Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 192
Filtrar
1.
Cell Physiol Biochem ; 58(5): 477-490, 2024 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-39248192

RESUMEN

BACKGROUND/AIMS: Tumor response to radiation is thought to depend on the direct killing of tumor cells. Our laboratory has called this into question. Firstly, we showed that the biology of the host, specifically the endothelial expression of acid sphingomyelinase (ASMase), was critical in determining tumor radiocurability. Secondly, we have shown that the immune system can enhance radiation response by allowing a complete tumor control in hemi-irradiated tumors. In this paper, we focus on the integration of these two findings. METHODS: We used Lewis Lung Carcinoma (LLC) cells, injected in the flank of either: (i) ASMase knockout or (ii) WT of matched background (sv129xBl/6) or (iii) C57Bl/6 mice. Radiation therapy (RT) was delivered to 50% or 100% of the LLC tumor volume. Tumor response, immune infiltration (CD8+ T cells), ICAM-1, and STING activation were measured. Radiotherapy was also combined with methyl-cyclodextrin, to inhibit the ASMase-mediated formation of ceramide-enriched lipid rafts. RESULTS: We recapitulated our previous finding, namely that tumor hemi-irradiation was sufficient for tumor control in the LLC/C57Bl/6 model. However, in ASMase KO mice hemi-irradiation was ineffective. Likewise, pharmacological inhibition of ASMase significantly reduced the tumor response to hemi-irradiation. Further, we demonstrated elevated ICAM-1 expression, increased levels of CD8+ T cells, ICAM-1, and STING activation in tumors growing in C57Bl/6 mice, as well as the ASMase WT strain. However, no such changes were seen in tumors growing in ASMase KO mice. CONCLUSION: ASMase and ceramide generation are necessary to mediate a radiation-induced anti-tumor immune response via STING activation.


Asunto(s)
Linfocitos T CD8-positivos , Carcinoma Pulmonar de Lewis , Molécula 1 de Adhesión Intercelular , Ratones Endogámicos C57BL , Ratones Noqueados , Esfingomielina Fosfodiesterasa , Animales , Esfingomielina Fosfodiesterasa/metabolismo , Esfingomielina Fosfodiesterasa/genética , Carcinoma Pulmonar de Lewis/inmunología , Carcinoma Pulmonar de Lewis/patología , Carcinoma Pulmonar de Lewis/radioterapia , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/metabolismo , Ratones , Molécula 1 de Adhesión Intercelular/metabolismo , Molécula 1 de Adhesión Intercelular/genética , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/efectos de la radiación , Linfocitos T CD8-positivos/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Ceramidas/metabolismo , Microdominios de Membrana/metabolismo , Línea Celular Tumoral
2.
J Surg Oncol ; 123(1): 117-126, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33084061

RESUMEN

BACKGROUND AND OBJECTIVES: Limited literature exists examining the immune microenvironment in liposarcoma, particularly with regard to the impact of radiotherapy. A major problem is the lack of scoring system for the tumour-infiltrating lymphocytes (TILs) in sarcoma. This study aims to describe the immune environment pre- and postradiotherapy and identify the optimal immune infiltrate scoring system for sarcoma. METHODS: Thirty-nine paired tissue samples (pre- and postradiotherapy) from patients with liposarcoma were scored by two pathologists for TILs using pre-existing systems (for breast cancer and melanoma) and compared for interobserver reliability. Immunohistochemical staining was performed for various immune markers. RESULTS: The TIL scoring system for breast cancer yielded perfect agreement (κ = 1.000). 21% of patients had increased TILs after radiotherapy, 87.5% of whom had dedifferentiated liposarcoma. Immune suppressor expression was increased frequently after radiotherapy (CD68 increased in 59.4%, PD-L1 increased in 25%). Immune effector expression (CD8) was unchanged in 84.4%. CONCLUSIONS: Breast cancer TIL scoring is reproducible in liposarcoma and has high interobserver reliability. Radiotherapy was observed to have a limited impact on immune effectors but seemed to have more impact in upregulating immune suppressors, suggesting radiotherapy may contribute to disease control through immunomodulatory effects. Dedifferentiated liposarcoma represents a uniquely responsive subtype.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Liposarcoma/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Recurrencia Local de Neoplasia/inmunología , Radioterapia/métodos , Microambiente Tumoral/inmunología , Adolescente , Adulto , Anciano , Linfocitos T CD8-positivos/efectos de la radiación , Femenino , Estudios de Seguimiento , Humanos , Liposarcoma/patología , Liposarcoma/radioterapia , Linfocitos Infiltrantes de Tumor/efectos de la radiación , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/patología , Recurrencia Local de Neoplasia/radioterapia , Pronóstico , Estudios Retrospectivos , Microambiente Tumoral/efectos de la radiación , Adulto Joven
3.
Int J Mol Sci ; 22(4)2021 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-33669885

RESUMEN

The low overall survival rate of patients with pancreatic cancer has driven research to seek a new therapeutic protocol. Radiotherapy (RT) is frequently an option in the neoadjuvant or palliative settings for pancreatic cancer treatment. This study explored the effect of RT protocols on the tumor microenvironment (TME) and their consequent impact on anti-programmed cell death ligand-1 (PD-L1) therapy. Using a murine orthotopic pancreatic tumor model, UN-KC-6141, RT-disturbed TME was examined by immunohistochemical staining. The results showed that ablative RT is more effective than fractionated RT at recruiting T cells. On the other hand, fractionated RT induces more myeloid-derived suppressor cell infiltration than ablative RT. The RT-disturbed TME presents a higher perfusion rate per vessel. The increase in vessel perfusion is associated with a higher amount of anti-PD-L1 antibody being delivered to the tumor. Animal survival is increased by anti-PD-L1 therapy after ablative RT, with 67% of treated animals surviving more than 30 days after tumor inoculation compared to a median survival time of 16.5 days for the control group. Splenocytes isolated from surviving animals were specifically cytotoxic for UN-KC-6141 cells. We conclude that the ablative RT-induced TME is more suited than conventional RT-induced TME to combination therapy with immune checkpoint blockade.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/radioterapia , Microambiente Tumoral , Animales , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/efectos de la radiación , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Inhibidores de Puntos de Control Inmunológico/farmacología , Ratones Endogámicos C57BL , Neoplasias Pancreáticas/irrigación sanguínea , Neoplasias Pancreáticas/inmunología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/efectos de la radiación
4.
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
5.
J Pathol ; 247(5): 606-614, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30632153

RESUMEN

Historically, our understanding of the cytotoxicity of radiation has centred on tumour cell-autonomous mechanisms of cell death. Here, tumour cell death occurs when a threshold number of radiation-induced non-reparable double-stranded DNA breaks is exceeded. However, in recent years, the importance of immune mechanisms of cell death has been increasingly recognised, as well as the impact of radiotherapy on non-malignant cellular components of the tumour microenvironment. Conserved antiviral pathways that detect foreign nucleic acid in the cytosol and drive downstream interferon (IFN) responses via the cyclic guanosine monophosphate-adenosine monophosphate synthase/stimulator of IFN genes (cGAS/STING) pathway are key components of the immune response to radiation-induced DNA damage. In preclinical models, acute induction of a type 1 IFN response is important for both direct and abscopal tumour responses to radiation. Inhibitors of the DNA damage response show promise in augmenting this inflammatory IFN response. However, a substantial proportion of tumours show chronic IFN signalling prior to radiotherapy, which paradoxically drives immunosuppression. This chronic IFN signalling leads to treatment resistance, and heterotypic interactions between stromal fibroblasts and tumour cells contribute to an aggressive tumour phenotype. The effect of radiotherapy on myeloid cell populations, particularly tumour-associated macrophages, has an additional impact on the immune tumour microenvironment. It is not yet clear how the above preclinical findings translate into a human context. Human tumours show greater intratumoural genomic heterogeneity and more variable levels of chromosomal instability than experimental murine models. High-quality translational studies of immunological changes occurring during radiotherapy that incorporate intrinsic tumour biology will enable a better understanding of the immunological consequences of radiation-induced DNA damage in patients. Copyright © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Daño del ADN/efectos de la radiación , Animales , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/efectos de la radiación , Fibroblastos Asociados al Cáncer/inmunología , Inestabilidad Cromosómica/genética , Inestabilidad Cromosómica/inmunología , Terapia Combinada , Daño del ADN/inmunología , Modelos Animales de Enfermedad , Humanos , Tolerancia Inmunológica/inmunología , Factores Inmunológicos/uso terapéutico , Interferón Tipo I/biosíntesis , Interferón Tipo I/efectos de la radiación , Ratones , Células Mieloides/inmunología , Células Mieloides/efectos de la radiación , Neoplasias/inmunología , Neoplasias/radioterapia , Dosis de Radiación , Transducción de Señal/inmunología
6.
Cancer Sci ; 109(4): 956-965, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29465830

RESUMEN

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


Asunto(s)
Carcinoma Pulmonar de Lewis/radioterapia , Proliferación Celular/efectos de la radiación , Receptor Toll-Like 3/metabolismo , Animales , Antígenos de Neoplasias/metabolismo , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/efectos de la radiación , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Carcinoma Pulmonar de Lewis/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Terapia Combinada/métodos , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Células Dendríticas/efectos de la radiación , Modelos Animales de Enfermedad , Inmunoterapia Adoptiva/métodos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , Poli I-C/farmacología , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/metabolismo , Linfocitos T Citotóxicos/efectos de la radiación
7.
Bioelectromagnetics ; 39(1): 68-76, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29171038

RESUMEN

We investigated whether exposure to the 915 MHz radiofrequency identification (RFID) signal affected circulating blood cells in rats. Sprague-Dawley rats were exposed to RFID at a whole-body specific absorption rate of 2 W/kg for 8 h per day, 5 days per week, for 2 weeks. Complete blood counts were performed after RFID exposure, and the CD4+ /CD8+ ratio was determined by flow cytometry. The number of red blood cells (RBCs) and the values of hemoglobin, hematocrit, and RBC indices were increased in the RFID-exposed group compared with those in the cage-control and sham-exposed groups (P < 0.05). However, the RBCs and platelet numbers were within normal physiologic response ranges. The number of white blood cells, including lymphocytes, was decreased in RFID-exposed rats. However, there was no statistically significant difference between the sham-exposed and RFID-exposed groups in terms of T-cell counts or CD4+ /CD8+ ratio (P > 0.05). Although the number of circulating blood cells was significantly altered by RFID exposure at a whole-body specific absorption rate of 2 W/kg for 2 weeks, these changes do not necessarily indicate that RFID exposure is harmful, as they were within the normal physiological response range. Bioelectromagnetics. 39:68-76, 2018. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Células Sanguíneas/efectos de la radiación , Campos Electromagnéticos/efectos adversos , Dispositivo de Identificación por Radiofrecuencia , Animales , Células Sanguíneas/citología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/efectos de la radiación , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/efectos de la radiación , Recuento de Células , Eritrocitos/citología , Eritrocitos/efectos de la radiación , Masculino , Ratas , Ratas Sprague-Dawley , Irradiación Corporal Total/efectos adversos
8.
Am J Dermatopathol ; 40(10): 727-735, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30188378

RESUMEN

Hypopigmented interface T-cell dyscrasia (HITCD) is a distinct form of lymphoid dyscrasia that may progress to hypopigmented mycosis fungoides (HMF). We compared both diseases as regards their CD4/CD8 phenotype and expression of granzyme B and tumor necrosis factor-alpha (TNF-α) and how these are affected by narrow-band UVB (nb-UVB). The study included 11 patients with HITCD and 9 patients with HMF. They received nb-UVB thrice weekly until complete repigmentation or a maximum of 48 sessions. Pretreatment and posttreatment biopsies were stained using anti CD4, CD8, TNF-α, and granzyme B monoclonal antibodies. Epidermal lymphocytes were CD8 predominant in 54.5% and 66.7% of HITCD and HMF cases, respectively, whereas dermal lymphocytes were CD4 predominant in 63.6% and 66.7%, respectively. Significantly, more dermal infiltrate was encountered in HMF (P = 0.041). In both diseases, granzyme B was only expressed in the dermis, whereas TNF-α was expressed both in the epidermis and dermis. No difference existed as regards the number of sessions needed to achieve repigmentation or cumulative nb-UVB dose reached at end of study. (P > 0.05). Narrow-band UVB significantly reduced only the epidermal lymphocytes in both diseases (P ≤ 0.05) with their complete disappearance in 8 (72.7%) HITCD and 6 (66.7%) HMF cases. In both diseases, nb-UVB did not affect granzyme B or TNF-α expression (P > 0.05). In conclusion, both diseases share the same phenotype, with HITCD being a milder form of T-cell dysfunction. In both diseases, epidermal lymphocytes are mainly CD8-exhausted cells lacking cytotoxicity, whereas dermal cells are mostly reactive cells exerting antitumor cytotoxicity. Tumor necrosis factor-alpha mediates hypopigmentation in both diseases and prevents disease progression. Repigmentation after nb-UVB in both diseases occurs before and independently from disappearance of the dermal infiltrate.


Asunto(s)
Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/patología , Hipopigmentación/patología , Trastornos Inmunoproliferativos/patología , Micosis Fungoide/patología , Neoplasias Cutáneas/patología , Pigmentación de la Piel , Piel/patología , Adolescente , Adulto , Biopsia , Linfocitos T CD4-Positivos/química , Linfocitos T CD4-Positivos/efectos de la radiación , Linfocitos T CD8-positivos/química , Linfocitos T CD8-positivos/efectos de la radiación , Niño , Estudios Transversales , Femenino , Granzimas/análisis , Humanos , Hipopigmentación/metabolismo , Hipopigmentación/radioterapia , Inmunohistoquímica , Trastornos Inmunoproliferativos/metabolismo , Trastornos Inmunoproliferativos/radioterapia , Masculino , Persona de Mediana Edad , Micosis Fungoide/química , Micosis Fungoide/radioterapia , Fenotipo , Piel/química , Piel/efectos de la radiación , Neoplasias Cutáneas/química , Neoplasias Cutáneas/radioterapia , Pigmentación de la Piel/efectos de la radiación , Resultado del Tratamiento , Factor de Necrosis Tumoral alfa/análisis , Terapia Ultravioleta , Adulto Joven
9.
Future Oncol ; 13(13): 1173-1180, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28498036

RESUMEN

This study aimed to detect the effect of combination radiotherapy and cantharidin on lung cancer growth. We found that combination therapy with radiotherapy and cantharidin was more effective in inhibiting the tumor growth than radiotherapy or cantharidin alone. It decreased the percentage of CD4+ Tregs and enhanced the percentage of CD8+ T cells, CD4+ Teff cells when comparing to that of single treatment. Combination therapy promoted a great increase in double producing CD8+ T cells and CD4+ Teff cells in tumor infiltrating lymphocytes. Overexpression of CTLA4 reversed the inhibitory action of combination treatment on cancer growth. Our data suggest that combining radiotherapy and cantharidin may have synergistic effects in driving tumor rejection by increasing T-cell infiltration, proliferation and cytokine production.


Asunto(s)
Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Carcinoma Pulmonar de Lewis/radioterapia , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/radioterapia , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/efectos de la radiación , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/efectos de la radiación , Antígeno CTLA-4/inmunología , Cantaridina/administración & dosificación , Cantaridina/efectos adversos , Carcinoma Pulmonar de Lewis/inmunología , Carcinoma Pulmonar de Lewis/patología , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Terapia Combinada , Modelos Animales de Enfermedad , Humanos , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/efectos de la radiación , Ratones , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/efectos de la radiación
10.
Nutr Cancer ; 68(5): 791-803, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27046042

RESUMEN

We herein compared the effects of the chronic feeding of high-fat (HF), high-sucrose (HS), and low-fat/low-sucrose (control) diets on carcinogenesis following chronic ultraviolet B (UVB) irradiation in hairless mice. UVB irradiation-induced carcinogenesis was more prominent in HF diet-fed group than in control diet- and HS diet-fed groups. The HS diet group, as well as the HF diet one, showed tumor development and growth, increased skin matrix metalloproteinase (MMP) and blood plasminogen activator inhibitor-1 (PAI-1) levels, and decreased blood leptin and adiponectin levels after long-term UVB irradiation. These changes were smaller in the HS diet group than in the HF diet group. In addition, no difference was noted in the above changes between the control and HS diet groups. The increase induced in adipose tissue weight by the HF diet was markedly reduced by UVB irradiation. This result suggests that the abundant availability of lipids in hypertrophic adipose tissue may be related to tumor incidence and growth through increases in blood PAI-1 and skin MMP-9 expression levels and decreases in blood adiponectin levels by UVB irradiation. In conclusion, HF diet-induced hypertrophic adipose tissue is an important cancer risk factor that promotes UV irradiation-induced carcinogenesis and tumor growth.


Asunto(s)
Carcinogénesis/efectos de la radiación , Dieta Alta en Grasa/efectos adversos , Sacarosa en la Dieta/efectos adversos , Rayos Ultravioleta/efectos adversos , Adiponectina/sangre , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/efectos de la radiación , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/efectos de la radiación , Quimiocina CCL2/sangre , Insulina/sangre , Leptina/sangre , Masculino , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Pelados , Ratones Endogámicos C57BL , Factores de Riesgo , Serpina E2/sangre , Piel/efectos de los fármacos , Piel/metabolismo , Piel/efectos de la radiación
11.
Georgian Med News ; (256-257): 106-11, 2016 Jul.
Artículo en Ruso | MEDLINE | ID: mdl-27661286

RESUMEN

The study aim was to investigate the combined influence of emotional stress and low doses of ionizing radiation (0.2 Gr) on cellular immunity of laboratory animals in the remote period. One hundred and twenty Wistar rats were divided into 4 groups: 1 group control, 2 group - exposed to gamma-radiation, 3 group - exposed to emotional stress, 4 group was exposed to the combined influence of emotional stress and gamma-radiation. Emotional stress was simulated by tail suspension. Animals from groups 2 and 4 were exposed to a single dose of 0.2 Gr 90 days prior the investigation via «TERAGAM¼ 60Co (ISOTREND spol. s.r.o., Check Republic). The results of our study show that in a remote period after exposure to a low dose of gamma-radiation the decrease of quantitative and increase of qualitative indicators of cellular immunity are observed, which is manifested by lymphopenia and decease of CD3+- CD4+ - and CD8+-lymphocytes subpopulations, and lymphokin-producing capacity of leucocytes. The late phase of stress-reaction is characterized by lymphocytosis, increase in absolute numbers of CD3+- and CD4+- lymphocytes, normal range of CD8+- cells and lymphokin-producing capacity of leucocytes and decrease of immunoregulatory index.


Asunto(s)
Inmunidad Celular/efectos de la radiación , Leucocitos/efectos de la radiación , Traumatismos Experimentales por Radiación/inmunología , Estrés Psicológico/inmunología , Animales , Complejo CD3/sangre , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/efectos de la radiación , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Rayos gamma , Leucocitos/inmunología , Linfocinas/sangre , Linfopenia/sangre , Linfopenia/inmunología , Masculino , Ratas Wistar
12.
Lancet Oncol ; 16(13): e498-509, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26433823

RESUMEN

Checkpoint blockade immunotherapy has received mainstream attention as a result of striking and durable clinical responses in some patients with metastatic disease and a reasonable response rate in many tumour types. The activity of checkpoint blockade immunotherapy is not restricted to melanoma or lung cancer, and additional indications are expected in the future, with responses already reported in renal cancer, bladder cancer, and Hodgkin's lymphoma among many others. Additionally, the interactions between radiation and the immune system have been investigated, with several studies describing the synergistic effects on local and distant tumour control when radiation therapy is combined with immunotherapy. Clinical enthusiasm for this approach is strengthened by the many ongoing trials combining immunotherapy with definitive and palliative radiation. Herein, we discuss the biological and mechanistic rationale behind combining radiation with checkpoint blockade immunotherapy, with a focus on the preclinical data supporting this potentially synergistic combination. We explore potential hypotheses and important considerations for clinical trial designs. Finally, we reintroduce the notion of radiosensitising immunotherapy, akin to radiosensitising chemotherapy, as a potential definitive therapeutic modality.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Quimioradioterapia/métodos , Inmunoterapia/métodos , Neoplasias/terapia , Tolerancia a Radiación , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Transducción de Señal/efectos de los fármacos , Transducción de Señal/efectos de la radiación , Animales , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/efectos de la radiación , Muerte Celular/efectos de los fármacos , Muerte Celular/efectos de la radiación , Citotoxicidad Inmunológica/efectos de los fármacos , Citotoxicidad Inmunológica/efectos de la radiación , Células Dendríticas/diagnóstico por imagen , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Humanos , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/efectos de la radiación , Terapia Molecular Dirigida , Neoplasias/inmunología , Neoplasias/patología , Dosis de Radiación , Radiografía , Resultado del Tratamiento
13.
Apoptosis ; 20(3): 371-82, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25398538

RESUMEN

We tested apoptosis levels in in vitro irradiated T-lymphocytes from breast cancer (BC) patients with radiotherapy-induced late effects. Previous results reported in the literature were revised. We also examined the effect of TP53 Arg72Pro polymorphism on irradiation-induced apoptosis (IA). Twenty BC patients, ten with fibrosis and/or telangiectasias and ten matched controls with no late reactions, were selected from those receiving radiotherapy between 1993 and 2007. All patients were followed-up at least 6 years after radiotherapy. Using the combination of both CD3 and CD8 antibodies the in vitro IA was measured in CD3, CD8 and CD4 T-lymphocytes, and CD8 natural killer lymphocytes (CD8 NK) by flow cytometry. The TP53 Arg72Pro genotype was determined by sequencing. Patients with late radiotherapy toxicity showed less IA for all T-lymphocytes except for the CD8 NK. CD8 NK showed the highest spontaneous apoptosis and the lowest IA. IA in patients with toxicity appears to be lower than the control patients only in TP53 Arg/Arg patients (P = 0.077). This difference was not present in patients carrying at least one Pro allele (P = 0.8266). Our data indicate that late side effects induced by radiotherapy of BC are associated to low levels of IA. CD8 NK cells have a different response to in vitro irradiation compared to CD8 T-lymphocytes. It would be advisable to distinguish the CD8 NK lymphocytes from the pool of CD8+ lymphocytes in IA assays using CD8+ cells. Our data suggest that the 72Pro TP53 allele may influence the IA of patients with radiotherapy toxicity.


Asunto(s)
Neoplasias de la Mama/radioterapia , Linfocitos T CD4-Positivos/efectos de la radiación , Linfocitos T CD8-positivos/efectos de la radiación , Rayos gamma/efectos adversos , Células Asesinas Naturales/efectos de la radiación , Subgrupos Linfocitarios/efectos de la radiación , Polimorfismo Genético , Proteína p53 Supresora de Tumor/genética , Adulto , Apoptosis/efectos de la radiación , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Estudios de Casos y Controles , Células Cultivadas , Femenino , Fibrosis , Expresión Génica , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/patología , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/patología , Persona de Mediana Edad , Pronóstico , Estudios Prospectivos , Tolerancia a Radiación , Telangiectasia/genética , Telangiectasia/metabolismo , Telangiectasia/patología , Resultado del Tratamiento , Proteína p53 Supresora de Tumor/metabolismo
14.
J Immunol ; 190(4): 1873-81, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23315072

RESUMEN

Adoptive cell transfer (ACT) of ex vivo-activated autologous tumor-reactive T cells is currently one of the most promising approaches for cancer immunotherapy. Recent studies provided some evidence that IL-17-producing CD8(+) (Tc17) cells may exhibit potent antitumor activity, but the specific mechanisms have not been completely defined. In this study, we used a murine melanoma lung-metastasis model and tested the therapeutic effects of gp100-specific polarized type I CD8(+) cytotoxic T (Tc1) or Tc17 cells combined with autologous bone marrow transplantation after total body irradiation. Bone marrow transplantation combined with ACT of antitumor (gp100-specific) Tc17 cells significantly suppressed the growth of established melanoma, whereas Tc1 cells induced long-term tumor regression. After ACT, Tc1 cells maintained their phenotype to produce IFN-γ, but not IL-17. However, although Tc17 cells largely preserved their ability to produce IL-17, a subset secreted IFN-γ or both IFN-γ and IL-17, indicating the plasticity of Tc17 cells in vivo. Furthermore, after ACT, the Tc17 cells had a long-lived effector T cell phenotype (CD127(hi)/KLRG-1(low)) as compared with Tc1 cells. Mechanistically, Tc1 cells mediated antitumor immunity primarily through the direct effect of IFN-γ on tumor cells. In contrast, despite the fact that some Tc17 cells also secreted IFN-γ, Tc17-mediated antitumor immunity was independent of the direct effects of IFN-γ on the tumor. Nevertheless, IFN-γ played a critical role by creating a microenvironment that promoted Tc17-mediated antitumor activity. Taken together, these studies demonstrate that both Tc1 and Tc17 cells can mediate effective antitumor immunity through distinct effector mechanisms, but Tc1 cells are superior to Tc17 cells in mediating tumor regression.


Asunto(s)
Traslado Adoptivo/métodos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/trasplante , Interleucina-17/biosíntesis , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/terapia , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Animales , Trasplante de Médula Ósea/inmunología , Trasplante de Médula Ósea/métodos , Trasplante de Médula Ósea/patología , Linfocitos T CD8-positivos/efectos de la radiación , Citotoxicidad Inmunológica/efectos de la radiación , Interleucina-17/efectos de la radiación , Neoplasias Pulmonares/patología , Activación de Linfocitos/inmunología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Irradiación Corporal Total/métodos , Antígeno gp100 del Melanoma/biosíntesis , Antígeno gp100 del Melanoma/inmunología
15.
Int J Mol Sci ; 16(12): 30405-21, 2015 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-26703577

RESUMEN

Sub-lethal doses of radiation can modulate gene expression, making tumor cells more susceptible to T-cell-mediated immune attack. Proteasome inhibitors demonstrate broad anti-tumor activity in clinical and pre-clinical cancer models. Here, we use a combination treatment of proteasome inhibition and irradiation to further induce immunomodulation of tumor cells that could enhance tumor-specific immune responses. We investigate the effects of the 26S proteasome inhibitor, bortezomib, alone or in combination with radiotherapy, on the expression of immunogenic genes in normal colon and colorectal cancer cell lines. We examined cells for changes in the expression of several death receptors (DR4, DR5 and Fas) commonly used by T cells for killing of target cells. Our results indicate that the combination treatment resulted in increased cell surface expression of death receptors by increasing their transcript levels. The combination treatment further increases the sensitivity of carcinoma cells to apoptosis through FAS and TRAIL receptors but does not change the sensitivity of normal non-malignant epithelial cells. Furthermore, the combination treatment significantly enhances tumor cell killing by tumor specific CD8⁺ T cells. This study suggests that combining radiotherapy and proteasome inhibition may simultaneously enhance tumor immunogenicity and the induction of antitumor immunity by enhancing tumor-specific T-cell activity.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis , Bortezomib/farmacología , Linfocitos T CD8-positivos/efectos de los fármacos , Inhibidores de Proteasoma/farmacología , Radiación Ionizante , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/efectos de la radiación , Línea Celular , Células Epiteliales/efectos de los fármacos , Células Epiteliales/efectos de la radiación , Células HCT116 , Humanos , Receptores de Muerte Celular/metabolismo
16.
Int J Cancer ; 134(10): 2383-92, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24154990

RESUMEN

Radiation therapy (RT) continues to be a cornerstone in the treatment for many cancers. Unfortunately, not all individuals respond effectively to RT resulting clinically in two groups consisting of nonresponders (progressive disease) and responders (tumor control/cure). The mechanisms that govern the outcome of radiotherapy are poorly understood. Interestingly, a new paradigm has emerged demonstrating that the immune system mediates many of the antitumor effects of RT. Therefore, we hypothesized that the immune response following RT may dictate the efficacy of treatment. To examine this, we developed a tumor model that mirrors this clinically relevant phenomenon in which mice bearing Colon38, a colon adenocarcinoma, were treated locally with 15Gy RT resulting in both nonresponders and responders. More importantly, we were able to distinguish responders from nonresponders as early as 4 days post-RT allowing for the unique opportunity to identify critical events that ultimately determined the effectiveness of therapy. Intratumoral immune cells and interferon-gamma were increased in responsive tumors and licensed CD8 T cells to exhibit lytic activity against tumor cells, a response that was diminished in tumors refractory to RT. Combinatorial treatment with RT and the immunomodulatory cytokine IL-12 resulted in complete remission of cancer in 100% of cases compared to a cure rate of only 12% with RT alone. Similar data were obtained when IL-12 was delivered by microspheres. Therefore, the efficacy of RT may depend on the strength of the immune response induced after radiotherapy. Additionally, immunotherapy that further stimulates the immune cells may enhance the effectiveness of RT.


Asunto(s)
Adenocarcinoma/radioterapia , Linfocitos T CD8-positivos/efectos de la radiación , Neoplasias del Colon/radioterapia , Citotoxicidad Inmunológica/efectos de la radiación , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/inmunología , Análisis de Varianza , Animales , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Quimioradioterapia , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/inmunología , Citotoxicidad Inmunológica/efectos de los fármacos , Sistema Inmunológico/efectos de los fármacos , Sistema Inmunológico/patología , Sistema Inmunológico/efectos de la radiación , Interferón gamma/inmunología , Interferón gamma/metabolismo , Interleucina-12/farmacología , Ratones , Resultado del Tratamiento
17.
Cancer Immunol Immunother ; 63(3): 259-71, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24357146

RESUMEN

The need for an intact immune system for cancer radiation therapy to be effective suggests that radiation not only acts directly on the tumor but also indirectly, through the activation of host immune components. Recent studies demonstrated that endogenous type I interferons (type I IFNs) play a role in radiation-mediated anti-tumor immunity by enhancing the ability of dendritic cells to cross-prime CD8(+) T cells. However, it is still unclear to what extent endogenous type I IFNs contribute to the recruitment and function of CD8(+) T cells. Little is also known about the effects of type I IFNs on myeloid cells. In the current study, we demonstrate that type I and type II IFNs (IFN-γ) are both required for the increased production of CXCL10 (IP-10) chemokine by myeloid cells within the tumor after radiation treatment. Radiation-induced intratumoral IP-10 levels in turn correlate with tumor-infiltrating CD8(+) T cell numbers. Moreover, type I IFNs promote potent tumor-reactive CD8(+) T cells by directly affecting the phenotype, effector molecule production, and enhancing cytolytic activity. Using a unique inducible expression system to increase local levels of IFN-α exogenously, we show here that the capacity of radiation therapy to result in tumor control can be enhanced. Our preclinical approach to study the effects of local increase in IFN-α levels can be used to further optimize the combination therapy strategy in terms of dosing and scheduling, which may lead to better clinical outcome.


Asunto(s)
Linfocitos T CD8-positivos/efectos de la radiación , Interferón-alfa/metabolismo , Neoplasias Mamarias Animales/radioterapia , Melanoma Experimental/inmunología , Melanoma Experimental/radioterapia , Células Mieloides/efectos de los fármacos , Animales , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Movimiento Celular/efectos de la radiación , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Citotoxicidad Inmunológica/genética , Citotoxicidad Inmunológica/efectos de la radiación , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/efectos de la radiación , Humanos , Interferón-alfa/genética , Interferón-alfa/farmacología , Interferón gamma/genética , Interferón gamma/metabolismo , Recuento de Linfocitos , Neoplasias Mamarias Animales/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/inmunología , Trasplante de Neoplasias
18.
Blood ; 119(16): 3844-53, 2012 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-22101894

RESUMEN

It is currently thought that acute GVHD cannot be elicited in the absence of Ag presentation by radiosensitive host hematopoietic-derived APCs after allogeneic BM transplantation. Because clinical data suggest that sex-mismatched H-Y Ags may be important minor histocompatibility Ags for GVH responses, we directly tested their relevance and ability to initiate GVHD when presented by either the hematopoietic- (host or donor) or the nonhematopoietic-derived APCs. H-Y minor Ag incompatibility elicited both CD4(+) and CD8(+) T-cell driven GVHD lethality. Studies with various well-established BM chimera recipients, in contrast to the current views, have reported that in the absence of functional radiosensitive host hematopoietic-derived APCs, H-Y Ag presentation by either the donor hematopoietic-derived or the host nonhematopoietic-derived APCs is sufficient for inducing GVHD. Our data further suggest that infusion of sufficient numbers of alloreactive donor T cells will induce GVHD in the absence of radiosensitive host hematopoietic-derived APCs.


Asunto(s)
Trasplante de Médula Ósea/efectos adversos , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/terapia , Antígeno H-Y/inmunología , Hematopoyesis/inmunología , Histocompatibilidad/inmunología , Enfermedad Aguda , Animales , Células Presentadoras de Antígenos/citología , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/efectos de la radiación , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/efectos de la radiación , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/efectos de la radiación , Células Cultivadas , Células Dendríticas/citología , Células Dendríticas/inmunología , Células Endoteliales/citología , Células Endoteliales/inmunología , Femenino , Isoantígenos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Mutantes , Tolerancia a Radiación , Timectomía
19.
Strahlenther Onkol ; 190(2): 210-6, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24362501

RESUMEN

BACKGROUND AND PURPOSE: A close relationship exists between immune response and tumor behavior. This study aimed to explore the associations between radiation-induced apoptosis (RIA) in peripheral blood lymphocytes (PBL) and clinical pathological variables. Furthermore, it assessed the role of RIA as a prognostic factor for survival in cervical carcinoma patients. PATIENTS AND METHODS: Between February 1998 and October 2003, 58 consecutive patients with nonmetastatic, localized stage I-II cervical carcinoma who had been treated with radiotherapy (RT) ± chemotherapy were included in this study. Follow-up ended in January 2013. PBL subpopulations were isolated and irradiated with 0, 1, 2 and 8 Gy then incubated for 24, 48 and 72 h. Apoptosis was measured by flow cytometry and the ß value, a parameter defining RIA of lymphocytes, was calculated. RESULTS: Mean follow-up duration was 111.92 ± 40.31 months. Patients with lower CD8 T lymphocyte ß values were at a higher risk of local relapse: Exp(B) = 5.137, confidence interval (CI) 95 % = 1.044-25.268, p = 0.044. Similar results were observed for regional relapse: Exp(B) = 8.008, CI 95 % = 1.702-37.679, p = 0.008 and disease relapse: Exp(B) = 6.766, CI 95 % = 1.889-24.238, p = 0.003. In multivariate analysis, only the CD8 T lymphocyte ß values were found to be of prognostic significance for local disease-free survival (LDFS, p = 0.049), regional disease-free survival (RDFS, p = 0.002), metastasis-free survival (MFS, p = 0.042), disease-free survival (DFS, p = 0.001) and cause-specific survival (CSS p = 0.028). CONCLUSION: For the first time, RIA in CD8 T lymphocytes was demonstrated to be a predictive factor for survival in cervical carcinoma patients.


Asunto(s)
Apoptosis/efectos de la radiación , Linfocitos T CD8-positivos/efectos de la radiación , Neoplasias del Cuello Uterino/inmunología , Neoplasias del Cuello Uterino/radioterapia , Adulto , Anciano , Anciano de 80 o más Años , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Braquiterapia , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Quimioradioterapia , Cisplatino/administración & dosificación , Terapia Combinada , Supervivencia sin Enfermedad , Femenino , Citometría de Flujo , Estudios de Seguimiento , Humanos , Persona de Mediana Edad , Estadificación de Neoplasias , Pronóstico , Dosificación Radioterapéutica , Neoplasias del Cuello Uterino/tratamiento farmacológico , Neoplasias del Cuello Uterino/mortalidad , Neoplasias del Cuello Uterino/patología
20.
Stem Cells ; 31(1): 137-45, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22961695

RESUMEN

The regeneration of the hematopoietic system following total body irradiation is supported by host-derived mesenchymal stromal cells (MSCs) within the bone marrow. The mechanisms used by MSCs to survive radiation doses that are lethal to the hematopoietic system are poorly understood. The DNA damage response (DDR) represents a cohort of signaling pathways that enable cells to execute biological responses to genotoxic stress. Here, we examine the role of the DDR in mediating the resistance of MSCs to ionizing radiation (IR) treatment using two authentic clonal mouse MSC lines, MS5 and ST2, and primary bulk mouse MSCs. We show that multiple DDR mechanisms contribute to the radio-resistance of MSCs: robust DDR activation via rapid γ-H2AX formation, activation of effective S and G(2)/M DNA damage checkpoints, and efficient repair of IR-induced DNA double-strand breaks. We show that MSCs are intrinsically programmed to maximize survival following IR treatment by expressing high levels of key DDR proteins including ATM, Chk2, and DNA Ligase IV; high levels of the anti-apoptotic, Bcl-2 and Bcl-(XL); and low levels of the pro-apoptotic, Bim and Puma. As a result, we demonstrate that irradiated mouse MSCs withstand IR-induced genotoxic stress, continue to proliferate, and retain their capacity to differentiate long-term along mesenchymal-derived lineages. We have shown, for the first time, that the DDR plays key roles in mediating the radioresistance of mouse MSCs which may have important implications for the study and application of MSCs in allogeneic bone marrow transplantation, graft-versus-host disease, and cancer treatment.


Asunto(s)
Daño del ADN/efectos de la radiación , Reparación del ADN/efectos de la radiación , Células Madre Mesenquimatosas/efectos de la radiación , Tolerancia a Radiación/fisiología , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Linfocitos T CD4-Positivos/efectos de la radiación , Linfocitos T CD8-positivos/efectos de la radiación , Puntos de Control del Ciclo Celular , Proteínas de Ciclo Celular/biosíntesis , Diferenciación Celular/efectos de la radiación , Línea Celular , Proliferación Celular/efectos de la radiación , Quinasa de Punto de Control 2 , Roturas del ADN de Doble Cadena/efectos de la radiación , ADN Ligasa (ATP) , ADN Ligasas/biosíntesis , Proteínas de Unión al ADN/biosíntesis , Histonas/biosíntesis , Histonas/metabolismo , Células Madre Mesenquimatosas/citología , Ratones , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Radiación Ionizante , Transducción de Señal , Proteínas Supresoras de Tumor/biosíntesis , Proteína bcl-X/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA