Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 258
Filtrar
1.
Nature ; 618(7966): 834-841, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37286599

RESUMO

Tumours most often arise from progression of precursor clones within a single anatomical niche. In the bone marrow, clonal progenitors can undergo malignant transformation to acute leukaemia, or differentiate into immune cells that contribute to disease pathology in peripheral tissues1-4. Outside the marrow, these clones are potentially exposed to a variety of tissue-specific mutational processes, although the consequences of this are unclear. Here we investigate the development of blastic plasmacytoid dendritic cell neoplasm (BPDCN)-an unusual form of acute leukaemia that often presents with malignant cells isolated to the skin5. Using tumour phylogenomics and single-cell transcriptomics with genotyping, we find that BPDCN arises from clonal (premalignant) haematopoietic precursors in the bone marrow. We observe that BPDCN skin tumours first develop at sun-exposed anatomical sites and are distinguished by clonally expanded mutations induced by ultraviolet (UV) radiation. A reconstruction of tumour phylogenies reveals that UV damage can precede the acquisition of alterations associated with malignant transformation, implicating sun exposure of plasmacytoid dendritic cells or committed precursors during BPDCN pathogenesis. Functionally, we find that loss-of-function mutations in Tet2, the most common premalignant alteration in BPDCN, confer resistance to UV-induced cell death in plasmacytoid, but not conventional, dendritic cells, suggesting a context-dependent tumour-suppressive role for TET2. These findings demonstrate how tissue-specific environmental exposures at distant anatomical sites can shape the evolution of premalignant clones to disseminated cancer.


Assuntos
Transformação Celular Neoplásica , Células Dendríticas , Leucemia Mieloide Aguda , Neoplasias Cutâneas , Pele , Raios Ultravioleta , Humanos , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Células da Medula Óssea/efeitos da radiação , Morte Celular/efeitos da radiação , Linhagem da Célula/genética , Linhagem da Célula/efeitos da radiação , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Transformação Celular Neoplásica/efeitos da radiação , Células Clonais/metabolismo , Células Clonais/patologia , Células Clonais/efeitos da radiação , Células Dendríticas/metabolismo , Células Dendríticas/patologia , Células Dendríticas/efeitos da radiação , Leucemia Mieloide Aguda/etiologia , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Mutação/efeitos da radiação , Especificidade de Órgãos , Análise da Expressão Gênica de Célula Única , Neoplasias Cutâneas/etiologia , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Raios Ultravioleta/efeitos adversos , Pele/patologia , Pele/efeitos da radiação
2.
Expert Rev Mol Med ; 26: e30, 2024 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-39438247

RESUMO

Radiotherapy (RT) plays a key role in the tumour microenvironment (TME), impacting the immune response via cellular and humoral immunity. RT can induce local immunity to modify the TME. It can stimulate dendritic cell maturation and T-cell infiltration. Moreover, B cells, macrophages and other immune cells may also be affected. Tertiary lymphoid structure (TLS) is a unique structure within the TME and a class of aggregates containing T cells, B cells and other immune cells. The maturation of TLS is determined by the presence of mature dendritic cells, the density of TLS is determined by the number of immune cells. TLS maturation and density both affect the antitumour immune response in the TME. This review summarized the recent research on the impact and the role of RT on TLS, including the changes of TLS components and formation conditions and the mechanism of how RT affects TLS and transforms the TME. RT may promote TLS maturation and density to modify the TME regarding enhanced antitumour immunity.


Assuntos
Neoplasias , Estruturas Linfoides Terciárias , Microambiente Tumoral , Humanos , Microambiente Tumoral/imunologia , Microambiente Tumoral/efeitos da radiação , Estruturas Linfoides Terciárias/imunologia , Estruturas Linfoides Terciárias/patologia , Neoplasias/imunologia , Neoplasias/radioterapia , Neoplasias/patologia , Neoplasias/terapia , Animais , Radioterapia/métodos , Células Dendríticas/imunologia , Células Dendríticas/efeitos da radiação
3.
Future Oncol ; 17(13): 1683-1694, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33726502

RESUMO

Radiation therapy (RT) in some cases results in a systemic anticancer response known as the abscopal effect. Multiple hypotheses support the role of immune activation initiated by RT-induced DNA damage. Optimal radiation dose is necessary to promote the cGAS-STING pathway in response to radiation and initiate an IFN-1 signaling cascade that promotes the maturation and migration of dendritic cells to facilitate antigen presentation and stimulation of cytotoxic T cells. T cells then exert a targeted response throughout the body at areas not subjected to RT. These effects are further augmented through the use of immunotherapeutic drugs resulting in increased T-cell activity. Tumor-infiltrating lymphocyte presence and TREX1, KPNA2 and p53 signal expression are being explored as prognostic biomarkers.


Assuntos
Quimiorradioterapia/métodos , Células Dendríticas/imunologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Neoplasias/radioterapia , Biomarcadores Tumorais/análise , Biomarcadores Tumorais/metabolismo , Movimento Celular/efeitos da radiação , Ensaios Clínicos como Assunto , Dano ao DNA/imunologia , Dano ao DNA/efeitos da radiação , Células Dendríticas/efeitos da radiação , Exodesoxirribonucleases/análise , Exodesoxirribonucleases/metabolismo , Humanos , Inibidores de Checkpoint Imunológico/farmacologia , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Linfócitos do Interstício Tumoral/efeitos da radiação , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/mortalidade , Fosfoproteínas/análise , Fosfoproteínas/metabolismo , Prognóstico , Intervalo Livre de Progressão , Dosagem Radioterapêutica , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T/efeitos da radiação , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia , Microambiente Tumoral/efeitos da radiação , Proteína Supressora de Tumor p53/análise , Proteína Supressora de Tumor p53/metabolismo , alfa Carioferinas/análise , alfa Carioferinas/metabolismo
4.
Immunology ; 159(2): 133-141, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31593303

RESUMO

The majority of human vaccines are administered above the deltoid muscle of the arm, a site that is chronically sun-exposed in many people. It is known that exposure of the skin to the UV wavelengths in sunlight stimulates systemic immunosuppression, an outcome that is associated with reduced immunity to microbial infections in animal models. Here we consider whether immunization of humans through a UV-irradiated skin site will lead to a less effective immune response compared with immunization through an unexposed site. Studies showing that the efficacy of vaccination can be reduced when surrogates of increased levels of sun exposure, such as latitude of residence and season of the year, are considered. Results from a limited number of intervention experiments in humans demonstrate a similar pattern. To provide an explanation for these findings, changes in the number and functional potential of immune cells in chronically sun-exposed compared with unexposed skin are outlined. UV radiation-induced changes to skin cells are also relevant when considering skin sites for administration of immune-tolerizing peptides. The review provides the basis for further research into the effects of acute and chronic UV radiation exposure on skin cells in the context of vaccination.


Assuntos
Tolerância Imunológica/efeitos da radiação , Imunogenicidade da Vacina , Pele/efeitos da radiação , Luz Solar , Vacinação , Vacinas/administração & dosagem , Animais , Células Dendríticas/imunologia , Células Dendríticas/efeitos da radiação , Humanos , Memória Imunológica/efeitos da radiação , Injeções Intradérmicas , Mastócitos/imunologia , Mastócitos/efeitos da radiação , Estações do Ano , Pele/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/efeitos da radiação
5.
Eur J Immunol ; 49(11): 2095-2102, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31334839

RESUMO

There is increasing evidence of the relevant connection and regulation between the gut and skin immune axis. In fact, oral administration of lipoteichoic acid (LTA) from Lactobacillus rhamnosus GG (LGG) prevents the development of UV-induced skin tumors in chronically exposed mice. Here we aim to evaluate whether this LTA is able to revert UV-induced immunosuppression as a mechanism involved in its anti-tumor effect and whether it has an immunotherapeutic effect against cutaneous squamous cell carcinoma. Using a mouse model of contact hypersensitivity, we demonstrate that LTA overcomes UV-induced skin immunosuppression. This effect was in part achieved by modulating the phenotype of lymph node resident dendritic cells (DC) and the homing of skin migratory DC. Importantly, oral LTA reduced significantly the growth of established skin tumors once UV radiation was discontinued, demonstrating that it has a therapeutic, besides the already demonstrated preventive antitumor effect. The data presented here strongly indicates that oral administration of LTA represents a promising immunotherapeutic approach for different conditions in which the skin immune system is compromised.


Assuntos
Antineoplásicos/farmacologia , Carcinoma de Células Escamosas/tratamento farmacológico , Lacticaseibacillus rhamnosus/química , Lipopolissacarídeos/farmacologia , Neoplasias Cutâneas/tratamento farmacológico , Ácidos Teicoicos/farmacologia , Raios Ultravioleta/efeitos adversos , Administração Oral , Animais , Antineoplásicos/isolamento & purificação , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/imunologia , Carcinoma de Células Escamosas/patologia , Movimento Celular/efeitos dos fármacos , Movimento Celular/imunologia , Movimento Celular/efeitos da radiação , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Células Dendríticas/patologia , Células Dendríticas/efeitos da radiação , Dermatite de Contato/genética , Dermatite de Contato/imunologia , Dermatite de Contato/patologia , Feminino , Trato Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/patologia , Trato Gastrointestinal/efeitos da radiação , Lipopolissacarídeos/isolamento & purificação , Linfonodos/efeitos dos fármacos , Linfonodos/imunologia , Linfonodos/patologia , Linfonodos/efeitos da radiação , Camundongos , Camundongos Endogâmicos C57BL , Pele/efeitos dos fármacos , Pele/imunologia , Pele/patologia , Pele/efeitos da radiação , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/patologia , Ácidos Teicoicos/isolamento & purificação
6.
FASEB J ; 33(2): 3074-3081, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30192655

RESUMO

Many vaccines require adjuvants to enhance immunogenicity, but there are few safe and effective intradermal (i.d.) adjuvants. Murine studies have validated the potency of laser illumination of skin as an adjuvant for i.d. vaccination with advantages over traditional adjuvants. We report a pilot clinical trial of low-power, continuous-wave, near-infrared laser adjuvant treatment, representing the first human trial of the safety, tolerability, and cutaneous immune cell trafficking changes produced by the laser adjuvant. In this trial we demonstrated a maximum tolerable energy dose of 300 J/cm2 to a spot on the lower back. The irradiated spot was biopsied 4 h later, as was a control spot. Paired biopsies were submitted for histomorphologic and immunohistochemical evaluation in a blinded fashion as well as quantitative PCR analysis for chemokines and cytokines. Similar to prior murine studies, highly significant reductions in CD1a+ Langerhans cells in the dermis and CD11c+ dermal dendritic cells were observed, corresponding to the increased migratory activity of these cells; changes in the epidermis were not significant. There was no evidence of skin damage. The laser adjuvant is a safe, well-tolerated adjuvant for i.d. vaccination in humans and results in significant cutaneous immune cell trafficking.-Gelfand, J. A., Nazarian, R. M., Kashiwagi, S., Brauns, T., Martin, B., Kimizuka, Y., Korek, S., Botvinick, E., Elkins, K., Thomas, L., Locascio, J., Parry, B., Kelly, K. M., Poznansky, M. C. A pilot clinical trial of a near-infrared laser vaccine adjuvant: safety, tolerability, and cutaneous immune cell trafficking.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Células Dendríticas/imunologia , Lasers , Pele/imunologia , Vacinas/administração & dosagem , Adolescente , Adulto , Células Cultivadas , Células Dendríticas/efeitos da radiação , Feminino , Humanos , Injeções Intradérmicas , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , Projetos Piloto , Pele/efeitos da radiação , Vacinação , Vacinas/imunologia , Adulto Jovem
7.
Mol Pharm ; 17(7): 2532-2545, 2020 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-32407125

RESUMO

Recent cancer immunotherapy has attracted much attention due to high specificity and recurrence prevention of tumor. Nevertheless, its therapeutic effects are still challenging in solid cancer. To establish superior antitumor immunity, chlorin e6 (Ce6)-loaded pH sensitive carbon dots were investigated (Ce6@IDCDs). At tumoral pH 6.5, Ce6 was released four times compared with the release at physiological pH 7.4 due to an imbalance between hydrophilic and hydrophobic forces via protonation of imidazole groups in Ce6@IDCDs. This result led to the superior singlet oxygen generating activity of Ce6@IDCDs without Ce6 quenching. The maturation effects of dendritic cells after co-incubation with supernatant media obtained from Ce6@IDCDs with laser-treated cells at pH 6.5 were much higher than at physiological pH. Furthermore, Ce6@IDCDs following a laser at pH 6.5 significantly promoted calreticulin exposure and high-mobility group box 1 release, as major immunogenic cell death markers. In bilateral CT-26-bearing mice model, the Ce6@IDCDs elicited significant antitumoral effects at laser treated-primary tumor regions via therapeutic reactive oxygen species. Furthermore, Ce6@IDCDs upon laser irradiation induced a large amount of activated CD8+ T cells, natural killer cells, and mature dendritic cells recruitment into tumoral tissue and hampered tumor growth even at untreated sites approximately four-fold compared with those of others. Overall, this pH-sensitive immunoinducer can accomplish primary and distant tumor ablation via photomediated cancer immunotherapy.


Assuntos
Carbono/química , Células Dendríticas/efeitos dos fármacos , Imunoterapia/métodos , Nanopartículas/química , Neoplasias/tratamento farmacológico , Fotoquimioterapia/métodos , Porfirinas/administração & dosagem , Radiossensibilizantes/administração & dosagem , Animais , Calreticulina/metabolismo , Morte Celular/efeitos dos fármacos , Morte Celular/imunologia , Morte Celular/efeitos da radiação , Linhagem Celular Tumoral , Clorofilídeos , Células Dendríticas/imunologia , Células Dendríticas/efeitos da radiação , Concentração de Íons de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Imidazóis/química , Lasers , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Microscopia Eletrônica de Transmissão , Nanopartículas/efeitos da radiação , Nanopartículas/ultraestrutura , Neoplasias/imunologia , Neoplasias/radioterapia , Tamanho da Partícula , Porfirinas/química , Porfirinas/farmacocinética , Radiossensibilizantes/química , Radiossensibilizantes/efeitos da radiação , Espécies Reativas de Oxigênio/metabolismo , Espécies Reativas de Oxigênio/uso terapêutico , Espectroscopia de Infravermelho com Transformada de Fourier , Microambiente Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Am J Transplant ; 19(5): 1344-1355, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30500995

RESUMO

Dendritic cells (DCs) are key targets for immunity and tolerance induction; they present donor antigens to recipient T cells by donor- and recipient-derived pathways. Donor-derived DCs, which are critical during the acute posttransplant period, can be depleted in graft tissue by forced migration via ultraviolet B light (UVB) irradiation. Here, we investigated the tolerogenic potential of donor-derived DC depletion through in vivo and ex vivo UVB preirradiation (UV) combined with the injection of anti-CD154 antibody (Ab) into recipients in an MHC-mismatched hair follicle (HF) allograft model in humanized mice. Surprisingly, human HF allografts achieved long-term survival with newly growing pigmented hair shafts in both Ab-treated groups (Ab-only and UV plus Ab) and in the UV-only group, whereas the control mice rejected all HF allografts with no hair regrowth. Perifollicular human CD3+ T cell and MHC class II+ cell infiltration was significantly diminished in the presence of UV and/or Ab treatment. HF allografts in the UV-only group showed stable maintenance of the immune privilege in the HF epithelium without evidence of antigen-specific T cell tolerance, which is likely promoted by normal HFs in vivo. This immunomodulatory strategy targeting the donor tissue exhibited novel biological relevance for clinical allogeneic transplantation without generalized immunosuppression.


Assuntos
Células Dendríticas/imunologia , Rejeição de Enxerto/prevenção & controle , Sobrevivência de Enxerto/imunologia , Folículo Piloso/crescimento & desenvolvimento , Tolerância Imunológica/imunologia , Doadores de Tecidos , Raios Ultravioleta , Animais , Células Dendríticas/efeitos da radiação , Rejeição de Enxerto/etiologia , Rejeição de Enxerto/imunologia , Sobrevivência de Enxerto/efeitos da radiação , Folículo Piloso/imunologia , Folículo Piloso/efeitos da radiação , Humanos , Tolerância Imunológica/efeitos da radiação , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Transplante Homólogo
9.
Cancer Sci ; 109(4): 956-965, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29465830

RESUMO

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.


Assuntos
Carcinoma Pulmonar de Lewis/radioterapia , Proliferação de Células/efeitos da radiação , Receptor 3 Toll-Like/metabolismo , Animais , Antígenos de Neoplasias/metabolismo , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/efeitos da radiação , Carcinoma Pulmonar de Lewis/tratamento farmacológico , Carcinoma Pulmonar de Lewis/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Terapia Combinada/métodos , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Células Dendríticas/efeitos da radiação , Modelos Animais de Doenças , Imunoterapia Adotiva/métodos , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos/efeitos da radiação , Camundongos , Camundongos Endogâmicos C57BL , Poli I-C/farmacologia , Linfócitos T Citotóxicos/efeitos dos fármacos , Linfócitos T Citotóxicos/metabolismo , Linfócitos T Citotóxicos/efeitos da radiação
10.
Photochem Photobiol Sci ; 17(12): 1872-1884, 2018 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-29136080

RESUMO

The realisation that UV radiation (UVR) exposure could induce a suppressed immune environment for the initiation of carcinogenesis in the skin was first described more than 40 years ago. Van der Leun and his colleagues contributed to this area in the 1980s and 90s by experiments in mice involving UV wavelength and dose-dependency in the formation of such tumours, in addition to illustrating both the local and systemic effect of the UVR on the immune system. Since these early days, many aspects of the complex pathways of UV-induced immunosuppression have been studied and are outlined in this review. Although most experimental work has involved mice, it is clear that UVR also causes reduced immune responses in humans. Evidence showing the importance of the immune system in determining the risk of human skin cancers is explained, and details of how UVR exposure can down-regulate immunity in the formation and progression of such tumours reviewed. With increasing knowledge of these links and the mechanisms of UVR-induced immunosuppression, novel approaches to enhance immunity to skin tumour antigens in humans are becoming apparent which, hopefully, will reduce the burden of UVR-induced skin cancers in the future.


Assuntos
Tolerância Imunológica/efeitos da radiação , Neoplasias Cutâneas/etiologia , Raios Ultravioleta , Animais , Antígeno B7-H1/química , Antígeno B7-H1/metabolismo , Células Dendríticas/metabolismo , Células Dendríticas/efeitos da radiação , Humanos , Sistema Imunitário/metabolismo , Sistema Imunitário/efeitos da radiação , Neuropeptídeos/metabolismo , Receptores de Citocinas/agonistas , Receptores de Citocinas/metabolismo , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/metabolismo
11.
Med Sci Monit ; 24: 1457-1463, 2018 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-29525808

RESUMO

BACKGROUND Carbon ion radiotherapy has been shown to be more effective in cancer radiotherapy than photon irradiation. Influence of carbon ion radiation on cancer microenvironment is very important for the outcomes of radiotherapy. Tumor-infiltrating dendritic cells (DCs) play critical roles in cancer antigen processing and antitumor immunity. However, there is scant literature covering the effects of carbon ion radiation on DCs. In this study, we aimed to uncover the impact of carbon ion irradiation on bone marrow derived DCs. MATERIAL AND METHODS Bone marrow cells were co-cultured with GM-CSF and IL-4 for seven days, and the population of DCs was confirmed with flow cytometry. We used an Annexin V and PI staining method to detect cell apoptosis. Endocytosis assay of DCs was determined by using a flow cytometry method. DCs migration capacity was tested by a Transwell method. We also used ELISA assay and western blotting assay to examine the cytokines and protein expression, respectively. RESULTS Our data showed that carbon ion radiation induced apoptosis in both immature and mature DCs. After irradiation, the endocytosis and migration capacity of DCs was also impaired. Interestingly, carbon irradiation triggered a burst of IFN-g and IL-12 in LPS or CpG treated DCs, which provide novel insights into the combination of immunotherapy and carbon ion radiotherapy. Finally, we found that carbon ion irradiation induced apoptosis and migration suppression was p38 dependent. CONCLUSIONS Our present study demonstrated that carbon ion irradiation induced apoptosis in DCs, and impaired DCs function mainly through the p38 signaling pathway. Carbon ion irradiation also triggered anti-tumor cytokines secretion. This work provides novel information of carbon ion radiotherapy in DCs, and also provides new insights on the combination of immune adjuvant and carbon ion radiotherapy.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/efeitos da radiação , Radioterapia com Íons Pesados , Animais , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Células da Medula Óssea/citologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/efeitos da radiação , Movimento Celular/efeitos dos fármacos , Movimento Celular/efeitos da radiação , Células Dendríticas/citologia , Células Dendríticas/efeitos dos fármacos , Endocitose/efeitos dos fármacos , Endocitose/efeitos da radiação , Feminino , Interferon gama/metabolismo , Interleucina-12/metabolismo , Lipopolissacarídeos/farmacologia , Camundongos Endogâmicos C57BL , Oligodesoxirribonucleotídeos/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
12.
J Immunol ; 194(6): 2569-77, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25681340

RESUMO

Extracorporeal photopheresis (ECP) is a widely used clinical cell-based therapy exhibiting efficacy in heterogenous immune-mediated diseases such as cutaneous T cell lymphoma, graft-versus-host disease, and organ allograft rejection. Despite its documented efficacy in cancer immunotherapy, little is known regarding the induction of immunostimulatory mediators by ECP. In this article, we show that ECP promotes marked release of the prototypic immunostimulatory cytokine IL-1ß. ECP primes IL-1ß production and activates IL-1ß maturation and release in the context of caspase-1 activation in monocytes and myeloid dendritic cells. Of interest, IL-1ß maturation by ECP was fully intact in murine cells deficient in caspase-1, suggesting the predominance of an inflammasome-independent pathway for ECP-dependent IL-1ß maturation. Clinically, patient analysis revealed significantly increased IL-1ß production in stimulated leukapheresis concentrates and peripheral blood samples after ECP. Collectively, these results provide evidence for promotion of IL-1ß production by ECP and offer new insight into the immunostimulatory capacity of ECP.


Assuntos
Interleucina-1beta/biossíntese , Interleucina-1beta/sangue , Leucócitos Mononucleares/metabolismo , Fotoferese/métodos , Trifosfato de Adenosina/metabolismo , Trifosfato de Adenosina/farmacologia , Animais , Caspase 1/genética , Caspase 1/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Células Cultivadas , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Células Dendríticas/efeitos da radiação , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/efeitos da radiação , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Inflamassomos/efeitos dos fármacos , Inflamassomos/metabolismo , Inflamassomos/efeitos da radiação , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/efeitos da radiação , Receptores de Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/farmacologia , Metoxaleno/farmacologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Monócitos/efeitos da radiação , Fármacos Fotossensibilizantes/farmacologia , Raios Ultravioleta
13.
Cancer Immunol Immunother ; 65(7): 779-86, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26590829

RESUMO

Radiotherapy (RT) utilizes the DNA-damaging properties of ionizing radiation to control tumor growth and ultimately kill tumor cells. By modifying the tumor cell phenotype and the tumor microenvironment, it may also modulate the immune system. However, out-of-field reactions of RT mostly assume further immune activation. Here, the sequence of the applications of RT and immunotherapy is crucial, just as the dose and fractionation may be. Lower single doses may impact on tumor vascularization and immune cell infiltration in particular, while higher doses may impact on intratumoral induction and production of type I interferons. The induction of immunogenic cancer cell death seems in turn to be a common mechanism for most RT schemes. Dendritic cells (DCs) are activated by the released danger signals and by taking up tumor peptides derived from irradiated cells. DCs subsequently activate T cells, a process that has to be tightly controlled to ensure tolerance. Inhibitory pathways known as immune checkpoints exist for this purpose and are exploited by tumors to inhibit immune responses. Cytotoxic T lymphocyte antigen 4 (CTLA-4) and programmed cell death protein 1 (PD-1) on T cells are two major checkpoints. The biological concepts behind the findings that RT in combination with anti-CTLA-4 and/or anti-PD-L1 blockade stimulates CD8+ T cell-mediated anti-tumor immunity are reviewed in detail. On this basis, we suggest clinically significant combinations and sequences of RT and immune checkpoint inhibition. We conclude that RT and immune therapies complement one another.


Assuntos
Imunoterapia/métodos , Neoplasias/imunologia , Neoplasias/radioterapia , Adjuvantes Imunológicos/uso terapêutico , Animais , Terapia Combinada , Células Dendríticas/imunologia , Células Dendríticas/efeitos da radiação , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/terapia , Radiação Ionizante , Linfócitos T/imunologia , Linfócitos T/efeitos da radiação , Regulação para Cima
14.
Int J Hyperthermia ; 32(1): 23-30, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26754406

RESUMO

Mild hyperthermia (HT) (41.5 °C for 30-60 min) has been shown in various cell culture systems, preclinical and clinical models to be a very potent radiosensitiser. Recent research suggests that local HT application in combination with standard tumour therapies such as radiotherapy (RT) and/or chemotherapy may not only improve local tumour control but also lead to systemic and immune mediated anti-tumour responses. Melanoma has been proven to be rather radioresistant and mostly only the addition of immunotherapy is capable of inducing beneficial anti-melanoma responses. This work therefore focuses on whether HT increases the immunogenic potential of B16-F10 mouse melanoma cells in combination with RT. The in vitro experiments revealed that combination of RT with HT resulted in an increased percentage of apoptotic and necrotic melanoma cells and an increased release of the danger signal heat shock protein 70 (Hsp70) and high mobility group box protein 1 (HMGB1). HT alone was also capable of inducing this release. We set up local irradiation and heating procedures of B16-F10 tumour-bearing C57/BL6 mice and revealed that the tumour growth of tumours treated with RT plus HT was significantly retarded compared to tumours treated only with RT. This combined treatment generated a beneficial tumour microenvironment by enhancing the infiltration of CD11c + /MHCII + /CD86+ dendritic cells, CD8+ T cells, and NK cells, and decreasing that of regulatory T cells and myeloid-derived suppressor cells. We conclude that HT in combination with RT has an immune-stimulating potential that might result in anti-tumour immunity.


Assuntos
Hipertermia Induzida , Melanoma Experimental/terapia , Radiação Ionizante , Animais , Apoptose/efeitos da radiação , Linhagem Celular Tumoral , Terapia Combinada , Células Dendríticas/imunologia , Células Dendríticas/efeitos da radiação , Feminino , Proteína HMGB1/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/efeitos da radiação , Melanoma Experimental/imunologia , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos Endogâmicos C57BL , Necrose/radioterapia , Necrose/terapia , Linfócitos T/imunologia , Linfócitos T/efeitos da radiação , Carga Tumoral/efeitos da radiação
15.
Clin Exp Immunol ; 181(3): 491-510, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25902927

RESUMO

In this study we examined the effects of non-myeloablative total body irradiation (TBI) in combination with immunosuppressive chemotherapy on immune homeostasis in rhesus macaques. Our results show that the administration of cyclosporin A or tacrolimus without radiotherapy did not result in lymphopenia. The addition of TBI to the regimen resulted in lymphopenia as well as alterations in the memory/naive ratio following reconstitution of lymphocyte populations. Dendritic cell (DC) numbers in whole blood were largely unaffected, while the monocyte population was altered by immunosuppressive treatment. Irradiation also resulted in increased levels of circulating cytokines and chemokines that correlated with T cell proliferative bursts and with the shift towards memory T cells. We also report that anti-thymocyte globulin (ATG) treatment and CD3 immunotoxin administration resulted in a selective and rapid depletion of naive CD4 and CD8 T cells and increased frequency of memory T cells. We also examined the impact of these treatments on reactivation of latent simian varicella virus (SVV) infection as a model of varicella zoster virus (VZV) infection of humans. None of the treatments resulted in overt SVV reactivation; however, select animals had transient increases in SVV-specific T cell responses following immunosuppression, suggestive of subclinical reactivation. Overall, we provide detailed observations into immune modulation by TBI and chemotherapeutic agents in rhesus macaques, an important research model of human disease.


Assuntos
Sistema Imunitário/efeitos dos fármacos , Sistema Imunitário/efeitos da radiação , Imunossupressores/farmacologia , Irradiação Corporal Total/métodos , Animais , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Ciclosporina/farmacologia , Citocinas/sangue , Citocinas/metabolismo , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Células Dendríticas/efeitos da radiação , Ensaio de Imunoadsorção Enzimática , Feminino , Homeostase/efeitos dos fármacos , Homeostase/efeitos da radiação , Sistema Imunitário/citologia , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Leucócitos/efeitos dos fármacos , Leucócitos/metabolismo , Leucócitos/efeitos da radiação , Contagem de Linfócitos , Linfócitos/efeitos dos fármacos , Linfócitos/metabolismo , Linfócitos/efeitos da radiação , Macaca mulatta/virologia , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Monócitos/efeitos da radiação , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Neutrófilos/efeitos da radiação , Tacrolimo/farmacologia , Varicellovirus/efeitos dos fármacos , Varicellovirus/crescimento & desenvolvimento , Varicellovirus/efeitos da radiação , Carga Viral/efeitos dos fármacos , Carga Viral/efeitos da radiação , Ativação Viral/efeitos dos fármacos , Ativação Viral/efeitos da radiação
16.
Ann Neurol ; 75(5): 739-58, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24771567

RESUMO

OBJECTIVE: Environmental conditions (eg, latitude) play a critical role in the susceptibility and severity of many autoimmune disorders, including multiple sclerosis (MS). Here, we investigated the mechanisms underlying the beneficial effects of immune regulatory processes induced in the skin by moderate ultraviolet B (UVB) radiation on central nervous system (CNS) autoimmunity. METHODS: Effects of UVB light were analyzed in a murine model of CNS autoimmunity (experimental autoimmune encephalomyelitis). Additionally, patients with relapsing-remitting MS were treated with narrowband UVB phototherapy. Immunomodulatory effects were examined in skin biopsies, serum samples, and immune cells of the peripheral blood. RESULTS: Regulatory T cells (Tregs), which are induced locally in the skin-draining lymph nodes in response to UVB exposure, connect the cutaneous immune response to CNS immunity by migration to the sites of inflammation (blood, spleen, CNS). Here, they attenuate the inflammatory response and ameliorate disease symptoms. Treg-inducing tolerogenic dendritic cells (DCs) were further necessary for induction of this systemic immune regulation by UVB radiation, because ablation of Langerhans cells abolished the UVB-induced phenotype. MS patients treated with UVB phototherapy showed an increase in induced Tregs and tolerogenic DCs accompanied by the downregulation of the T-cell effector cytokine interleukin 21. The treatment further induced elevated serum levels of vitamin D. INTERPRETATION: Local UVB radiation of the skin influences systemic immune reactions and attenuates systemic autoimmunity via the induction of skin-derived tolerogenic DCs and Tregs. Our data could have implications for the understanding or therapeutic modulation of environmental factors that influence immune tolerance.


Assuntos
Encefalomielite Autoimune Experimental/radioterapia , Imunidade Celular/efeitos da radiação , Esclerose Múltipla Recidivante-Remitente/radioterapia , Linfócitos T Reguladores/efeitos da radiação , Raios Ultravioleta , Terapia Ultravioleta , Adulto , Animais , Células Cultivadas , Células Dendríticas/imunologia , Células Dendríticas/efeitos da radiação , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Feminino , Humanos , Imunidade Celular/imunologia , Masculino , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Esclerose Múltipla Recidivante-Remitente/imunologia , Esclerose Múltipla Recidivante-Remitente/patologia , Linfócitos T Reguladores/imunologia , Terapia Ultravioleta/métodos , Adulto Jovem
17.
Photochem Photobiol Sci ; 14(8): 1492-1509, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26062987

RESUMO

Photodynamic therapy (PDT) uses the combination of non-toxic photosensitizers and harmless light to generate reactive oxygen species that destroy tumors by a combination of direct tumor cell killing, vascular shutdown, and activation of the immune system. It has been shown in some animal models that mice that have been cured of cancer by PDT, may exhibit resistance to rechallenge. The cured mice can also possess tumor specific T-cells that recognize defined tumor antigens, destroy tumor cells in vitro, and can be adoptively transferred to protect naïve mice from cancer. However, these beneficial outcomes are the exception rather than the rule. The reasons for this lack of consistency lie in the ability of many tumors to suppress the host immune system and to actively evade immune attack. The presence of an appropriate tumor rejection antigen in the particular tumor cell line is a requisite for T-cell mediated immunity. Regulatory T-cells (CD25+, Foxp3+) are potent inhibitors of anti-tumor immunity, and their removal by low dose cyclophosphamide can potentiate the PDT-induced immune response. Treatments that stimulate dendritic cells (DC) such as CpG oligonucleotide can overcome tumor-induced DC dysfunction and improve PDT outcome. Epigenetic reversal agents can increase tumor expression of MHC class I and also simultaneously increase expression of tumor antigens. A few clinical reports have shown that anti-tumor immunity can be generated by PDT in patients, and it is hoped that these combination approaches may increase tumor cures in patients.


Assuntos
Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Fotoquimioterapia/métodos , Linfócitos T/imunologia , Animais , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Células Dendríticas/efeitos da radiação , Epigênese Genética/efeitos dos fármacos , Epigênese Genética/efeitos da radiação , Humanos , Neoplasias/genética , Linfócitos T/efeitos dos fármacos , Linfócitos T/efeitos da radiação
18.
J Immunol ; 190(11): 5471-84, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23636055

RESUMO

Alterations to dendritic cell (DC) progenitors in the bone marrow (BM) may contribute to long-lasting systemic immunosuppression (>28 d) following exposure of the skin of mice to erythemal UV radiation (UVR). DCs differentiated in vitro from the BM of mice 3 d after UVR (8 kJ/m(2)) have a reduced capacity to initiate immunity (both skin and airways) when adoptively transferred into naive mice. Studies in IL-10(-/-) mice suggested that UV-induced IL-10 was not significantly involved. To investigate the immune capabilities of peripheral tissue DCs generated in vivo from the BM of UV-irradiated mice, chimeric mice were established. Sixteen weeks after reconstitution, contact hypersensitivity responses were significantly reduced in mice reconstituted with BM from UV-irradiated mice (UV-chimeric). When the dorsal skin of UV-chimeric mice was challenged with innate inflammatory agents, the hypertrophy induced in the draining lymph nodes was minimal and significantly less than that measured in control-chimeric mice challenged with the same inflammatory agent. When DCs were differentiated from the BM of UV-chimeric mice using FLT3 ligand or GM-CSF + IL-4, the cells maintained a reduced priming ability. The diminished responses in UV-chimeric mice were not due to different numerical or proportional reconstitution of BM or the hematopoietic cells in blood, lymph nodes, and skin. Erythemal UVR may imprint a long-lasting epigenetic effect on DC progenitors in the BM and alter the function of their terminally differentiated progeny.


Assuntos
Transplante de Medula Óssea , Células Dendríticas/imunologia , Células Dendríticas/efeitos da radiação , Sobrevivência de Enxerto/imunologia , Raios Ultravioleta , Transferência Adotiva , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/imunologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/imunologia , Diferenciação Celular/efeitos da radiação , Movimento Celular/imunologia , Quimerismo/efeitos da radiação , Células Dendríticas/citologia , Células Dendríticas/efeitos dos fármacos , Dermatite de Contato/imunologia , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Hipertrofia , Imunidade Inata , Interleucina-4/farmacologia , Linfonodos/imunologia , Linfonodos/patologia , Linfonodos/efeitos da radiação , Proteínas de Membrana/farmacologia , Camundongos , Linfócitos T/imunologia , Linfócitos T/efeitos da radiação
19.
Cancer Cell ; 12(4): 300-1, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17936555

RESUMO

Recent literature has highlighted an important role of inflammation in promoting cancer. However, the immune system can also play a central role in protecting the body against cancer as well as infection, although its role in cancer is not well understood. A study published in the September issue of Nature Medicine adds a new twist to the role of inflammation in cancer. Apetoh et al. describe how activation of innate immunity after conventional radiation or chemotherapy can trigger protective antitumor immunity.


Assuntos
Antineoplásicos/farmacologia , Imunidade Celular , Imunidade Inata , Inflamação/imunologia , Neoplasias/tratamento farmacológico , Neoplasias/radioterapia , Receptor 4 Toll-Like/metabolismo , Animais , Antineoplásicos/uso terapêutico , Morte Celular , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Células Dendríticas/efeitos da radiação , Proteína HMGB1/metabolismo , Humanos , Imunidade Celular/efeitos dos fármacos , Imunidade Celular/efeitos da radiação , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/efeitos da radiação , Inflamação/patologia , Camundongos , Fator 88 de Diferenciação Mieloide/metabolismo , Neoplasias/imunologia , Neoplasias/patologia , Linfócitos T Citotóxicos/efeitos dos fármacos , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/efeitos da radiação
20.
J Am Chem Soc ; 136(31): 10823-5, 2014 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-25029205

RESUMO

The innate immune response is controlled, in part, by the synergistic interaction of multiple Toll-like receptors (TLRs). This multi-receptor cooperation is responsible for the potent activity of many vaccines, but few tools have been developed to understand the spatio-temporal elements of TLR synergies. In this Communication, we present photo-controlled agonists of TLR7/8. By strategically protecting the active agonist moiety based on an agonist-bound crystal structure, TLR activity is suppressed and then regained upon exposure to light. We confirmed NF-κB production upon light exposure in a model macrophage cell line. Primary cell activity was confirmed by examining cytokine and cell surface marker production in bone-marrow-derived dendritic cells. Finally, we used light to activate dendritic cell sub-populations within a larger population.


Assuntos
Células Dendríticas/imunologia , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/efeitos da radiação , Luz , Receptor 7 Toll-Like/agonistas , Receptor 8 Toll-Like/agonistas , Aminoquinolinas/química , Aminoquinolinas/farmacologia , Animais , Células da Medula Óssea/citologia , Linhagem Celular , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Células Dendríticas/efeitos da radiação , Imiquimode , Camundongos , Modelos Moleculares , Multimerização Proteica , Estrutura Quaternária de Proteína , Receptor 7 Toll-Like/metabolismo , Receptor 8 Toll-Like/química , Receptor 8 Toll-Like/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa