacología , Polaridad Celular/efectos de los fármacos , Polaridad Celular/inmunología , Citotoxicidad Inmunológica/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/patología , Ratones , Poli I-C/farmacología , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
15.
J Biol Chem ; 286(12): 10702-11, 2011 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-21266579

RESUMEN

The double-stranded RNA analog, poly(I:C), extracellularly activates both the endosomal Toll-like receptor (TLR) 3 and the cytoplasmic RNA helicase, melanoma differentiation-associated gene 5, leading to the production of type I interferons (IFNs) and inflammatory cytokines. The mechanism by which extracellular poly(I:C) is delivered to TLR3-positive organelles and the cytoplasm remains to be elucidated. Here, we show that the cytoplasmic lipid raft protein, Raftlin, is essential for poly(I:C) cellular uptake in human myeloid dendritic cells and epithelial cells. When Raftlin was silenced, poly(I:C) failed to enter cells and induction of IFN-ß production was inhibited. In addition, cellular uptake of B-type oligodeoxynucleotide that shares its uptake receptor with poly(I:C) was suppressed in Raftlin knockdown cells. Upon poly(I:C) stimulation, Raftlin was translocated from the cytoplasm to the plasma membrane where it colocalized with poly(I:C), and thereafter moved to TLR3-positive endosomes. Thus, Raftlin cooperates with the uptake receptor to mediate cell entry of poly(I:C), which is critical for activation of TLR3.


Asunto(s)
Células Dendríticas/metabolismo , Inductores de Interferón/farmacología , Proteínas de la Membrana/metabolismo , Células Mieloides/metabolismo , Poli I-C/farmacología , Receptor Toll-Like 3/metabolismo , Animales , Línea Celular Tumoral , Membrana Celular/genética , Membrana Celular/metabolismo , Células Dendríticas/citología , Endosomas/genética , Endosomas/metabolismo , Silenciador del Gen , Células HEK293 , Humanos , Interferón beta/biosíntesis , Interferón beta/genética , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Células Mieloides/citología , Receptor Toll-Like 3/agonistas , Receptor Toll-Like 3/genética
16.
Int Immunol ; 23(1): 29-41, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21131367

RESUMEN

Interleukin-17A (IL-17A) is a cytokine produced by T(h)17 cells that plays an important role in inflammatory and autoimmune diseases and cancer. Stimulation with IL-6, transforming growth factor-ß , IL-21, IL-1ß and IL-23 is required for differentiation of T(h)17 cells and the production of IL-17A. Recently, we reported that tumor-derived lactic acid enhances the toll-like receptor (TLR) ligand-mediated expression of IL-23, leading to increased IL-17A production. Tumor cells secrete large amounts of lactic acid due to the up-regulation of glycolysis, which is known as the Warburg effect. Even without TLR ligand stimulation, lactic acid enhanced antigen-dependent IL-17A production from splenocytes in an IL-23-dependent manner. Here, we show that macrophages and effector/memory CD4(+) T cells are the primary cell types involved in the ability of lactic acid to boost IL-17A production. Although lactic acid suppressed the proliferation of T(h)1 and T(h)17 cells, T(h)17 cells still secreted large amounts of IL-17A. CD40 ligand-CD40 interactions were involved in the up-regulation of IL-17A by lactic acid through IL-12/23p40 production. A new cytokine containing the IL-12/23p40 subunit, but not IL-23, IL-12 or the IL-12p40 homodimer, is a candidate for involvement in the up-regulation of IL-17A. IL-1ß also increased IL-17A expression; however, IL-1ß, CARD9 and MyD88 signaling pathways activated by known intrinsic inflammatory mediators were hardly required for the enhanced activity induced by lactic acid. Our results show that lactic acid functions as an intrinsic inflammatory mediator that activates IL-23-dependent and -independent pathways, resulting in the promotion of chronic inflammation in tumor microenvironments.


Asunto(s)
Subunidad p40 de la Interleucina-12/inmunología , Interleucina-17/biosíntesis , Interleucina-23/inmunología , Ácido Láctico/inmunología , Neoplasias/inmunología , Animales , Antígeno CD11b/metabolismo , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Células Cultivadas , Células Dendríticas/inmunología , Memoria Inmunológica , Interleucina-23/química , Ácido Láctico/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Células Th17/efectos de los fármacos , Células Th17/inmunología , Regulación hacia Arriba
17.
Microbiol Immunol ; 55(5): 373-7, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21517948

RESUMEN

The immune system has evolved mechanisms to sense not only microbes, but also necrotic cells. The pattern-recognition receptors in macrophages/dendritic cells that stimulate the acquired immune system are closely associated with danger signaling. In this study monoclonal antibodies (mAbs) that specifically interact with necrotic cells were developed. One IgG1 and two IgM mAbs were established, and they recognized a 80 kDa protein expressed in necrotic, but not live or apoptotic, cells. These mAbs, which serve as a probe for necrosis, facilitate analyses of the role of the immune complex that consists of necrotic cells and Ab and contributes to the formation of the inflammatory milieu induced by necrotic cell death.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Linfoma/inmunología , Proteínas de la Membrana/inmunología , Necrosis , Anticuerpos Monoclonales/metabolismo , Línea Celular Tumoral , Humanos , Inmunoglobulina G/inmunología , Inmunoglobulina G/metabolismo , Inmunoglobulina M/inmunología , Inmunoglobulina M/metabolismo , Linfoma/patología , Proteínas de la Membrana/metabolismo
18.
Cancer Sci ; 101(2): 313-20, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20059475

RESUMEN

Dendritic cells (DC) begin maturation in response to complex stimuli consisting of antigens and pattern molecules (PAMP) for the activation of the immune system. Immune adjuvant usually contains PAMP. Infection represents one event that is capable of inducing such a complex set of stimuli. Recently, DC were subdivided into a number of subsets with distinct cell-surface markers, with each subset displaying unique differential maturation in response to pattern molecules to induce various types of effector cells. In the present study, we review how pattern recognition molecules and adaptors in each DC subset drive immune effector cells and their effect in the stimulated DC. Although tumor cells harbor tumor-associated antigens, they usually lack PAMP. Hence, we outline the properties of exogenously-added PAMP in the modulation of raising tumor immunity. In addition, we describe the mechanism by which DC-dependent natural killer activation is triggered for the induction of antitumor immunity.


Asunto(s)
Inmunidad Innata , Neoplasias/terapia , Receptores de Reconocimiento de Patrones/fisiología , Proteínas Adaptadoras del Transporte Vesicular/fisiología , Animales , Células Dendríticas/inmunología , Humanos , Inmunoterapia , Células Asesinas Naturales/inmunología , Activación de Linfocitos , Células Mieloides/inmunología , Factor 88 de Diferenciación Mieloide/fisiología , Neoplasias/inmunología , Receptores Toll-Like/fisiología
19.
Cancer Sci ; 101(7): 1596-603, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20507323

RESUMEN

The development of effective immunoadjuvants for tumor immunotherapy is of fundamental importance. The use of Mycobacterium bovis bacillus Calmette-Guérin cell wall skeleton (BCG-CWS) in tumor immunotherapy has been examined in various clinical applications. Because BCG-CWS is a macromolecule that cannot be chemically synthesized, the development of an alternative synthetic molecule is necessary to ensure a constant supply of adjuvant. In the present study, a new adjuvant was designed based on the structure of macrophage-activating lipopeptide (MALP)-2, which is a Toll-like receptor (TLR)-2 ligand similar to BCG-CWS. Macrophage-activating lipopeptide-2, [S-(2,3-bispalmitoyloxypropyl)Cys (P2C) - GNNDESNISFKEK], originally identified in a Mycoplasma species, is a lipopeptide that can be chemically synthesized. A MALP-2 peptide was substituted with a functional motif, RGDS, creating a novel molecule named P2C-RGDS. RGDS was selected because its sequence constitutes an integrin-binding motif and various integrins are expressed in immune cells including dendritic cells (DCs). Thus, this motif adds functionality to the ligand. P2C-RGDS activated DCs and splenocytes more efficiently than MALP-2 over short incubation times in vitro, and the RGDS motif contributed to their activation. Furthermore, P2C-RGDS showed higher activity than MALP-2 in inducing migration of DCs to draining lymph node, and in inhibiting tumor growth in vivo. This process of designing and developing synthetic adjuvants has been named "adjuvant engineering," and the evaluation and improvement of P2C-RGDS constitutes a first step in the development of stronger synthetic adjuvants in the future.


Asunto(s)
Inmunoterapia/métodos , Neoplasias/inmunología , Ingeniería de Tejidos/métodos , Receptor Toll-Like 2/inmunología , Adyuvantes Inmunológicos/química , Adyuvantes Inmunológicos/uso terapéutico , Secuencia de Aminoácidos , Animales , Vacuna BCG/uso terapéutico , Adhesión Celular/inmunología , Adhesión Celular/fisiología , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Dendríticas/inmunología , Ligandos , Lipopéptidos/uso terapéutico , Ganglios Linfáticos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oligopéptidos/química , Oligopéptidos/inmunología , Receptor Toll-Like 1/deficiencia , Receptor Toll-Like 1/genética
20.
Mol Biol Cell ; 18(5): 1701-9, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17332504

RESUMEN

The tumor suppressor p53 is a key transcriptional factor regulating the induction of cellular senescence by oncogenic signals. The activity of p53 is regulated by recruitment into promyelocytic leukemia (PML)-nuclear bodies (NBs) as well as by stabilization through posttranslational modifications such as phosphorylation and acetylation. Here we found that MORC3 (microrchidia3)-ATPase activated p53 and induced cellular senescence in normal human and mouse fibroblasts but not p53-/- fibroblasts. Conversely, genotoxic stress-induced phosphorylation and stabilization of p53 but barely increased its transcriptional activity in Morc3-/- fibroblasts. MORC3 localized on PML-NBs in presence of PML and mediated recruitment of p53 and CREB-binding protein (CBP) into PML-NBs. In contrast, expression of ATPase activity-deficient mutant MORC3-E35A or siRNA repression of MORC3 impaired the localization of p53 and Sp100 but not CBP on PML-NBs. These results suggest that MORC3 regulates p53 activity and localization into PML-NBs. We identified a new molecular mechanism that regulates the activity of nuclear proteins by localization to a nuclear subdomain.


Asunto(s)
Proteínas Nucleares/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Antígenos Nucleares/metabolismo , Autoantígenos/metabolismo , Línea Celular , Núcleo Celular/metabolismo , Senescencia Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Doxorrubicina/farmacología , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Genes p53 , Células HeLa , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transfección , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA