Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Mol Ther ; 23(9): 1541-50, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25896248

RESUMEN

Here, we describe a fatal serious adverse event observed in a patient infused with autologous T-cell receptor (TCR) transduced T cells. This TCR, originally obtained from a melanoma patient, recognizes the well-described HLA-A*0201 restricted 26-35 epitope of MART-1, and was not affinity enhanced. Patient 1 with metastatic melanoma experienced a cerebral hemorrhage, epileptic seizures, and a witnessed cardiac arrest 6 days after cell infusion. Three days later, the patient died from multiple organ failure and irreversible neurologic damage. After T-cell infusion, levels of IL-6, IFN-γ, C-reactive protein (CRP), and procalcitonin increased to extreme levels, indicative of a cytokine release syndrome or T-cell-mediated inflammatory response. Infused T cells could be recovered from blood, broncho-alveolar lavage, ascites, and after autopsy from tumor sites and heart tissue. High levels of NT-proBNP indicate semi-acute heart failure. No cross reactivity of the modified T cells toward a beating cardiomyocyte culture was observed. Together, these observations suggest that high levels of inflammatory cytokines alone or in combination with semi-acute heart failure and epileptic seizure may have contributed substantially to the occurrence of the acute and lethal event. Protocol modifications to limit the risk of T-cell activation-induced toxicity are discussed.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/efectos adversos , Inmunoterapia Adoptiva/efectos adversos , Antígeno MART-1/inmunología , Receptores de Antígenos de Linfocitos T/genética , Especificidad del Receptor de Antígeno de Linfocitos T/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Adulto , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Resultado Fatal , Femenino , Humanos , Inmunoterapia Adoptiva/métodos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Antígeno MART-1/metabolismo , Melanoma/diagnóstico , Melanoma/genética , Melanoma/inmunología , Melanoma/terapia , Estadificación de Neoplasias , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción Genética
2.
Nucleic Acids Res ; 40(21): 10821-31, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23012265

RESUMEN

DNA double-strand breaks (DSBs) are biologically one of the most important cellular lesions and possess varying degrees of chemical complexity. The notion that the repairability of more chemically complex DSBs is inefficient led to the concept that the extent of DSB complexity underlies the severity of the biological consequences. The repair of DSBs by non-homologous end joining (NHEJ) has been extensively studied but it remains unknown whether more complex DSBs require a different sub-set of NHEJ protein for their repair compared with simple DSBs. To address this, we have induced DSBs in fluorescently tagged mammalian cells (Ku80-EGFP, DNA-PKcs-YFP or XRCC4-GFP, key proteins in NHEJ) using ultra-soft X-rays (USX) or multi-photon near infrared (NIR) laser irradiation. We have shown in real-time that simple DSBs, induced by USX or NIR microbeam irradiation, are repaired rapidly involving Ku70/80 and XRCC4/Ligase IV/XLF. In contrast, DSBs with greater chemical complexity are repaired slowly involving not only Ku70/80 and XRCC4/Ligase IV/XLF but also DNA-PKcs. Ataxia telangiectasia-mutated inhibition only retards repair of the more chemically complex DSBs which require DNA-PKcs. In summary, the repair of DSBs by NHEJ is highly regulated with pathway choice and kinetics of repair dependent on the chemical complexity of the DSB.


Asunto(s)
Antígenos Nucleares/metabolismo , Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades , Proteína Quinasa Activada por ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Radiación Ionizante , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/antagonistas & inhibidores , Línea Celular , Proteínas de Unión al ADN/antagonistas & inhibidores , Cinética , Autoantígeno Ku , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Supresoras de Tumor/antagonistas & inhibidores
3.
Radiat Res ; 169(2): 214-22, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18220463

RESUMEN

We developed a biochemical kinetics approach to describe the repair of double-strand breaks (DSBs) produced by low-LET radiation by modeling molecular events associated with non-homologous end joining (NHEJ). A system of coupled nonlinear ordinary differential equations describes the induction of DSBs and activation pathways for major NHEJ components including Ku70/80, DNA-PKcs, and the ligase IV-XRCC4 heterodimer. The autophosphorylation of DNA-PKcs and subsequent induction of gamma-H2AX foci observed after ionizing radiation exposure were modeled. A two-step model of regulation of repair by DNA-PKcs was developed with an initial step allowing access of other NHEJ components to breaks and a second step limiting access to ligase IV-XRCC4. Our model assumes that the transition from the first to the second step depends on DSB complexity, with a much slower rate for complex DSBs. The model faithfully reproduced several experimental data sets, including DSB rejoining as measured by pulsed-field gel electrophoresis (PFGE) at 10 min postirradiation or longer and quantification of the induction of gamma-H2AX foci. A process that is independent of DNA-PKcs is required for the model to reproduce experimental data for rejoining before 10 min postirradiation. Predictions are made for the behaviors of NHEJ components at low doses and dose rates, and a steady state is found at dose rates of 0.1 Gy/h or lower.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN/genética , Reparación del ADN/efectos de la radiación , ADN/química , ADN/genética , Histonas/química , Modelos Genéticos , Bioquímica/métodos , Simulación por Computador , ADN/efectos de la radiación , Histonas/genética , Histonas/efectos de la radiación , Humanos , Cinética , Modelos Químicos
4.
Photochem Photobiol ; 84(6): 1506-14, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18557822

RESUMEN

DNA double strand breaks (DSBs) are amongst the most deleterious lesions induced within the cell following exposure to ionizing radiation. Mammalian cells repair these breaks predominantly via the nonhomologous end joining pathway which is active throughout the cell cycle and is error prone. The alternative pathway for repair of DSBs is homologous recombination (HR) which is error free and active during S- and G2/M-phases of the cell cycle. We have utilized near-infrared laser radiation to induce DNA damage in individual mammalian cells through multiphoton excitation processes to investigate the dynamics of single cell DNA damage processing. We have used immunofluorescent imaging of gamma-H2AX (a marker for DSBs) in mammalian cells and investigated the colocalization of this protein with ATM, p53 binding protein 1 and RAD51, an integral protein of the HR DNA repair pathway. We have observed persistent DSBs at later times postlaser irradiation which are indicative of DSBs arising at replication, presumably from UV photoproducts or clustered damage containing single strand breaks. Cell cycle studies have shown that in G1 cells, a significant fraction of multiphoton laser-induced prompt DSBs persists for > 4 h in addition to those induced at replication.


Asunto(s)
Ciclo Celular/efectos de la radiación , Roturas del ADN de Doble Cadena/efectos de la radiación , Replicación del ADN/genética , Replicación del ADN/efectos de la radiación , ADN/genética , Fotones , Animales , Línea Celular , Cricetinae , Cricetulus , Roturas del ADN de Cadena Simple/efectos de la radiación , Cinética , Recombinasa Rad51/metabolismo
5.
Int J Radiat Biol ; 82(2): 111-8, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16546909

RESUMEN

PURPOSE: To investigate quantitatively the induction and rejoining of DNA double strand breaks (DSB) in V79-4 and xrs-5 Chinese hamster cells and HF19 human fibroblast cells, using the phosphorylation of the histone protein H2AX (gamma-H2AX) as an indicator of DSB, exposed to low doses of either low linear energy transfer (LET) (60)Co gamma-rays or high LET a-particles. MATERIALS AND METHODS: Cells were irradiated with low or high LET (20 - 2000 mGy). The gamma-H2AX foci were detected using immunohistochemistry and quantified by image analysis. RESULTS: The number of DSB determined 30 min post gamma-irradiation at 37 degrees C is 12.2 (+/-1.5), 13.5 (+/-1.6) and 19.1 (+/-1.7) foci/cell/Gy for V79-4, xrs-5 and HF19 cells respectively, comparable with levels detected in V79-4 cells using pulse field gel electrophoresis. 6 h post gamma-irradiation, gamma-H2AX foci levels in V79-4 and HF19 cells approach control levels but remain higher in DSB repair deficient xrs-5 cells. Gamma-H2AX foci levels remain significantly higher than controls at 6 h in a-irradiated cells. CONCLUSIONS: Gamma-radiation and alpha-radiation induced the phosphorylation of H2AX in response to DSB at low doses; the variation in the rate of dephosphorylation of induced foci are dependent both on radiation quality and cell characteristics.


Asunto(s)
Daño del ADN , ADN/efectos de la radiación , Fibroblastos/efectos de la radiación , Histonas/genética , Histonas/efectos de la radiación , Partículas alfa , Animales , Línea Celular , Cricetinae , Cricetulus , Relación Dosis-Respuesta en la Radiación , Rayos gamma , Humanos , Transferencia Lineal de Energía , Dosis de Radiación
6.
Methods Mol Biol ; 296: 157-66, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15576930

RESUMEN

The method described in the following chapter utilizes a double thymidine block (an inhibitor of DNA synthesis) followed by treatment of cells with nocodazole (a mitotic inhibitor) to obtain large cell populations at distinct phases of the cell cycle. Treatment with double thymidine results in a G1/S-phase arrested cell population, and the use of flow cytometry allows progression of the cells through the cell cycle to be monitored. Flow cytometry enables the calculation of timings for collection of cells at distinct cell cycle phases from G1/S (following treatment with thymidine) through to G2/M (owing to the presence of nocodazole).


Asunto(s)
Ciclo Celular/fisiología , Técnicas Citológicas/métodos , Animales , Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Citometría de Flujo/métodos , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Nocodazol/farmacología , Ratas , Timidina/metabolismo , Timidina/farmacología
7.
Methods Mol Biol ; 296: 113-53, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15576929

RESUMEN

In recent years, we have witnessed major advances in our understanding of the mammalian cell cycle and how it is regulated. Normal mammalian cellular proliferation is tightly regulated at each phase of the cell cycle by the activation and deactivation of a series of proteins that constitute the cell cycle machinery. This review article describes the various phases of the mammalian cell cycle and focuses on the cell cycle regulatory molecules that act at each stage to ensure normal cellular progression.


Asunto(s)
Ciclo Celular/fisiología , Animales , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/fisiología , Quinasas Ciclina-Dependientes/fisiología , Ciclinas/fisiología , Citocinesis , Replicación del ADN , Mamíferos , Mitosis , Modelos Biológicos
8.
J Pharm Pharmacol ; 55(4): 519-26, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12803774

RESUMEN

Abnormal vascular smooth muscle cell (VSMC) proliferation plays an important role in the pathogenesis of both atherosclerosis and restenosis. Recent studies suggest that high-dose salicylates, in addition to inhibiting cyclooxygenase activity, exert an antiproliferative effect on VSMC growth both in-vitro and in-vivo. However, whether all non-steroidal anti-inflammatory drugs (NSAIDs) exert similar antiproliferative effects on VSMCs, and do so via a common mechanism of action, remains to be shown. In this study, we demonstrate that the NSAIDs aspirin, sodium salicylate, diclofenac, ibuprofen, indometacin and sulindac induce a dose-dependent inhibition of proliferation in rat A10 VSMCs in the absence of significant cytotoxicity. Flow cytometric analyses showed that exposure of A10 cells to diclofenac, indometacin, ibuprofen and sulindac, in the presence of the mitotic inhibitor, nocodazole, led to a significant G0/G1 arrest. In contrast, the salicylates failed to induce a significant G1 arrest since flow cytometry profiles were not significantly different from control cells. Cyclin A levels were elevated, and hyperphosphorylated p107 was present at significant levels, in salicylate-treated A10 cells, consistent with a post-G1/S block, whereas cyclin A levels were low, and hypophosphorylated p107 was the dominant form, in cells treated with other NSAIDs consistent with a G1 arrest. The ubiquitously expressed cyclin-dependent kinase (CDK) inhibitors, p21 and p27, were increased in all NSAID-treated cells. Our results suggest that diclofenac, indometacin, ibuprofen and sulindac inhibit VSMC proliferation by arresting the cell cycle in the G1 phase, whereas the growth inhibitory effect of salicylates probably affects the late S and/or G2/M phases. Irrespective of mechanism, our results suggest that NSAIDs might be of benefit in the treatment of certain vasculoproliferative disorders.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , División Celular/efectos de los fármacos , Animales , Antiinflamatorios no Esteroideos/administración & dosificación , Aorta/citología , Aorta/efectos de los fármacos , Aspirina/administración & dosificación , Aspirina/farmacología , Línea Celular/efectos de los fármacos , Línea Celular/metabolismo , Diclofenaco/administración & dosificación , Diclofenaco/farmacología , Relación Dosis-Respuesta a Droga , Citometría de Flujo , Ibuprofeno/administración & dosificación , Ibuprofeno/farmacología , Indometacina/administración & dosificación , Indometacina/farmacología , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Ratas , Salicilato de Sodio/administración & dosificación , Salicilato de Sodio/farmacología , Sulindac/administración & dosificación , Sulindac/farmacología
9.
Sci Transl Med ; 5(197): 197ra103, 2013 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-23926201

RESUMEN

MAGE A3, which belongs to the family of cancer-testis antigens, is an attractive target for adoptive therapy given its reactivation in various tumors and limited expression in normal tissues. We developed an affinity-enhanced T cell receptor (TCR) directed to a human leukocyte antigen (HLA)-A*01-restricted MAGE A3 antigen (EVDPIGHLY) for use in adoptive therapy. Extensive preclinical investigations revealed no off-target antigen recognition concerns; nonetheless, administration to patients of T cells expressing the affinity-enhanced MAGE A3 TCR resulted in a serious adverse event (SAE) and fatal toxicity against cardiac tissue. We present a description of the preclinical in vitro functional analysis of the MAGE A3 TCR, which failed to reveal any evidence of off-target activity, and a full analysis of the post-SAE in vitro investigations, which reveal cross-recognition of an off-target peptide. Using an amino acid scanning approach, a peptide from the muscle protein Titin (ESDPIVAQY) was identified as an alternative target for the MAGE A3 TCR and the most likely cause of in vivo toxicity. These results demonstrate that affinity-enhanced TCRs have considerable effector functions in vivo and highlight the potential safety concerns for TCR-engineered T cells. Strategies such as peptide scanning and the use of more complex cell cultures are recommended in preclinical studies to mitigate the risk of off-target toxicity in future clinical investigations.


Asunto(s)
Presentación de Antígeno/inmunología , Antígenos de Neoplasias/inmunología , Conectina/química , Reacciones Cruzadas/inmunología , Antígeno HLA-A1/inmunología , Proteínas de Neoplasias/inmunología , Péptidos/inmunología , Linfocitos T/metabolismo , Secuencia de Aminoácidos , Antígenos de Neoplasias/química , Antineoplásicos/farmacología , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Conectina/inmunología , Reacciones Cruzadas/efectos de los fármacos , Células HEK293 , Humanos , Activación de Linfocitos/efectos de los fármacos , Datos de Secuencia Molecular , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Proteínas de Neoplasias/química , Péptidos/química , Ingeniería de Proteínas , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/efectos de los fármacos
10.
Nat Med ; 18(6): 980-7, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22561687

RESUMEN

T cell immunity can potentially eradicate malignant cells and lead to clinical remission in a minority of patients with cancer. In the majority of these individuals, however, there is a failure of the specific T cell receptor (TCR)­mediated immune recognition and activation process. Here we describe the engineering and characterization of new reagents termed immune-mobilizing monoclonal TCRs against cancer (ImmTACs). Four such ImmTACs, each comprising a distinct tumor-associated epitope-specific monoclonal TCR with picomolar affinity fused to a humanized cluster of differentiation 3 (CD3)-specific single-chain antibody fragment (scFv), effectively redirected T cells to kill cancer cells expressing extremely low surface epitope densities. Furthermore, these reagents potently suppressed tumor growth in vivo. Thus, ImmTACs overcome immune tolerance to cancer and represent a new approach to tumor immunotherapy.


Asunto(s)
Citotoxicidad Inmunológica , Neoplasias Experimentales/terapia , Receptores de Antígenos de Linfocitos T/fisiología , Animales , Linfocitos T CD8-positivos/inmunología , Humanos , Memoria Inmunológica , Inmunoterapia , Interferón gamma/biosíntesis , Activación de Linfocitos , Ratones , Ratones SCID , Neoplasias Experimentales/inmunología
11.
DNA Repair (Amst) ; 9(8): 907-13, 2010 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-20634148

RESUMEN

When cells are exposed to radiation serious lesions are introduced into the DNA including double strand breaks (DSBs), single strand breaks (SSBs), base modifications and clustered damage sites (a specific feature of ionizing radiation induced DNA damage). Radiation induced DNA damage has the potential to initiate events that can lead ultimately to mutations and the onset of cancer and therefore understanding the cellular responses to DNA lesions is of particular importance. Using gammaH2AX as a marker for DSB formation and RAD51 as a marker of homologous recombination (HR) which is recruited in the processing of frank DSBs or DSBs arising from stalled replication forks, we have investigated the contribution of SSBs and non-DSB DNA damage to the induction of DSBs in mammalian cells by ionizing radiation during the cell cycle. V79-4 cells and human HF19 fibroblast cells have been either irradiated with 0-20Gy of gamma radiation or, for comparison, treated with a low concentration of hydrogen peroxide, which is known to induce SSBs but not DSBs. Inhibition of the repair of oxidative DNA lesions by poly(ADP ribose) polymerase (PARP) inhibitor leads to an increase in radiation induced gammaH2AX and RAD51 foci which we propose is due to these lesions colliding with replication forks forming replication induced DSBs. It was confirmed that DSBs are not induced in G(1) phase cells by treatment with hydrogen peroxide but treatment does lead to DSB induction, specifically in S phase cells. We therefore suggest that radiation induced SSBs and non-DSB DNA damage contribute to the formation of replication induced DSBs, detected as RAD51 foci.


Asunto(s)
Roturas del ADN de Doble Cadena/efectos de la radiación , Roturas del ADN de Cadena Simple/efectos de la radiación , Replicación del ADN/efectos de la radiación , Animales , Línea Celular , Cricetinae , Cricetulus , Roturas del ADN de Cadena Simple/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Fibroblastos/metabolismo , Fibroblastos/patología , Fase G1/efectos de la radiación , Histonas/genética , Humanos , Peróxido de Hidrógeno/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Recombinasa Rad51/genética , Radiación Ionizante , Fase S/efectos de la radiación
12.
Radiat Res ; 174(2): 195-205, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20681786

RESUMEN

Cellular lesions (e.g. DSBs) are induced into DNA upon exposure to radiation, with DSB complexity increasing with radiation ionization density. Using M059K and M059J human glioblastoma cells (proficient and deficient in DNA-PKcs activity, respectively), we investigated the repair of DNA damage, including DSBs, induced by high- and low-LET radiation [gamma rays, alpha particles and high-charge and energy (HZE) ions]. In the absence of DNA-PKcs activity, less DSB repair and increased recruitment of RAD51 was seen at 24 h. After exposure to (56)Fe heavy ions, the number of cells with RAD51 tracks was less than the number of cells with gamma-H2AX at 24 h with both cell lines. Using alpha particles, comparable numbers of cells with visible gamma-H2AX and RAD51 were seen at 24 h in both cell lines. M059J cells irradiated with alpha particles accumulated in S phase, with a greater number of cyclin A and RAD51 co-stained cells seen at 24 h compared with M059K cells, where an S-phase block is absent. It is proposed that DNA-PKcs plays a role in the repair of some frank DSBs, which are longer-lived in NHEJ-deficient cells, and some non-DSB clustered damage sites that are converted into DSBs at replication as the cell cycles through to S phase.


Asunto(s)
Proteína Quinasa Activada por ADN/efectos de la radiación , Rayos gamma/efectos adversos , Radioisótopos de Hierro/farmacología , Línea Celular Tumoral , Roturas del ADN de Doble Cadena , Reparación del ADN/efectos de la radiación , Proteína Quinasa Activada por ADN/aislamiento & purificación , Proteína Quinasa Activada por ADN/metabolismo , Electroforesis en Gel de Campo Pulsado , Fibroblastos/enzimología , Fibroblastos/fisiología , Fibroblastos/efectos de la radiación , Glioblastoma , Humanos , Proteína Quinasa C/aislamiento & purificación , Proteína Quinasa C/metabolismo , Proteína Quinasa C/efectos de la radiación , Dosis de Radiación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA