Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 738
Filtrar
1.
BMC Immunol ; 21(1): 18, 2020 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-32299365

RESUMEN

BACKGROUND: The use of immunotherapy strategies for the treatment of advanced cancer is rapidly increasing. Most immunotherapies rely on induction of CD8+ tumor-specific cytotoxic T cells that are capable of directly killing cancer cells. Tumors, however, utilize a variety of mechanisms that can suppress anti-tumor immunity. CD4+ regulatory T cells can directly inhibit cytotoxic T cell activity and these cells can be recruited, or induced, by cancer cells allowing escape from immune attack. The use of ionizing radiation as a treatment for cancer has been shown to enhance anti-tumor immunity by several mechanisms including immunogenic tumor cell death and phenotypic modulation of tumor cells. Less is known about the impact of radiation directly on suppressive regulatory T cells. In this study we investigate the direct effect of radiation on human TREG viability, phenotype, and suppressive activity. RESULTS: Both natural and TGF-ß1-induced CD4+ TREG cells exhibited increased resistance to radiation (10 Gy) as compared to CD4+ conventional T cells. Treatment, however, decreased Foxp3 expression in natural and induced TREG cells and the reduction was more robust in induced TREGS. Radiation also modulated the expression of signature iTREG molecules, inducing increased expression of LAG-3 and decreased expression of CD25 and CTLA-4. Despite the disconcordant modulation of suppressive molecules, irradiated iTREGS exhibited a reduced capacity to suppress the proliferation of CD8+ T cells. CONCLUSIONS: Our findings demonstrate that while human TREG cells are more resistant to radiation-induced death, treatment causes downregulation of Foxp3 expression, as well as modulation in the expression of TREG signature molecules associated with suppressive activity. Functionally, irradiated TGF-ß1-induced TREGS were less effective at inhibiting CD8+ T cell proliferation. These data suggest that doses of radiotherapy in the hypofractionated range could be utilized to effectively target and reduce TREG activity, particularly when used in combination with cancer immunotherapies.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/efectos de la radiación , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/efectos de la radiación , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/efectos de la radiación , Antígeno CTLA-4/inmunología , Muerte Celular/inmunología , Muerte Celular/efectos de la radiación , Células Cultivadas , Factores de Transcripción Forkhead/inmunología , Humanos , Inmunoterapia/métodos , Activación de Linfocitos/inmunología , Activación de Linfocitos/efectos de la radiación , Neoplasias/inmunología , Fenotipo , Radiación Ionizante , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/efectos de la radiación , Factor de Crecimiento Transformador beta1/inmunología
2.
Electromagn Biol Med ; 39(4): 316-322, 2020 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-32783480

RESUMEN

PURPOSE: Theoretical and experimental evidences support the hypothesis that Extremely Low-Frequency Electromagnetic Fields (ELF-EMF) can modulate voltage-gated channels. In this work we investigated the effect of ELF-EMF on Kv1.3, a member of the family of the voltage-gated potassium channels that is thought to be involved in key physiological functions, including the regulation of T-cells activation during the immune response. MATERIALS AND METHODS: Kv1.3 expressing CHO-K1 cells were exposed to a 20 Hz electromagnetic field at two different intensities: 268 µT and 902 µT. Kv1.3 potassium currents were recorded by whole-cell patch-clamp before, during and after field exposure. RESULTS: We found that the Kv1.3 current was increased significantly by the ELF-EMF in a subpopulation of CHO-K1 cells. The increase developed after a few seconds from the start of exposure, reached a steady-state and took several minutes to return to the baseline after field removal. CONCLUSIONS: These findings suggest that Kv1.3 may mediate interactions between ELF-EMF and living cells, disclosing new research opportunities on the molecular mechanisms with which electromagnetic fields affect physiological and pathological processes, including immunomodulation, inflammation and cancer.


Asunto(s)
Campos Electromagnéticos , Canal de Potasio Kv1.3/metabolismo , Animales , Células CHO , Cricetulus , Fenómenos Electrofisiológicos/efectos de la radiación , Activación de Linfocitos/efectos de la radiación , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/efectos de la radiación
3.
Strahlenther Onkol ; 194(5): 392-402, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29663037

RESUMEN

BACKGROUND: For head and neck squamous cell cancer (HNSCC), standard therapy consists of surgery, radiation, and/or chemotherapy. Antineoplastic immunotherapy could be an option in an adjuvant setting and is already in palliation. A functional immune system is a prerequisite for successful immunotherapy. However, effects of the standard-of-care therapy on the patients' immune system are not fully understood. METHODS: Peripheral blood mononuclear cells (PBMC) were collected from patients with HNSCC (n = 37) and healthy controls (n = 10). PBMC were stimulated with staphylococcal enterotoxin B (SEB). Simultaneous expression of various cytokines was measured in CD4+ and CD8+ T cells by multicolor flow cytometry, and polyfunctional cytokine expression profiles were determined on a single-cell basis. RESULTS: Expression levels of all measured cytokines in CD4+ T cells were higher in patients after chemoradiotherapy (CRT) as compared to untreated HNSCC patients or normal controls. After CRT, the frequency of polyfunctional CD4+ T cells, which simultaneously expressed multiple cytokines, was significantly increased as compared to untreated patients (p < 0.01). CONCLUSION: CRT increases polyfunctionality of CD4+ T cells in HNSCC patients, suggesting that standard-of-care therapy can promote immune activity in immune cells. These polyfunctional CD4+ T cells in the blood of treated HNSCC patients are expected to be responsive to subsequent immunotherapeutic approaches.


Asunto(s)
Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/efectos de la radiación , Carcinoma de Células Escamosas/inmunología , Carcinoma de Células Escamosas/terapia , Quimioradioterapia , Neoplasias de Oído, Nariz y Garganta/inmunología , Neoplasias de Oído, Nariz y Garganta/terapia , Anciano , Carcinoma de Células Escamosas/patología , Terapia Combinada , Citocinas/sangre , Femenino , Citometría de Flujo , Humanos , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/efectos de la radiación , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Neoplasias de Oído, Nariz y Garganta/patología
4.
Transfusion ; 58(6): 1506-1515, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29607502

RESUMEN

INTRODUCTION: Transfusion-associated graft-versus-host disease (TA-GVHD) is a rare complication after transfusion of components containing viable donor T cells. Gamma irradiation with doses that stop T-cell proliferation is the predominant method to prevent TA-GVHD. Treatment with pathogen inactivation methodologies has been found to also be effective against proliferating white blood cells, including T cells. In this study, T-cell inactivation was compared, between amotosalen/ultraviolet A (UVA) treatment and gamma-irradiation (2500 cGy), using a sensitive limiting dilution assay (LDA) with an enhanced dynamic range. METHODS AND MATERIALS: Matched plasma units (N = 8), contaminated with 1 × 106 peripheral blood mononuclear cells (PBMCs) per mL, were either treated with amotosalen/UVA or gamma irradiation, or retained as untreated control. Posttreatment, cells were cultured under standardized conditions. T-cell proliferation was determined by the incorporation of 3 H-thymidine and correlated with microscopic detection. RESULTS: Range-finding experiments showed that after gamma irradiation (2500 cGy), significant T-cell proliferation could be observed at a 1 × 107 cell culture density, some proliferation at 1 × 106 , and none at 1 × 105 cells/well. Based on these facts, a quantitative comparison was carried out between amotosalen/UVA at the highest challenge of 1 × 107 PBMCs/well, and gamma irradiation at 1 × 106 and 1 × 105 PBMCs/well. Complete inactivation of the T cells after amotosalen/UVA treatment was observed, equivalent to greater than 6.2 log inactivation. Complete inactivation of the T cells was also observed after gamma irradiation when 1 × 105 PBMCs/well were cultured (>4.2 log inactivation). Proliferation was observed when 1 × 106 PBMCs/well were cultured (≤5.2 log inactivation) after gamma irradiation. CONCLUSION: Amotosalen/UVA treatment more effectively inactivates T cells than the current standard of gamma irradiation (2500 cGy) for the prevention of TA-GVHD.


Asunto(s)
Rayos gamma , Enfermedad Injerto contra Huésped/prevención & control , Linfocitos T/efectos de la radiación , Reacción a la Transfusión/prevención & control , Rayos Ultravioleta , Transfusión Sanguínea , Proliferación Celular/efectos de la radiación , Furocumarinas/farmacología , Furocumarinas/uso terapéutico , Humanos , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/efectos de la radiación , Linfocitos T/citología , Linfocitos T/efectos de los fármacos , Resultado del Tratamiento
5.
Arterioscler Thromb Vasc Biol ; 37(1): 66-74, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27765767

RESUMEN

OBJECTIVE: UVB irradiation is an established treatment for immunoinflammatory cutaneous disorders and has been shown to suppress cutaneous and systemic inflammatory diseases through modulation of the adaptive immune response. However, it remains unknown whether UVB irradiation prevents an immunoinflammatory disease of arteries such as atherosclerosis. APPROACH AND RESULTS: Here, we show that UVB exposure inhibits the development and progression of atherosclerosis in atherosclerosis-prone mice by expanding and enhancing the functional capacity of CD4+ forkhead box P3+ regulatory T cells and regulating proatherogenic T-cell responses. Experimental studies in Langerhans cell-depleted mice revealed that epidermal Langerhans cells play a critical role in UVB-dependent induction of CD4+ forkhead box P3+ regulatory T cells, suppression of proatherogenic T-cell responses, and prevention of atherosclerotic plaque development. CONCLUSIONS: Our findings suggest the skin immune system as a novel therapeutic target for atherosclerosis and provide a novel strategy for the treatment and prevention of atherosclerosis.


Asunto(s)
Aorta/efectos de la radiación , Enfermedades de la Aorta/prevención & control , Aterosclerosis/prevención & control , Inflamación/prevención & control , Piel/efectos de la radiación , Linfocitos T Reguladores/efectos de la radiación , Rayos Ultravioleta , Animales , Aorta/inmunología , Aorta/metabolismo , Aorta/patología , Enfermedades de la Aorta/inmunología , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/patología , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/inmunología , Aterosclerosis/metabolismo , Aterosclerosis/patología , Células Cultivadas , Modelos Animales de Enfermedad , Factores de Transcripción Forkhead/inmunología , Factores de Transcripción Forkhead/metabolismo , Predisposición Genética a la Enfermedad , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Células de Langerhans/inmunología , Células de Langerhans/metabolismo , Células de Langerhans/efectos de la radiación , Activación de Linfocitos/efectos de la radiación , Ratones Noqueados , Fenotipo , Placa Aterosclerótica , Transducción de Señal/efectos de la radiación , Piel/inmunología , Piel/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo
6.
Eur J Nucl Med Mol Imaging ; 44(2): 242-246, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27717998

RESUMEN

PURPOSE: Therapy with the alpha-emitter radium-223 chloride (223Ra) is an innovative therapeutic option in patients with metastasized, castration-resistant prostate cancer. However, radiotherapy can lead to hematopoietic toxicity. The aim of this study was to determine if 223Ra therapy induces an impairment of cellular antimicrobial immune responses. METHODS: In 11 patients receiving 223Ra treatment, lymphocyte proliferation and the production of pro- and anti-inflammatory cytokines (interferon-γ and interleukin-10) were determined, using lymphocyte transformation testing and ELISpot, respectively. Lymphocyte function after stimulation with mitogens and microbial antigens was assessed prior to therapy and at day 1, 7 and 28 after therapy. RESULTS: Lymphocyte proliferation and the production of interferon-γ and interleukin-10 towards mitogens and antigens remained unchanged after therapy. Consistent with these in vitro data, we did not observe infectious complications after treatment. CONCLUSIONS: The results argue against an impairment of lymphocyte function after 223Ra therapy. Thus, immune responses against pathogens should remain unaffected.


Asunto(s)
Activación de Linfocitos/inmunología , Linfocitos/inmunología , Linfocitos/efectos de la radiación , Neoplasias de la Próstata Resistentes a la Castración/inmunología , Neoplasias de la Próstata Resistentes a la Castración/radioterapia , Radio (Elemento)/administración & dosificación , Anciano , Anciano de 80 o más Años , Humanos , Interferón gamma/inmunología , Metástasis Linfática , Activación de Linfocitos/efectos de la radiación , Masculino , Persona de Mediana Edad , Neoplasias de la Próstata Resistentes a la Castración/secundario , Dosis de Radiación , Radioisótopos/administración & dosificación , Radiofármacos/administración & dosificación , Ganglio Linfático Centinela/inmunología , Ganglio Linfático Centinela/efectos de la radiación , Resultado del Tratamiento
7.
J Autoimmun ; 73: 10-23, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27289166

RESUMEN

The ultraviolet (UV) radiation contained in sunlight is a powerful immune suppressant. While exposure to UV is associated with protection from the development of autoimmune diseases, particularly multiple sclerosis, the precise mechanism by which UV achieves this protection is not currently well understood. Regulatory B cells play an important role in preventing autoimmunity and activation of B cells is a major way in which UV suppresses adaptive immune responses. Whether UV-protection from autoimmunity is mediated by the activation of regulatory B cells has never been considered before. When C57BL/6 mice were exposed to low, physiologically relevant doses of UV, a unique population of B cells was activated in the skin draining lymph nodes. As determined by flow cytometry, CD1d(low)CD5(-)MHC-II(hi)B220(hi) UV-activated B cells expressed significantly higher levels of CD19, CD21/35, CD25, CD210 and CD268 as well as the co-stimulatory molecules CD80, CD86, CD274 and CD275. Experimental autoimmune encephalomyelitis (EAE) in mice immunized with MOG/CFA was reduced by exposure to UV. UV significantly inhibited demyelination and infiltration of inflammatory cells into the spinal cord. Consequently, UV-exposed groups showed elevated IL-10 levels in secondary lymphoid organs, delayed EAE onset, reduced peak EAE score and significantly suppressed overall disease incidence and burden. Importantly, protection from EAE could be adoptively transferred using B cells isolated from UV-exposed, but not unirradiated hosts. Indeed, UV-protection from EAE was dependent on UV activation of lymph node B cells because UV could not protect mice from EAE who were pharmacologically depleted of B cells using antibodies. Thus, UV maintenance of a pool of unique regulatory B cells in peripheral lymph nodes appears to be essential to prevent an autoimmune attack on the central nervous system.


Asunto(s)
Autoinmunidad/efectos de la radiación , Linfocitos B Reguladores/efectos de la radiación , Sistema Nervioso Central/inmunología , Encefalomielitis Autoinmune Experimental/prevención & control , Activación de Linfocitos/efectos de la radiación , Luz Solar , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/inmunología , Antígenos CD/metabolismo , Linfocitos B Reguladores/inmunología , Linfocitos B Reguladores/metabolismo , Sistema Nervioso Central/patología , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/inmunología , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Antígenos de Histocompatibilidad Clase II/metabolismo , Inyecciones Intraperitoneales , Interleucina-10/metabolismo , Interleucina-10/efectos de la radiación , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Ratones , Ratones Endogámicos C57BL , Glicoproteína Mielina-Oligodendrócito/toxicidad
8.
J Immunol ; 192(7): 3101-10, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24600032

RESUMEN

As a side effect of cancer radiotherapy, immune cells receive varying doses of radiation. Whereas high doses of radiation (>10 Gy) can lead to lymphopenia, lower radiation doses (2-4 Gy) represent a valid treatment option in some hematological cancers, triggering clinically relevant immunological changes. Based on our earlier observations, we hypothesized that lower radiation doses have a direct positive effect on T cells. In this study, we show that 0.6-2.4 Gy radiation enhances proliferation and IFN-γ production of PBMC or purified T cells induced by stimulation via the TCR. Radiation with 1.2 Gy also lowered T cell activation threshold and broadened the Th1 cytokine profile. Although radiation alone did not activate T cells, when followed by TCR stimulation, ERK1/2 and Akt phosphorylation increased above that induced by stimulation alone. These changes were followed by an early increase in glucose uptake. Naive (CD45RA(+)) or memory (CD45RA(-)) T cell responses to stimulation were boosted at similar rates by radiation. Whereas increased Ag-specific cytotoxic activity of a CD8(+) T cell line manifested in a 4-h assay (10-20% increase), highly significant (5- to 10-fold) differences in cytokine production were detected in 6-d Ag-stimulation assays of PBMC, probably as a net outcome of death of nonstimulated and enhanced response of Ag-stimulated T cells. T cells from patients receiving pelvic radiation (2.2-2.75 Gy) also displayed increased cytokine production when stimulated in vitro. We report in this study enhanced T cell function induced by synergistic radiation treatment, with potential physiological significance in a wide range of T cell responses.


Asunto(s)
Proliferación Celular/efectos de la radiación , Péptidos/inmunología , Linfocitos T/inmunología , Linfocitos T/efectos de la radiación , Secuencia de Aminoácidos , Línea Celular Tumoral , Células Cultivadas , Citotoxicidad Inmunológica/inmunología , Citotoxicidad Inmunológica/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Epítopos de Linfocito T/inmunología , Quinasas MAP Reguladas por Señal Extracelular/inmunología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Citometría de Flujo , Glucosa/inmunología , Glucosa/farmacocinética , Humanos , Interferón gamma/inmunología , Interferón gamma/metabolismo , Antígenos Comunes de Leucocito/inmunología , Antígenos Comunes de Leucocito/metabolismo , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/efectos de la radiación , Activación de Linfocitos/inmunología , Activación de Linfocitos/efectos de la radiación , Masculino , Fosforilación/inmunología , Fosforilación/efectos de la radiación , Neoplasias de la Próstata/sangre , Neoplasias de la Próstata/radioterapia , Proteínas Proto-Oncogénicas c-akt/inmunología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/metabolismo
9.
Cytotherapy ; 17(2): 152-62, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25453724

RESUMEN

BACKGROUND AIMS: The discovery of regenerative and immunosuppressive capacities of mesenchymal stromal cells (MSCs) raises hope for patients with tissue-damaging or severe, treatment-refractory autoimmune disorders. We previously presented a method to expand human MSCs in a bioreactor under standardized Good Manufacturing Practice conditions. Now we characterized the impact of critical treatment conditions on MSCs with respect to immunosuppressive capabilities and proliferation. METHODS: MSC proliferation and survival after γ irradiation were determined by 5-carboxyfluorescein diacetate N-succinimidyl ester and annexinV/4',6-diamidino-2-phenylindole (DAPI) staining, respectively. T-cell proliferation assays were used to assess the effect of γ irradiation, passaging, cryopreservation, post-thaw equilibration time and hypoxia on T-cell suppressive capacities of MSCs. Quantitative polymerase chain reaction and ß-galactosidase staining served as tools to investigate differences between immunosuppressive and non-immunosuppressive MSCs. RESULTS: γ irradiation of MSCs abrogated their proliferation while vitality and T-cell inhibitory capacity were preserved. Passaging and long cryopreservation time decreased the T-cell suppressive function of MSCs, and postthaw equilibration time of 5 days restored this capability. Hypoxic culture markedly increased MSC proliferation without affecting their T-cell-suppressive capacity and phenotype. Furthermore, T-cell suppressive MSCs showed higher CXCL12 expression and less ß-galactosidase staining than non-suppressive MSCs. DISCUSSION: We demonstrate that γ irradiation is an effective strategy to abrogate MSC proliferation without impairing the cells' immunosuppressive function. Hypoxia significantly enhanced MSC expansion, allowing for transplantation of MSCs with low passage number. In summary, our optimized MSC expansion protocol successfully addressed the issues of safety and preservation of immunosuppressive MSC function after ex vivo expansion for therapeutic purposes.


Asunto(s)
Hipoxia de la Célula/fisiología , Proliferación Celular/efectos de la radiación , Rayos gamma/efectos adversos , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/efectos de la radiación , Adulto , Células Cultivadas , Quimiocina CXCL12/biosíntesis , Criopreservación , Fluoresceínas , Humanos , Terapia de Inmunosupresión/métodos , Inmunosupresores/efectos de la radiación , Activación de Linfocitos/inmunología , Activación de Linfocitos/efectos de la radiación , Células Madre Mesenquimatosas/citología , Persona de Mediana Edad , Linfocitos T/inmunología , beta-Galactosidasa/metabolismo
10.
Photochem Photobiol Sci ; 14(10): 1903-9, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26295729

RESUMEN

Transfusion of allogeneic white blood cells (WBCs) may cause adverse reactions in immunocompromised recipients, including transfusion-associated graft-versus-host disease (TA-GVHD), which is often fatal and incurable. In this study, the in vitro effect of methylene blue with visible light (MB + L) treatment on lymphocyte proliferation and cytokine production was measured to investigate whether MB + L can be used to prevent immune reactions that result from transfused lymphocytes. WBCs and 3 µM of MB were mixed and transferred into medical PVC bags, which were then exposed to visible light. Gamma irradiation was conducted as a parallel positive control. The cells without treatment were used as untreated group. All the groups were tested for the ability of cell proliferation and cytokine production upon stimulation. After incubation with mitogen phytohemagglutinin (PHA) or plate-bound anti-CD3 plus anti-CD28, the proliferation of MB + L/gamma-irradiation treated lymphocytes was significantly inhibited (P < 0.01) as compared to the untreated ones; the proliferation inhibitive rate of the MB + L group was even higher than that of gamma-irradiated cells (73.77% ± 28.75% vs. 44.72% ± 38.20%). MB + L treated cells incubated up to 7 days with PHA also showed no significant proliferation. The levels of TNF-α, IFN-γ, IL-6, IL-8, IL-10 and IL-1ß present in the supernatant of MB + L treated lymphocytes upon stimulation were significantly lower than those of untreated lymphocytes. These results demonstrated that MB + L treatment functionally and irreversibly inactivated lymphocytes by inhibiting lymphocyte proliferation and the production of cytokines. MB + L treatment might be a promising method for the prevention of adverse immune responses caused by WBCs.


Asunto(s)
Luz , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/efectos de la radiación , Linfocitos/efectos de los fármacos , Linfocitos/efectos de la radiación , Azul de Metileno/farmacología , Fármacos Fotosensibilizantes/farmacología , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Citocinas/biosíntesis , Humanos , Linfocitos/inmunología , Linfocitos/metabolismo
11.
Int J Mol Sci ; 16(12): 28334-46, 2015 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-26633364

RESUMEN

Carbon ion radiation is a promising treatment for brain cancer; however, the immune system involved long-term systemic effects evoke a concern of complementary and alternative therapies in clinical treatment. To clarify radiotherapy caused fundamental changes in peripheral immune system, examinations were performed based on established models in vitro and in vivo. We found that brain-localized carbon ion radiation of neural cells induced complex changes in the peripheral blood, thymus, and spleen at one, two, and three months after its application. Atrophy, apoptosis, and abnormal T-cell distributions were observed in rats receiving a single high dose of radiation. Radiation downregulated the expression of proteins involved in T-cell development at the transcriptional level and increased the proportion of CD3⁺CD4(-)CD8⁺ T-cells in the thymus and the proportion of CD3⁺CD4⁺CD8(-) T-cells in the spleen. These data show that brain irradiation severely affects the peripheral immune system, even at relatively long times after irradiation. In addition, they provide valuable information that will implement the design of biological-based strategies that will aid brain cancer patients suffering from the long-term side effects of radiation.


Asunto(s)
Carbono , Iones , Sistema Nervioso/inmunología , Sistema Nervioso/efectos de la radiación , Traumatismos Experimentales por Radiación/inmunología , Radiación Ionizante , Animales , Apoptosis/inmunología , Apoptosis/efectos de la radiación , Biomarcadores , Encéfalo/inmunología , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/efectos de la radiación , Línea Celular , Modelos Animales de Enfermedad , Humanos , Terapia de Inmunosupresión , Activación de Linfocitos/inmunología , Activación de Linfocitos/efectos de la radiación , Masculino , Traumatismos Experimentales por Radiación/metabolismo , Traumatismos Experimentales por Radiación/patología , Ratas , Bazo/inmunología , Bazo/metabolismo , Bazo/patología , Bazo/efectos de la radiación , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/patología , Linfocitos T/efectos de la radiación , Timo/inmunología , Timo/patología , Timo/efectos de la radiación
12.
Transfus Apher Sci ; 50(3): 330-9, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24837416

RESUMEN

Extracorporeal Photochemotherapy (ECP) consists in illumination of the patient's leukocytes in the presence of 8-Methoxy Psoralen (8-MOP) and its reinjection to the same patient. ECP is responsible for many cellular events, the most important being the induction of cell apoptosis. Apoptosis appears first in lymphocytes and activated lymphocytes (allo or auto) which are more sensitive and undergo faster apoptosis rather than other cells. Monocytes develop apoptosis later. The injection of apoptotic cells induces tolerance in patients with graft versus host disease (GvHD) and acute heart or lung graft rejection. In these patients, phagocytosis of apoptotic cells by antigen-presenting cells (APCs) and in particular dendritic cells is responsible for a shift from Th1 to Th2 immune response, an increase in anti-inflammatory cytokines such as interleukine 10 (IL-10) and Tumor Growth Factor Beta (TGF-ß), a decrease in pro-inflammatory cytokines and finally, for the proliferation of regulatory cells. Among CD4/CD25 positive cells, only CD4(+)CD25(hi) are T-regulatory cells (T-regs). One subpopulation of T-regs produces IL-10 and inhibits Th1 CD4 cells, whereas other populations act as suppressors and inhibit the cytotoxic T-cells responsible for organ rejection and GvHD in an antigen specific fashion. It is not clear why the injection of early apoptotic cells induces tolerance in GvHD and organ graft rejection, but in Sézary syndrome, it induces up-regulation of anti-tumor immune response. Immune response modulation (up- or down-regulation) after ECP depends on many factors: early apoptotic cell injection; anti-inflammatory environment; impaired function of dendritic cells; dendritic type 2 cell dominance, lead to immune tolerance, whereas late apoptotic or necrotic cell injection and pro-inflammatory cytokines enhance immune response. Therefore, immune response to ECP depends on various factors responsible for the diversity of its mode of action in different diseases and further investigations are required.


Asunto(s)
Inmunidad Celular , Metoxaleno/uso terapéutico , Fotoféresis/métodos , Fármacos Fotosensibilizantes/uso terapéutico , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Células Dendríticas/inmunología , Células Dendríticas/patología , Rechazo de Injerto/inmunología , Rechazo de Injerto/patología , Rechazo de Injerto/terapia , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/patología , Enfermedad Injerto contra Huésped/terapia , Humanos , Inmunidad Celular/efectos de los fármacos , Inmunidad Celular/efectos de la radiación , Interleucina-10/inmunología , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/efectos de la radiación , Monocitos/inmunología , Monocitos/patología , Trasplante de Órganos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología , Células TH1/inmunología , Células TH1/patología , Células Th2/inmunología , Células Th2/patología , Factor de Crecimiento Transformador beta/inmunología
13.
Adv Exp Med Biol ; 810: 182-91, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25207366

RESUMEN

Carcinogenesis involves multiple mechanisms that disturb genomic integrity and encourage abnormal proliferation. The immune system plays an integral role in maintaining homeostasis and these mechanisms may arrest or enhance dysplasia. There exists a large body of evidence from organ transplantation literature supporting the significance of the immune suppression in the development of skin cancer. Nonmelanoma skin cancers are the most frequent neoplasms after organ transplantation, with organ transplant recipients having a 65-fold increase in squamous cell carcinoma incidence and 10-fold increase in basal cell carcinoma incidence. Similarly, UV-radiation (UVR) induced immunosuppression is correlated with the development of cutaneous malignancies in a dose-dependent manner. This was first shown several decades ago by Margaret Kripke, when transplanted tumors were rejected in mice with competent immune systems, but grew unchecked in immunosuppressed specimens. After UV exposure, chromophores initiate a cascade that leads to immunosuppression via derangement of Langerhans cells' antigen-presenting capacity. UV-irradiated Langerhans cells present antigens to Th2 cells, but fail to stimulate Th1 cells. A subset of T regulatory cells, specific for the antigen encountered after UVR, is also stimulated to proliferate. In general UV irradiation leads to a greater number of T regulatory cells and fewer effector T cells in the skin, shiftingthe balance from T-cell-mediated immunity to immunosuppression. These regulatory cells have the phenotype CD4+, CD25+, Foxp3+, CTLA-4+. These and many other changes in local immunity lead to a suppressed immune state, which allow for skin cancer development.


Asunto(s)
Carcinoma Basocelular/inmunología , Carcinoma de Células Escamosas/inmunología , Sistema Inmunológico/efectos de la radiación , Neoplasias Cutáneas/inmunología , Subgrupos de Linfocitos T/efectos de la radiación , Carcinoma Basocelular/epidemiología , Carcinoma Basocelular/patología , Carcinoma de Células Escamosas/epidemiología , Carcinoma de Células Escamosas/patología , Proliferación Celular/efectos de la radiación , Humanos , Inmunofenotipificación , Terapia de Inmunosupresión , Incidencia , Activación de Linfocitos/efectos de la radiación , Trasplante de Órganos , Piel/inmunología , Piel/patología , Neoplasias Cutáneas/epidemiología , Neoplasias Cutáneas/patología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/patología , Rayos Ultravioleta
14.
Exp Dermatol ; 22(8): 558-60, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23879817

RESUMEN

Blue light is a UV-free irradiation suitable for treating chronic skin inflammation, for example, atopic dermatitis, psoriasis, and hand- and foot eczema. However, a better understanding of the mode of action is still missing. For this reason, we investigated whether dendritic cells (DC) are directly affected by blue light irradiation in vitro. Here, we report that irradiation neither induced apoptosis nor maturation of monocyte-derived and myeloid DC. However, subsequent DC maturation upon LPS/IFNγ stimulation was impaired in a dose-dependent manner as assessed by maturation markers and cytokine release. Moreover, the potential of this DC to induce cytokine secretion from allogeneic CD4 T cells was reduced. In conclusion, unlike UV irradiation, blue light irradiation at high and low doses only resulted in impaired DC maturation upon activation and a reduced subsequent stimulatory capacity in allogeneic MLRs with strongest effects at higher doses.


Asunto(s)
Células Dendríticas/citología , Células Dendríticas/efectos de la radiación , Luz , Activación de Linfocitos/efectos de la radiación , Rayos Ultravioleta , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/efectos de la radiación , Separación Celular , Técnicas de Cocultivo , Citocinas/metabolismo , Células Dendríticas/inmunología , Humanos , Terapia de Inmunosupresión , Inflamación , Interferón gamma/farmacología , Lipopolisacáridos/farmacología , Activación de Linfocitos/inmunología
15.
J Immunol ; 187(3): 1142-9, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21697461

RESUMEN

Epidermal Langerhans cells (LC) are potent APCs surveying the skin. They are crucial regulators of T cell activation in the context of inflammatory skin disease and graft-versus-host disease (GVHD). In contrast to other dendritic cell subtypes, murine LC are able to reconstitute after local depletion without the need of peripheral blood-derived precursors. In this study, we introduce an experimental model of human skin grafted to NOD-SCID IL2Rγ(null) mice. In this model, we demonstrate that xenografting leads to the transient loss of LC from the human skin grafts. Despite the lack of a human hematopoietic system, human LC repopulated the xenografts 6 to 9 wk after transplantation. By staining of LC with the proliferation marker Ki67, we show that one third of the replenishing LC exhibit proliferative activity in vivo. We further used the skin xenograft as an in vivo model for human GVHD. HLA-disparate third-party T cells stimulated with skin donor-derived dendritic cells were injected intravenously into NOD-SCID IL2Rγ(null) mice that had been transplanted with human skin. The application of alloreactive T cells led to erythema and was associated with histological signs of GVHD limited to the transplanted human skin. The inflammation also led to the depletion of LC from the epidermis. In summary, we provide evidence that human LC are able to repopulate the skin independent of blood-derived precursor cells and that this at least partly relates to their proliferative capacity. Our data also propose xeno-transplantation of human skin as a model system for studying the role of skin dendritic cells in the efferent arm of GVHD.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Epidermis/inmunología , Células de Langerhans/inmunología , Activación de Linfocitos/inmunología , Trasplante de Piel/inmunología , Trasplante Heterólogo/inmunología , Animales , Linfocitos T CD8-positivos/patología , Linfocitos T CD8-positivos/trasplante , Muerte Celular/inmunología , Muerte Celular/efectos de la radiación , Diferenciación Celular/inmunología , Diferenciación Celular/efectos de la radiación , Movimiento Celular/inmunología , Movimiento Celular/efectos de la radiación , Proliferación Celular/efectos de la radiación , Células Cultivadas , Modelos Animales de Enfermedad , Epidermis/patología , Epidermis/trasplante , Femenino , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/patología , Humanos , Células de Langerhans/patología , Células de Langerhans/trasplante , Activación de Linfocitos/efectos de la radiación , Masculino , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Ratones Transgénicos , Trasplante de Piel/patología , Trasplante Heterólogo/patología
16.
Bull Exp Biol Med ; 155(5): 689-91, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24288741
17.
J Immunol ; 185(8): 4633-40, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20844203

RESUMEN

Ultraviolet exposure alters the morphology and function of epidermal Langerhans cells (LCs), which play a role in UV-induced immune suppression. It is generally believed that UV exposure triggers the migration of immature LCs from the skin to the draining lymph nodes (LNs), where they induce tolerance. However, because most of the previous studies employed in vitro UV-irradiated LCs, the data generated may not adequately reflect what is happening in vivo. In this study, we isolated migrating LCs from the LNs of UV-irradiated mice and studied their function. We found prolonged LC survival in the LNs of UV-irradiated mice. LCs were necessary for UV-induced immune suppression because no immune suppression was observed in LC-deficient mice. Transferring LCs from UV-irradiated mice into normal recipient animals transferred immune suppression and induced tolerance. We found that LCs colocalized with LN NKT cells. No immune suppression was observed when LCs were transferred from UV-irradiated mice into NKT cell-deficient mice. NKT cells isolated from the LNs of UV-irradiated mice secreted significantly more IL-4 than NKT cells isolated from nonirradiated controls. Injecting the wild-type mice with anti-IL-4 blocked the induction of immune suppression. Our findings indicate that UV exposure activates the migration of mature LC to the skin draining LNs, where they induce immune regulation in vivo by activating NKT cells.


Asunto(s)
Tolerancia Inmunológica/inmunología , Células de Langerhans/inmunología , Activación de Linfocitos/inmunología , Células T Asesinas Naturales/inmunología , Animales , Quimiotaxis de Leucocito/inmunología , Quimiotaxis de Leucocito/efectos de la radiación , Técnica del Anticuerpo Fluorescente , Tolerancia Inmunológica/efectos de la radiación , Células de Langerhans/efectos de la radiación , Activación de Linfocitos/efectos de la radiación , Ratones , Células T Asesinas Naturales/efectos de la radiación , Rayos Ultravioleta/efectos adversos
18.
J Immunol ; 184(6): 2751-5, 2010 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-20164430

RESUMEN

Polyinosinic:polycytidylic acid (poly I:C), a synthetic analog of double-stranded viral RNA, serves as a potent adjuvant for vaccination against soluble proteins, pathogens, and tumors. Poly I:C is sensed by both TLR3 in the endosomes and melanoma differentiation-associated protein 5 (MDA5) in the cytoplasm. Although it is known that TLR3 is required for cross-priming of CD8 T cells specific for viral Ags, the role of MDA5 in inducing CD8 T cell responses is still unclear. In this study, we demonstrate that in mice lacking MDA5, the majority of CD8 T cells do not survive after primary immunization with poly I:C and Ag, impairing memory response to subsequent Ag challenge. Furthermore, bone marrow chimera experiments revealed that MDA5 expression in radiation-resistant stromal cells, but not in radiation-sensitive hematopoietic cells, is essential for establishing CD8 T cell memory. We conclude that MDA5 and TLR3 mediate substantially distinct yet complementary functions during poly I:C-mediated activation of Ag-specific CD8 T cell responses.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , ARN Helicasas DEAD-box/fisiología , Regulación Neoplásica de la Expresión Génica/inmunología , Memoria Inmunológica , Activación de Linfocitos/inmunología , Poli I-C/farmacología , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/efectos de la radiación , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/efectos de la radiación , ARN Helicasas DEAD-box/biosíntesis , ARN Helicasas DEAD-box/deficiencia , ARN Helicasas DEAD-box/genética , Epítopos de Linfocito T/inmunología , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/efectos de la radiación , Helicasa Inducida por Interferón IFIH1 , Activación de Linfocitos/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Quimera por Radiación , Células del Estroma/inmunología , Células del Estroma/metabolismo , Células del Estroma/efectos de la radiación , Receptor Toll-Like 3/deficiencia , Receptor Toll-Like 3/genética , Receptor Toll-Like 3/fisiología
19.
J Immunol ; 184(10): 5670-7, 2010 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-20400709

RESUMEN

The mechanism underlying the local UVB-induced immunosuppression is a central issue to be clarified in photoimmunology. There have been reported a considerable number of cells and factors that participate in the sensitization phase-dependent suppression, including Langerhans cells (LCs), regulatory T cells, IL-10, and TNF-alpha. The recent important finding that LC-depleted mice rather exhibit enhanced contact hypersensitivity responses urged us to re-evaluate the role of LCs along with dermal dendritic cells (dDCs) in the mechanism of UVB-induced immunosuppression. We studied the surface expression of OX40 ligand (OX40L) and the intracellular expression of IL-10 in LCs and dDCs from UVB-irradiated (300 mJ/cm(2)) skin of BALB/c mice and those migrating to the regional lymph nodes from UVB-irradiated, hapten-painted mice. In epidermal and dermal cell suspensions prepared from the UVB-irradiated skin, LCs expressed OX40L as well as CD86 and produced IL-10 at a higher level than Langerin(-) dDCs. The UVB-induced immunosuppression was attenuated by the administration of IL-10-neutralizing or OX40L-blocking Abs. In mice whose UVB-irradiated, hapten-painted skin was dissected 1 d after hapten application, the contact hypersensitivity response was restored, because this treatment allowed dDCs but not LCs to migrate to the draining lymph nodes. Moreover, LC-depleted mice by using Langerin-diphtheria toxin receptor-knocked-in mice showed impaired UVB-induced immunosuppression. These results suggest that IL-10-producing and OX40L-expressing LCs in the UVB-exposed skin are mandatory for the induction of Ag-specific regulatory T cells.


Asunto(s)
Diferenciación Celular/inmunología , Terapia de Inmunosupresión , Interleucina-10/biosíntesis , Células de Langerhans/inmunología , Glicoproteínas de Membrana/biosíntesis , Factores de Necrosis Tumoral/biosíntesis , Rayos Ultravioleta , Animales , Antígenos de Superficie/genética , Diferenciación Celular/efectos de la radiación , Femenino , Técnicas de Sustitución del Gen , Factor de Crecimiento Similar a EGF de Unión a Heparina , Terapia de Inmunosupresión/métodos , Péptidos y Proteínas de Señalización Intercelular/genética , Interleucina-10/efectos de la radiación , Células de Langerhans/metabolismo , Células de Langerhans/efectos de la radiación , Lectinas Tipo C/genética , Ligandos , Activación de Linfocitos/inmunología , Activación de Linfocitos/efectos de la radiación , Lectinas de Unión a Manosa/genética , Glicoproteínas de Membrana/efectos de la radiación , Ratones , Ratones Endogámicos BALB C , Ligando OX40 , Técnicas de Cultivo de Órganos , Piel/citología , Piel/inmunología , Piel/efectos de la radiación , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/efectos de la radiación , Factores de Necrosis Tumoral/efectos de la radiación
20.
Proc Natl Acad Sci U S A ; 106(29): 11966-71, 2009 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-19570996

RESUMEN

Deletion or mutation of the SAP gene is associated with the X-linked lymphoproliferative disease (XLP) that is characterized by extreme sensitivity to Epstein-Barr virus (EBV). Primary infection of the affected individuals leads to serious, sometimes fatal infectious mononucleosis (IM) and proneness to lymphoma. Our present results revealed a proapoptotic function of SAP by which it contributes to the maintenance of T-cell homeostasis and to the elimination of potentially dangerous DNA-damaged cells. Therefore, the loss of this function could be responsible for the uncontrolled T-cell proliferation in fatal IM and for the generation of lymphomas. We show now the role of SAP in apoptosis in T and B lymphocyte-derived lines. Among the clones of T-ALL line, the ones with higher SAP levels succumbed more promptly to activation induced cell death (AICD). Importantly, introduction of SAP expression into lymphoblastoid cell lines (LCL) established from XLP patients led to elevated apoptotic response to DNA damage. Similar results were obtained in the osteosarcoma line, Saos-2. We have shown that the anti-apoptotic protein VCP (valosin-containing protein) binds to SAP, suggesting that it could be instrumental in the enhanced apoptotic response modulated by SAP.


Asunto(s)
Apoptosis , Genes Ligados a X , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Trastornos Linfoproliferativos/metabolismo , Trastornos Linfoproliferativos/patología , Adenosina Trifosfatasas/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Biomarcadores/metabolismo , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Membrana Celular/efectos de la radiación , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Células Clonales , Daño del ADN , Fase G2/efectos de los fármacos , Fase G2/efectos de la radiación , Rayos gamma , Humanos , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/efectos de la radiación , Mitosis/efectos de los fármacos , Mitosis/efectos de la radiación , Fitohemaglutininas/farmacología , Unión Proteica/efectos de los fármacos , Unión Proteica/efectos de la radiación , Retroviridae , Proteína Asociada a la Molécula de Señalización de la Activación Linfocitaria , Linfocitos T/citología , Linfocitos T/efectos de los fármacos , Transducción Genética , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/efectos de la radiación , Proteína que Contiene Valosina , Proteína bcl-X/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda