Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Radiat Environ Biophys ; 58(1): 129-135, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30456560

RESUMO

Sustained pain relief following radon spa therapy in patients suffering from chronic painful diseases has been well described. But still, the underlying mechanisms are not fully understood. We conducted the prospective and explorative RAD-ON01 study which included 103 patients who suffered from chronic painful musculoskeletal disorders of the spine and/or joints and present here the data of the examination of pro- and anti-inflammatory cytokines in the serum of the patients before and at weeks 6, 12 and 30 after therapy. While TNFα, IL-1ß, IFNγ, IL-1Ra and IL-10 were not altered, TGFß was temporarily significantly (p = 0.013) elevated 6 weeks after therapy. Importantly, this elevation positively correlated with lowered pain sensitivity (r = 0.41). Further, the amount of IL-18 in the serum positively correlated with lowered pain sensitivity. Therefore, IL-18 can be considered as predictive marker for pain sensitivity of radon spa patients. We conclude that alterations in TGFß and general IL-18 levels in serum have prognostic and predictive value in situations of lowered pain by exposure of patients to very low-doses of radiation as it is the case in radon spa.


Assuntos
Banhos , Interleucina-18/sangue , Dor/sangue , Dor/radioterapia , Radônio/uso terapêutico , Fator de Crescimento Transformador beta/sangue , Biomarcadores/sangue , Feminino , Humanos , Masculino , Estudos Prospectivos
2.
Radiat Oncol ; 10: 197, 2015 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-26383236

RESUMO

BACKGROUND: Radiotherapy is an integral part of breast cancer treatment. Immune activating properties of especially hypofractionated irradiation are in the spotlight of clinicians, besides the well-known effects of radiotherapy on cell cycle and the reduction of the clonogenic potential of tumor cells. Especially combination of radiotherapy with further immune stimulation induces immune-mediated anti-tumor responses. We therefore examined whether hypofractionated irradiation alone or in combination with hyperthermia as immune stimulants is capable of inducing breast cancer cells with immunogenic potential. METHODS: Clonogenic assay, AnnexinA5-FITC/Propidium iodide assay and ELISA analyses of heat shock protein 70 and high mobility group box 1 protein were applied to characterize colony forming capability, cell death induction, cell death forms and release of danger signals by breast cancer cells in response to hypofractionated radiation (4x4Gy, 6x3Gy) alone and in combination with hyperthermia (41.5 °C for 1 h). Caspase-3 deficient, hormone receptor positive, p53 wild type MCF-7 and caspase-3 intact, hormone receptor negative, p53 mutated MDA-MB231 breast cancer cells, the latter in absence or presence of the pan-caspase inhibitor zVAD-fmk, were used. Supernatants of the treated tumor cells were analyzed for their potential to alter the surface expression of activation markers on human-monocyte-derived dendritic cells. RESULTS: Irradiation reduced the clonogenicity of caspase deficient MCF-7 cells more than of MDA-B231 cells. In contrast, higher amounts of apoptotic and necrotic cells were induced in MDA-B231 cells after single irradiation with 4Gy, 10Gy, or 20Gy or after hypofractionated irradiation with 4x4Gy or 6x3Gy. MDA-B231 cells consecutively released higher amounts of Hsp70 and HMGB1 after hypofractionated irradiation. However, only the release of Hsp70 was further increased by hyperthermia. Both, apoptosis induction and release of the danger signals, was dependent on caspase-3. Only supernatants of MDA-B231 cells after hypofractionated irradiation resulted in slight changes of activation markers on dendritic cells; especially that of CD86 was upregulated and HT did not further impact on it. CONCLUSIONS: Hypofractionated irradiation is the main stimulus for cell death induction and consecutive dendritic cell activation in caspase proficient breast cancer cells. For the assessment of radiosensitivity and immunological effects of radio- and immunotherapies the readout system is crucial.


Assuntos
Neoplasias da Mama/imunologia , Neoplasias da Mama/radioterapia , Caspase 3/metabolismo , Radioterapia/métodos , Neoplasias da Mama/metabolismo , Morte Celular/efeitos da radiação , Linhagem Celular Tumoral , Células Dendríticas/imunologia , Células Dendríticas/efeitos da radiação , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Hipertermia Induzida/métodos , Técnicas In Vitro , Hipofracionamento da Dose de Radiação , Tolerância a Radiação/fisiologia
3.
Radiat Oncol ; 9(1): 89, 2014 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-24678590

RESUMO

BACKGROUND: Glioblastoma multiforme (GBM) is the most common primary brain tumor in adults. Despite a multimodal therapy consisting of resection followed by fractionated radiotherapy (RT) combined with the chemotherapeutic agent (CT) temozolomide (TMZ), its recurrence is almost inevitable. Since the immune system is capable of eliminating small tumor masses, a therapy should also aim to stimulate anti-tumor immune responses by induction of immunogenic cell death forms. The histone deacetylase inhibitor valproic acid (VPA) might foster this. METHODS: Reflecting therapy standards, we applied in our in vitro model fractionated RT with a single dose of 2Gy and clinically relevant concentrations of CT. Not only the impact of RT and/or CT with TMZ and/or VPA on the clonogenic potential and cell cycle of the glioblastoma cell lines T98G, U251MG, and U87MG was analyzed, but also the resulting cell death forms and release of danger signals such as heat-shock protein70 (Hsp70) and high-mobility group protein B1 (HMGB1). RESULTS: The clonogenic assays revealed that T98G and U251MG, having mutated tumor suppressor protein p53, are more resistant to RT and CT than U87MG with wild type (WT) p53. In all glioblastoma cells lines, fractionated RT induced a G2 cell cycle arrest, but only in the case of U87MG, TMZ and/or VPA alone resulted in this cell cycle block. Further, fractionated RT significantly increased the number of apoptotic and necrotic tumor cells in all three cell lines. However, only in U87MG, the treatment with TMZ and/or VPA alone, or in combination with fractionated RT, induced significantly more cell death compared to untreated or irradiated controls. While necrotic glioblastoma cells were present after VPA, TMZ especially led to significantly increased amounts of U87MG cells in the radiosensitive G2 cell cycle phase. While CT did not impact on the release of Hsp70, fractionated RT resulted in significantly increased extracellular concentrations of Hsp70 in p53 mutated and WT glioblastoma cells. CONCLUSIONS: Our results indicate that fractionated RT is the main stimulus for induction of glioblastoma cell death forms with immunogenic potential. The generated tumor cell microenvironment might be beneficial to include immune therapies for GBM in the future.


Assuntos
Neoplasias Encefálicas/radioterapia , Fracionamento da Dose de Radiação , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Glioblastoma/radioterapia , Proteínas de Choque Térmico HSP70/metabolismo , Proteína Supressora de Tumor p53/genética , Antineoplásicos/administração & dosagem , Apoptose , Ciclo Celular , Morte Celular , Linhagem Celular Tumoral , Dacarbazina/administração & dosagem , Dacarbazina/análogos & derivados , Fase G2 , Proteína HMGB1/metabolismo , Humanos , Mutação , Necrose , Temozolomida , Ácido Valproico/química
4.
J Immunotoxicol ; 9(3): 301-13, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22800185

RESUMO

Chemotherapeutic agents (CT) and ionizing radiation (X-ray) induce DNA damage and primarily aim to stop the proliferation of tumor cells. However, multimodal anti-cancer therapies should finally result in tumor cell death and, best, in the induction of systemic anti-tumor immunity. Since distinct therapy-induced tumor cell death forms may create an immune activating tumor microenvironment, this study examined whether sole treatment with CT that are used in the therapy for colorectal cancer or in combination with X-ray result in colorectal tumor cell death with immunogenic potential. 5-Fluorouracil (5-FU), Oxaliplatin (Oxp), or Irinotecan (Irino) in combination with X-ray were all potent inhibitors of colorectal tumor cell colony formation. This study then examined the forms of cell death with AnnexinA5-FITC/Propidium Iodide staining. Necrosis was the prominent form of cell death induced by CT and/or X-ray. While only a combination of Irino with X-ray leads to death induction already 1 day after treatment, also the combinations of Oxp or 5-FU with X-ray and X-ray alone resulted in high necrosis rates at later time points after treatment. Inhibition of apoptosis increased the amount of necrotic tumor cells, suggesting that a programmed form of necrosis can be induced by CT + X-ray. 5-FU and Oxp alone or in combination with X-ray and Irino plus X-ray were most effective in increasing the expression of RIP, IRF-5, and p53, proteins involved in necrotic and apoptotic cell death pathways. All treatments further resulted in the release of the immune activating danger signals high-mobility group box 1 (HMGB1) and heat shock protein 70 (HSP70). The supernatants of the treated tumor cells induced maturation of dendritic cells. It is, therefore, concluded that combination of CT with X-ray is capable of inducing in vitro cell death forms of colorectal tumors with immunogenic potential.


Assuntos
Adenocarcinoma , Apoptose/imunologia , Quimiorradioterapia/métodos , Neoplasias Colorretais , Microambiente Tumoral/imunologia , Adenocarcinoma/imunologia , Adenocarcinoma/terapia , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/terapia , Humanos , Proteínas de Neoplasias/imunologia , Raios X
5.
Strahlenther Onkol ; 186(11): 587-99, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21069267

RESUMO

PURPOSE: The malignancy of tumor cells can be attenuated by interfering with cell death pathways. Since hyperthermia (HT) is a very potent radiosensitizer, the influence of HT (41.5 °C for 1 hour) alone and in combination with ionising irradiation (X-ray; 5 Gy or 10 Gy) on the form of cell death as well as on the expression of proteins known to be major components in tumor cells' apoptotic and necrotic pathways were examined in colorectal tumor cells. MATERIAL AND METHODS: The expression of proteins was analysed by western blot and the relative activity of caspases-3/7 by fluorescence- based assay. Colony formation was analysed using the clonogenic assay and cell death was determined with annexin V-FITC/propidium iodide staining. RESULTS: Combining X-ray with HT led to similar activation of caspase-3/7 and p53 expression in comparison to irradiation only while the amount of the pro-apoptotic proteins PUMA and Bax was increased in HCT15 and SW480 cells. HT alone or combinations with X-ray further resulted in a temporarily increased level of the anti-apoptotic protein Bcl-2. Irradiation plus HT further led to an up-regulation of IRF-5. The levels of RIP-1, a marker for programmed necrosis, increased in tumor cells which were treated with HT and/or X-ray. Combining 5 Gy irradiation with HT compared to irradiation resulted in a significantly increased number of necrotic tumor cells and in decreased colony formation. CONCLUSION: The combined treatment of colorectal tumor cells with X-ray and HT activates distinct tumor cell pathways and fosters the early appearance of a necrotic tumor cell phenotype.


Assuntos
Apoptose/efeitos da radiação , Morte Celular/efeitos da radiação , Neoplasias Colorretais/patologia , Hipertermia Induzida , Proteínas Reguladoras de Apoptose/análise , Western Blotting , Caspase 3/análise , Caspase 7/análise , Terapia Combinada , Relação Dose-Resposta à Radiação , Ativação Enzimática/efeitos da radiação , Humanos , Necrose , Células-Tronco Neoplásicas , Proteínas Proto-Oncogênicas/análise , Proteínas Proto-Oncogênicas c-bcl-6/análise , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/análise , Proteína X Associada a bcl-2/análise
6.
Biochem Biophys Res Commun ; 391(1): 1014-20, 2010 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-19968962

RESUMO

Colorectal cancer is the second leading cause of death in developed countries. Tumor therapies should on the one hand aim to stop the proliferation of tumor cells and to kill them, and on the other hand stimulate a specific immune response against residual cancer cells. Dying cells are modulators of the immune system contributing to anti-inflammatory or pro-inflammatory responses, depending on the respective cell death form. The positive therapeutic effects of temperature-controlled hyperthermia (HT), when combined with ionizing irradiation (X-ray), were the origin to examine whether combinations of X-ray with HT can induce immune activating tumor cell death forms, also characterized by the release of the danger signal HMGB1. Human colorectal tumor cells with differing radiosensitivities were treated with combinations of HT (41.5 degrees C for 1h) and X-ray (5 or 10Gy). Necrotic cell death was prominent after X-ray and could be further increased by HT. Apoptosis remained quite low in HCT 15 and SW480 cells. X-ray and combinations with HT arrested the tumor cells in the radiosensitive G2 cell cycle phase. The amount of released HMGB1 protein was significantly enhanced after combinatorial treatments in comparison to single ones. We conclude that combining X-ray with HT may induce anti-tumor immunity as a result of the predominant induction of inflammatory necrotic tumor cells and the release of HMGB1.


Assuntos
Neoplasias Colorretais/terapia , Proteína HMGB1/metabolismo , Hipertermia Induzida , Apoptose/imunologia , Neoplasias Colorretais/patologia , Neoplasias Colorretais/radioterapia , Terapia Combinada , Fase G2/efeitos da radiação , Humanos , Sistema Imunitário/efeitos da radiação , Necrose/imunologia , Tolerância a Radiação
7.
Autoimmunity ; 42(4): 311-3, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19811286

RESUMO

Autoimmune diseases and cancer can be treated by influencing the immune system. Apo and nec cells are strong modulators of the immune system contributing to anti-inflammatory and pro-inflammatory responses, respectively. We examined which form of cell death was induced by HT and X-irradiation. Nec was the prominent form of cell death after combined treatment and the amount of dead cells was higher when exposing the cells to radiation before HT. Combined applications further led to an increased percentage of cells in a more radioresponsive G2 cell cycle phase. The danger signal HMGB1 is released when combining HT with radiation, a further hint that those treatments may induce inflammation and immune activation. We conclude that immune responses are appropriately adapted to the damage that has occurred and may contribute to anti-cancer immunity or chronic autoimmunity, respectively.


Assuntos
Apoptose/efeitos da radiação , Hipertermia Induzida/efeitos adversos , Inflamação/imunologia , Necrose/imunologia , Apoptose/imunologia , Linhagem Celular Tumoral , Proteína HMGB1/biossíntese , Proteína HMGB1/efeitos da radiação , Humanos , Raios X
8.
J Cell Mol Med ; 13(7): 1391-9, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18624762

RESUMO

Proteins of the annexin family bind to phospholipids in a Ca(2+) dependent manner. The exposure of phosphatidylserine (PS) by apoptotic as well as necrotic cells is one major eat-me-signal for macrophages. Annexin A5 (Anx A5) preferentially binds to PS. The availability of Anx A5 knock out (KO) mice allowed us to investigate for the first time if endogenous Anx A5 modulates the immune response towards allogeneic cells. Furthermore, the effect of Anx A5 gene deletion on the phagocytic process as well as on the inflammatory reaction of macrophages was explored. We found that Anx A5 KO mice have a strongly reduced allogeneic cellular immune reaction against primary as well as secondary necrotic cells. In vivo phagocytosis experiments revealed that macrophages of Anx A5 KO mice displayed an increased uptake of necrotic cells. Additionally, an increased secretion of the anti-inflammatory cytokine IL-10 of isolated macrophages of Anx A5 KO mice after contact with necrotic cells was observed. Furthermore, the promoter activity of the Anx A5 gene was enhanced after stimulation of macrophages. The tumour size of an allogeneic tumour regressed faster when endogenous Anx A5 was present. These data demonstrate that endogenous Anx A5 influences the phagocytosis of necrotic cells, modulates the immune response towards allogeneic cells and acts as an inflammatory protein.


Assuntos
Anexina A5/deficiência , Inflamação/imunologia , Macrófagos/imunologia , Necrose/imunologia , Animais , Anexina A5/metabolismo , Comunicação Celular , Linhagem Celular Tumoral , Interleucina-10/metabolismo , Ativação de Macrófagos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Regressão Neoplásica Espontânea/patologia , Fenótipo
9.
Strahlenther Onkol ; 182(8): 467-72, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16896593

RESUMO

BACKGROUND AND PURPOSE: Apoptosis can be induced by distinct but overlapping pathways. Ionizing radiation induces apoptosis by an "intrinsic", mitochondria-dependent pathway. Ligation of tumor necrosis factor-(TNF-)alpha, FAS (CD95) or TRAIL receptors are typical representatives of an extrinsic, death-receptor-mediated pathway. In this study the effect of irradiation, treatment with the cytokine TNF-alpha, or a combination of both on the induction of apoptosis and clonogenic survival of bladder carcinoma cells was investigated. MATERIAL AND METHODS: 5637 bladder carcinoma cells were treated with different concentrations of recombinant TNF-alpha (0-10 ng/ml), irradiated with single doses ranging from 0.5 to 10 Gy, or a combination of both modalities. Apoptotic cells were quantified by the TUNEL assay up to 96 h following treatment, clonogenic cell survival by a clonogenic assay. Synergistic effects of both modalities were evaluated using isobolographic analysis. RESULTS: Irradiation of 5637 carcinoma cells resulted in a discontinuous dose dependence of the apoptotic fraction with a pronounced increase in the range of 0-2 Gy and a slighter increase at 2-10 Gy. The percentage of apoptotic carcinoma cells also increased continuously after treatment with lower concentrations of TNF-alpha reaching a plateau at concentrations of 5.0-10.0 ng/ml. Isobolographic analysis revealed a supraadditive interrelationship between irradiation and TNF-alpha in the range between 0.005 and 0.5 ng/ml, and an additive effect for TNF-alpha concentrations>0.5 ng/ml. The additive effects were confirmed in clonogenic survival assays with reduced survival fractions following combined TNF-alpha administration and irradiation. CONCLUSION: The combination of two apoptosis-inducing modalities resulted in a synergistic effect on the induction of apoptosis in 5637 bladder carcinoma cells. Although a radiosensitizing effect still has to be proven in animal models, combined-modality treatment may increase the therapeutic effectiveness of irradiation in bladder cancer.


Assuntos
Apoptose , Fator de Necrose Tumoral alfa/uso terapêutico , Neoplasias da Bexiga Urinária/tratamento farmacológico , Neoplasias da Bexiga Urinária/radioterapia , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Apoptose/efeitos da radiação , Linhagem Celular Tumoral , Ensaio de Unidades Formadoras de Colônias , Terapia Combinada , Relação Dose-Resposta a Droga , Relação Dose-Resposta à Radiação , Humanos , Marcação In Situ das Extremidades Cortadas , Dosagem Radioterapêutica , Fatores de Tempo , Resultado do Tratamento , Fator de Necrose Tumoral alfa/administração & dosagem , Ensaio Tumoral de Célula-Tronco , Neoplasias da Bexiga Urinária/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...