Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 423
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Cell Biochem Funct ; 42(6): e4103, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39073207

RESUMO

The cancer treatment landscape is significantly evolving, focusing on advanced radiation therapy methods to maximize effectiveness and minimize the adverse effects. Recognized as a pivotal component in cancer and disease treatment, radiation therapy (RT) has drawn attention in recent research that delves into its intricate interplay with inflammation and the immune response. This exploration unveils the underlying processes that significantly influence treatment outcomes. In this context, the potential advantages of combining bronchoscopy with RT across diverse clinical scenarios, alongside the targeted impact of brachytherapy, are explored. Concurrently, radiation treatments serve multifaceted roles such as DNA repair, cell elimination, and generating immune stress signaling molecules known as damage-associated molecular patterns, elucidating their effectiveness in treating various diseases. External beam RT introduces versatility by utilizing particles such as photons, electrons, protons, or carbon ions, each offering distinct advantages. Advanced RT techniques contribute to the evolving landscape, with emerging technologies like FLASH, spatially fractionated RT, and others poised to revolutionize the field. The comprehension of RT, striving for improved treatment outcomes, reduced side effects, and facilitating personalized and innovative treatments for cancer and noncancer patients. After navigating these advancements, the goal is fixed to usher in a new era in which RT is a cornerstone of precision and effectiveness in medical interventions. In summarizing the myriad findings, the review underscores the significance of understanding the differential impacts of radiation approaches on inflammation and immune modulation, offering valuable insights for developing innovative therapeutic interventions that harness the immune system in conjunction with RT.


Assuntos
Sistema Imunitário , Neoplasias , Humanos , Neoplasias/radioterapia , Neoplasias/imunologia , Sistema Imunitário/efeitos da radiação , Sistema Imunitário/metabolismo , Radioterapia/efeitos adversos , Radioterapia/métodos , Inflamação/radioterapia , Inflamação/imunologia , Reparo do DNA
2.
Int J Clin Oncol ; 28(2): 201-208, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35556190

RESUMO

The effects of irradiation on tumor tissue and the host immune system are interrelated. The antitumor effect of irradiation is attenuated in the immunocompromised hosts. In addition, radiation alone positively and negatively influences the host immune system. The positive effects of radiation are summarized by the ability to help induce and enhance tumor-antigen-specific immune responses. The cancer-immunity cycle is a multistep framework that illustrates how the tumor-antigen-specific immune responses are induced and how the induced antigen-specific immune cells exert their functions in tumor tissues. Irradiation affects each step of this cancer-immunity cycle, primarily in a positive manner. In contrast, radiation also has negative effects on the immune system. The first is that irradiation has the possibility to kill irradiated effector immune cells. The second is that irradiation upregulates immunosuppressive molecules in the tumor microenvironment, whereas the third is that irradiation to the tumor condenses immunosuppressor cells in the tumor microenvironment. When used in conjunction with radiotherapy, immune checkpoint inhibitors can further leverage the positive effects of radiation on the immune system and compensate for the negative effects of irradiation, which supports the rationale for the combination of radiotherapy and immune checkpoint inhibitors. In this review, we summarize the preclinical evidence for the reciprocal effects of radiation exposure and the immune system, and up-front topics of the combination therapy of immune checkpoint inhibitors and radiotherapy.


Assuntos
Inibidores de Checkpoint Imunológico , Neoplasias , Humanos , Inibidores de Checkpoint Imunológico/uso terapêutico , Neoplasias/tratamento farmacológico , Antígenos de Neoplasias , Sistema Imunitário/patologia , Sistema Imunitário/efeitos da radiação , Terapia Combinada , Microambiente Tumoral , Imunoterapia , Radioterapia
3.
Cancer Immunol Immunother ; 71(3): 541-552, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34269847

RESUMO

PURPOSE: The influence of radiotherapy on patient immune cell subsets has been established by several groups. Following a previously published analysis of immune changes during and after curative radiotherapy for prostate cancer, this analysis focused on describing correlations of changes of immune cell subsets with radiation treatment parameters. PATIENTS AND METHODS: For 13 patients treated in a prospective trial with radiotherapy to the prostate region (primary analysis) and five patients treated with radiotherapy to prostate and pelvic nodal regions (exploratory analysis), already published immune monitoring data were correlated with clinical data as well as radiation planning parameters such as clinical target volume (CTV) and volumes receiving 20 Gy (V20) for newly contoured volumes of pelvic blood vessels and bone marrow. RESULTS: Most significant changes among immune cell subsets were observed at the end of radiotherapy. In contrast, correlations of age and CD8+ subsets (effector and memory cells) were observed early during and 3 months after radiotherapy. Ratios of T cells and T cell proliferation compared to baseline correlated with CTV. Early changes in regulatory T cells (Treg cells) and CD8+ effector T cells correlated with V20 of blood vessels and bone volumes. CONCLUSIONS: Patient age as well as radiotherapy planning parameters correlated with immune changes during radiotherapy. Larger irradiated volumes seem to correlate with early suppression of anti-cancer immunity. For immune cell analysis during normofractionated radiotherapy and correlations with treatment planning parameters, different time points should be looked at in future projects. TRIAL REGISTRATION NUMBER: NCT01376674, 20.06.2011.


Assuntos
Biomarcadores , Sistema Imunitário/efeitos da radiação , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/radioterapia , Dosagem Radioterapêutica , Planejamento da Radioterapia Assistida por Computador , Adulto , Fatores Etários , Humanos , Imunofenotipagem , Contagem de Leucócitos , Subpopulações de Linfócitos/metabolismo , Subpopulações de Linfócitos/efeitos da radiação , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Neoplasias da Próstata/patologia , Radioterapia Guiada por Imagem , Adulto Jovem
4.
J Appl Microbiol ; 130(1): 14-24, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32692438

RESUMO

The gut microbiome is well recognized to have a pivotal role in regulation of the health and behaviour of the host, affecting digestion, metabolism, immunity, and has been linked to changes in bones, muscles and the brain, to name a few. However, the impact of microgravity environment on gut bacteria is not well understood. In space environments, astronauts face several health issues including stress, high iron diet, radiation and being in a closed system during extended space missions. Herein, we discuss the role of gut bacteria in the space environment, in relation to factors such as microgravity, radiation and diet. Gut bacteria may exact their effects by synthesis of molecules, their absorption, and through physiological effects on the host. Moreover we deliberate the role of these challenges in the dysbiosis of the human microbiota and possible dysregulation of the immune system.


Assuntos
Meio Ambiente Extraterreno , Microbioma Gastrointestinal/fisiologia , Dieta/efeitos adversos , Disbiose/fisiopatologia , Humanos , Sistema Imunitário/fisiologia , Sistema Imunitário/efeitos da radiação , Radiação , Voo Espacial , Estresse Fisiológico/fisiologia , Estresse Fisiológico/efeitos da radiação , Ausência de Peso/efeitos adversos
5.
Int J Mol Sci ; 22(18)2021 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-34576217

RESUMO

Radiation therapy (RT) recruits myeloid cells, leading to an immunosuppressive microenvironment that impedes its efficacy against tumors. Combination of immunotherapy with RT is a potential approach to reversing the immunosuppressive condition and enhancing tumor control after RT. This study aimed to assess the effects of local interleukin-12 (IL-12) therapy on improving the efficacy of RT in a murine prostate cancer model. Combined treatment effectively shrunk the radioresistant tumors by inducing a T helper-1 immune response and influx of CD8+ T cells. It also delayed the radiation-induced vascular damage accompanied by increased α-smooth muscle actin-positive pericyte coverage and blood perfusion. Moreover, RT significantly reduced the IL-12-induced levels of alanine aminotransferase in blood. However, it did not further improve the IL-12-induced anti-tumor effect on distant tumors. Upregulated expression of T-cell exhaustion-associated genes was found in tumors treated with IL-12 only and combined treatment, suggesting that T-cell exhaustion is potentially correlated with tumor relapse in combined treatment. In conclusion, this study illustrated that combination of radiation and local IL-12 therapy enhanced the host immune response and promoted vascular maturation and function. Furthermore, combination treatment was associated with less systemic toxicity than IL-12 alone, providing a potential option for tumor therapy in clinical settings.


Assuntos
Sistema Imunitário/efeitos da radiação , Subunidade p35 da Interleucina-12/metabolismo , Radioterapia/métodos , Actinas/metabolismo , Animais , Linfócitos T CD8-Positivos/metabolismo , Imuno-Histoquímica , Imunossupressores/farmacologia , Imunoterapia , Interferon gama/metabolismo , Fígado/metabolismo , Fígado/patologia , Linfócitos do Interstício Tumoral/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso/metabolismo , Transplante de Neoplasias , Pericitos/metabolismo , Neoplasias da Próstata/metabolismo , Microambiente Tumoral/imunologia
6.
Int J Mol Sci ; 22(9)2021 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-33925430

RESUMO

Although ionizing radiation (radiation) is commonly used for medical diagnosis and cancer treatment, radiation-induced damages cannot be avoided. Such damages can be classified into direct and indirect damages, caused by the direct absorption of radiation energy into DNA and by free radicals, such as hydroxyl radicals (•OH), generated in the process of water radiolysis. More specifically, radiation damage concerns not only direct damages to DNA, but also secondary damages to non-DNA targets, because low-dose radiation damage is mainly caused by these indirect effects. Molecular hydrogen (H2) has the potential to be a radioprotective agent because it can selectively scavenge •OH, a reactive oxygen species with strong oxidizing power. Animal experiments and clinical trials have reported that H2 exhibits a highly safe radioprotective effect. This paper reviews previously reported radioprotective effects of H2 and discusses the mechanisms of H2, not only as an antioxidant, but also in intracellular responses including anti-inflammation, anti-apoptosis, and the regulation of gene expression. In doing so, we demonstrate the prospects of H2 as a novel and clinically applicable radioprotective agent.


Assuntos
Hidrogênio/farmacologia , Neoplasias/terapia , Lesões por Radiação/prevenção & controle , Protetores contra Radiação/farmacologia , Animais , Antioxidantes/farmacologia , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/prevenção & controle , Gastroenteropatias/etiologia , Gastroenteropatias/prevenção & controle , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos da radiação , Humanos , Hidrogênio/uso terapêutico , Sistema Imunitário/efeitos dos fármacos , Sistema Imunitário/efeitos da radiação , Masculino , Qualidade de Vida , Protetores contra Radiação/uso terapêutico , Pele/efeitos dos fármacos , Pele/efeitos da radiação , Espermatozoides/efeitos dos fármacos , Espermatozoides/efeitos da radiação
7.
Strahlenther Onkol ; 196(11): 1018-1033, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32519025

RESUMO

BACKGROUND: In this exploratory study, the impact of local irradiation on systemic changes in stress and immune parameters was investigated in eight patients treated with intensity-modulated radiation therapy (IMRT) or stereotactic ablative body radiotherapy (SABR) for prostate adenocarcinoma to gain deeper insights into how radiotherapy (RT) modulates the immune system. PATIENTS AND METHODS: RT-qPCR, flow cytometry, metabolomics, and antibody arrays were used to monitor a panel of stress- and immune-related parameters before RT, after the first fraction (SABR) or the first week of treatment (IMRT), after the last fraction, and 3 weeks later in the blood of IMRT (N = 4) or SABR (N = 4) patients. Effect size analysis was used for comparison of results at different timepoints. RESULTS: Several parameters were found to be differentially modulated in IMRT and SABR patients: the expression of TGFB1, IL1B, and CCL3 genes; the expression of HLA-DR on circulating monocytes; the abundance and ratio of phosphatidylcholine and lysophosphatidylcholine metabolites in plasma. More immune modulators in plasma were modulated during IMRT than SABR, with only two common proteins, namely GDF-15 and Tim­3. CONCLUSION: Locally delivered RT induces systemic modulation of the immune system in prostate adenocarcinoma patients. IMRT and SABR appear to specifically affect distinct immune components.


Assuntos
Adenocarcinoma/radioterapia , Adenocarcinoma/cirurgia , Sistema Imunitário/efeitos da radiação , Metaboloma/efeitos da radiação , Proteínas de Neoplasias/sangue , Neoplasias da Próstata/radioterapia , Neoplasias da Próstata/cirurgia , Proteoma/efeitos da radiação , Radiocirurgia/métodos , Radioterapia de Intensidade Modulada/métodos , Estresse Fisiológico/efeitos da radiação , Adenocarcinoma/imunologia , Adenocarcinoma/fisiopatologia , Idoso , Idoso de 80 Anos ou mais , Biomarcadores , Citocinas/sangue , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Antígenos HLA/sangue , Humanos , Mediadores da Inflamação/sangue , Lisofosfatidilcolinas/sangue , Masculino , Pessoa de Meia-Idade , Monócitos/imunologia , Fosfatidilcolinas/sangue , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/fisiopatologia
8.
Oncology ; 98(4): 202-215, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32079015

RESUMO

BACKGROUND: Malignant melanoma represents the deadliest form of skin cancer with a high tendency to metastasize during the early course of the disease. Radiation therapy has long played a key role in the management of both local and metastatic melanoma. Although local radiation therapy exerts antitumor effects by damaging the cellular DNA, it also induces an important out-of-field (distant) effect known as the "abscopal effect" in nonirradiated sites. Radiation therapy-induced abscopal effects are believed to be mediated by activation and stimulation of the immune system. OBJECTIVE: To provide a detailed overview of the current state of knowledge and clinical experience of radiation therapy-induced abscopal effects in patients with malignant melanoma. METHODS: Using electronic databases such as MEDLINE via PubMed and Google Scholar, a systematic literature review was performed to find published clinical evidence for radiation therapy-induced abscopal effects in patients with malignant melanoma. The clinical data on radiation therapy-induced abscopal effects were reviewed and the outcomes summarized. RESULTS: Clinical evidence of patients with malignant melanoma was gathered using databases from MEDLINE and those findings were summarized. Although the precise mechanism of the abscopal effect of radiation therapy is still not completely understood, evidence suggests that tumor cell destruction by radiation releases tumor antigens that stimulate the immune system of the host to activate the body's immune effector cells systemically and produce distant non-target antitumor effects. This forms a basis for using the radiation therapy with immunotherapy to augment the abscopal response rates. CONCLUSIONS: Current clinical evidence suggests that there is a large potential to enhance the abscopal effect when radiation therapy is combined with immunotherapeutic agents for the treatment of malignant melanoma. Ongoing and planned clinical trials may provide us with a more in-depth understanding of how this combination therapy can be optimally utilized clinically to achieve improved survival outcomes among patients with malignant melanoma.


Assuntos
Efeito Espectador , Sistema Imunitário/efeitos da radiação , Melanoma/radioterapia , Humanos
9.
Mol Ecol ; 28(20): 4620-4635, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31498518

RESUMO

Wildlife inhabiting environments contaminated by radionuclides face putative detrimental effects of exposure to ionizing radiation, with biomarkers such as an increase in DNA damage and/or oxidative stress commonly associated with radiation exposure. To examine the effects of exposure to radiation on gene expression in wildlife, we conducted a de novo RNA sequencing study of liver and spleen tissues from a rodent, the bank vole Myodes glareolus. Bank voles were collected from the Chernobyl Exclusion Zone (CEZ), where animals were exposed to elevated levels of radionuclides, and from uncontaminated areas near Kyiv, Ukraine. Counter to expectations, we did not observe a strong DNA damage response in animals exposed to radionuclides, although some signs of oxidative stress were identified. Rather, exposure to environmental radionuclides was associated with upregulation of genes involved in lipid metabolism and fatty acid oxidation in the livers - an apparent shift in energy metabolism. Moreover, using stable isotope analysis, we identified that fur from bank voles inhabiting the CEZ had enriched isotope values of nitrogen: such an increase is consistent with increased fatty acid metabolism, but also could arise from a difference in diet or habitat between the CEZ and elsewhere. In livers and spleens, voles inhabiting the CEZ were characterized by immunosuppression, such as impaired antigen processing, and activation of leucocytes involved in inflammatory responses. In conclusion, exposure to low dose environmental radiation impacts pathways associated with immunity and lipid metabolism, potentially as a stress-induced coping mechanism.


Assuntos
Acidente Nuclear de Chernobyl , Metabolismo dos Lipídeos/fisiologia , Fígado/patologia , Exposição à Radiação/efeitos adversos , Baço/patologia , Animais , Arvicolinae , Dano ao DNA/efeitos da radiação , Reparo do DNA/efeitos da radiação , Ácidos Graxos/metabolismo , Sistema Imunitário/efeitos da radiação , Metabolismo dos Lipídeos/genética , Fígado/imunologia , Mutagênicos/efeitos adversos , Oxirredução/efeitos da radiação , Estresse Oxidativo/efeitos da radiação , Radiação Ionizante , Radioisótopos/efeitos adversos , Baço/imunologia , Ucrânia
10.
Gynecol Oncol ; 154(1): 236-245, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30995960

RESUMO

Immunotherapy (IO) is an important new pillar in the treatment of solid tumors, and the integration of IO agents with chemotherapy, targeted therapy, surgery and radiation has yet to be defined. As preclinical and clinical studies have described synergistic activity with the combination of radiation and immunotherapy, many clinical trials are underway to explore both the safety and efficacy of this approach both in the metastatic and definitive setting. Through immune priming, radiation may enhance local tumor control at the irradiated site, as well as induce a systemic response to control distant metastasis, known as the abscopal effect. On a mechanistic level, radiation therapy releases tumor neoantigens and activates an adaptive immune response that is mediated by cytotoxic T-cells, which then hone to sites of irradiated tumor as well as non-irradiated tumor metastases to induce immunogenic tumor cell death. Although the abscopal effect is rare in clinical practice, strategies that combine immune checkpoint blockade with radiation are being studied to overcome immune tolerance or suppression and increase systemic response rates to IO agents. Gynecologic cancers are attractive targets for immune checkpoint blockade, and IO agents may be used in combination with definitive chemoradiotherapy to enhance radiosensitivity and thus local control for unresected disease as well as control distant micrometastatic spread. For patients with metastatic disease, immune checkpoint blockade in combination with stereotactic radiotherapy is being evaluated as a strategy for immune activation and tumor cytoreduction. In this review, we highlight the current use of IO agents in gynecologic cancer, describe the immunogenic potential of radiation through clinical observation and preclinical study, and discuss strategies for combining IO and radiation in reported and ongoing clinical trials.


Assuntos
Neoplasias dos Genitais Femininos/terapia , Sistema Imunitário/efeitos da radiação , Imunoterapia/métodos , Animais , Terapia Combinada , Feminino , Neoplasias dos Genitais Femininos/imunologia , Neoplasias dos Genitais Femininos/radioterapia , Humanos , Tolerância a Radiação/imunologia , Ensaios Clínicos Controlados Aleatórios como Assunto
11.
Nutr Cancer ; 71(8): 1325-1334, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30990087

RESUMO

The topical problem is to find new, more effective and safe treatments for cancer. The purpose of the present work was to study the combined effects of low-intensity extremely high-frequency electromagnetic radiation (EHF EMR) and consumption of n-3 polyunsaturated fatty acids (PUFAs) on tumor growth and the content of FAs in the thymus and tumor tissue in mice. Fatty acid composition was determined using gas chromatography. Exposure of tumor-bearing mice with solid Ehrlich carcinoma to EHF EMR with effective parameters (42.2 GHz, 0.1 mW/cm2, 20 min daily for 5 consecutive days beginning on the first day after the tumor inoculation) led to delaying the tumor growth and restored the content of almost all FAs in thymic tissue to the level of intact animals. Animal intake of the preparation enriched with n-3 PUFAs increased the content of n-3 PUFAs in thymic tissue significantly, but did not affect the tumor growth, even in combination with EHF EMR exposure. Combined action of EHF EMR exposure and n-3 preparation promoted recovery of thymus weight in tumor-bearing animals. The data obtained assume a complex interaction between the immune system and the tumor, and the important role of FAs in the regulation of this interaction.


Assuntos
Carcinoma de Ehrlich/terapia , Radiação Eletromagnética , Ácidos Graxos Ômega-3/metabolismo , Timo/metabolismo , Animais , Carcinoma de Ehrlich/metabolismo , Carcinoma de Ehrlich/patologia , Ácidos Graxos Ômega-3/administração & dosagem , Ácidos Graxos Ômega-3/análise , Sistema Imunitário/efeitos da radiação , Masculino , Camundongos , Timo/efeitos da radiação
12.
Am J Perinatol ; 36(3): 252-257, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30081405

RESUMO

Phototherapy in neonates for treatment of pathological jaundice is an effective therapeutic tool that is widely used in neonatal units. Over the past years, a greater concern has emerged about the effects on the immune and inflammatory system and its potential genotoxic and side effects, especially the late ones, possibly associated with childhood diseases, showing that this treatment is not as harmless as previously believed. Numerous studies assessing these possible adverse effects of phototherapy on neonates have been published over the past years. Through this review, we seek to analyze what we know about the side effects of phototherapy in the neonatal period. The main causes of jaundice, phototherapy techniques, acute and late side effects, and effects on the immune and inflammatory system were reviewed. It was concluded that phototherapy is not a treatment free of side effects and further studies need to be conducted to elucidate its harmful effects on neonates.


Assuntos
Icterícia Neonatal/terapia , Fototerapia/efeitos adversos , Permeabilidade do Canal Arterial/etiologia , Humanos , Sistema Imunitário/efeitos da radiação , Recém-Nascido , Recém-Nascido Prematuro , Doenças do Prematuro/etiologia , Doenças do Prematuro/terapia , Fototerapia/instrumentação , Fototerapia/métodos , Pele/efeitos da radiação
13.
Int J Mol Sci ; 20(13)2019 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-31261963

RESUMO

Radiotherapy (RT), besides cancer cells, also affects the tumor microenvironment (TME): tumor blood vessels and cells of the immune system. It damages endothelial cells and causes radiation-induced inflammation. Damaged vessels inhibit the infiltration of CD8+ T lymphocytes into tumors, and immunosuppressive pathways are activated. They lead to the accumulation of radioresistant suppressor cells, including tumor-associated macrophages (TAMs) with the M2 phenotype, myeloid-derived suppressor cells (MDSCs), and regulatory T cells (Tregs). The area of tumor hypoxia increases. Hypoxia reduces oxygen-dependent DNA damage and weakens the anti-cancer RT effect. It activates the formation of new blood vessels and leads to cancer relapse after irradiation. Irradiation may also activate the immune response through immunogenic cell death induction. This leads to the "in situ" vaccination effect. In this article, we review how changes in the TME affect radiation-induced anticancer efficacy. There is a very delicate balance between the activation of the immune system and the immunosuppression induced by RT. The effects of RT doses on immune system reactions and also on tumor vascularization remain unclear. A better understanding of these interactions will contribute to the optimization of RT treatment, which may prevent the recurrence of cancer.


Assuntos
Sistema Imunitário/efeitos da radiação , Neoplasias/radioterapia , Radioterapia/efeitos adversos , Microambiente Tumoral/efeitos da radiação , Animais , Humanos , Neoplasias/imunologia , Neoplasias/patologia , Radioterapia/métodos , Microambiente Tumoral/imunologia
14.
Semin Cancer Biol ; 46: 138-145, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28412456

RESUMO

Numerous plant products have been used to prevent and manage a wide variety of diseases for centuries. These products are now considered as promising options for the development of more effective and less toxic alternatives to the systems of medicine developed primarily in developed countries in the modern era. Grape seed proanthocyanidins (GSPs) are of great interest due to their anti-carcinogenic effects that have been demonstrated using various tumor models including ultraviolet (UV) radiation-induced non-melanoma skin cancer. In a pre-clinical mouse model supplementation of a control diet (AIN76A) with GSPs at concentrations of 0.2% and 0.5% (w/w) significantly inhibits the growth and multiplicity of UVB radiation-induced skin tumors. In this review, we summarize the evidence that this inhibition of UVB-induced skin tumor development by dietary GSPs is mediated by a multiplicity of coordinated effects including: (i) Promotion of the repair of damaged DNA by nuclear excision repair mechanisms, and (ii) DNA repair-dependent stimulation of the immune system following the functional activation of dendritic cells and effector T cells. Dietary GSPs hold promise for the development of an effective alternative strategy for the prevention of excessive solar UVB radiation exposure-induced skin diseases including the risk of non-melanoma skin cancer in humans.


Assuntos
Reparo do DNA/efeitos dos fármacos , Sistema Imunitário/efeitos dos fármacos , Proantocianidinas/uso terapêutico , Neoplasias Cutâneas/dietoterapia , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/efeitos da radiação , Reparo do DNA/efeitos da radiação , Humanos , Sistema Imunitário/efeitos da radiação , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/patologia , Neoplasias Cutâneas/radioterapia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Raios Ultravioleta/efeitos adversos
15.
Photochem Photobiol Sci ; 17(12): 1872-1884, 2018 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-29136080

RESUMO

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


Assuntos
Tolerância Imunológica/efeitos da radiação , Neoplasias Cutâneas/etiologia , Raios Ultravioleta , Animais , Antígeno B7-H1/química , Antígeno B7-H1/metabolismo , Células Dendríticas/metabolismo , Células Dendríticas/efeitos da radiação , Humanos , Sistema Imunitário/metabolismo , Sistema Imunitário/efeitos da radiação , Neuropeptídeos/metabolismo , Receptores de Citocinas/agonistas , Receptores de Citocinas/metabolismo , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/metabolismo
16.
Cancer Immunol Immunother ; 66(7): 819-832, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28361232

RESUMO

The cancer immunoediting hypothesis assumes that the immune system guards the host against the incipient cancer, but also "edits" the immunogenicity of surviving neoplastic cells and supports remodeling of tumor microenvironment towards an immunosuppressive and pro-neoplastic state. Local irradiation of tumors during standard radiotherapy, by killing neoplastic cells and generating inflammation, stimulates anti-cancer immunity and/or partially reverses cancer-promoting immunosuppression. These effects are induced by moderate (0.1-2.0 Gy) or high (>2 Gy) doses of ionizing radiation which can also harm normal tissues, impede immune functions, and increase the risk of secondary neoplasms. In contrast, such complications do not occur with exposures to low doses (≤0.1 Gy for acute irradiation or ≤0.1 mGy/min dose rate for chronic exposures) of low-LET ionizing radiation. Furthermore, considerable evidence indicates that such low-level radiation (LLR) exposures retard the development of neoplasms in humans and experimental animals. Here, we review immunosuppressive mechanisms induced by growing tumors as well as immunomodulatory effects of LLR evidently or likely associated with cancer-inhibiting outcomes of such exposures. We also offer suggestions how LLR may restore and/or stimulate effective anti-tumor immunity during the more advanced stages of carcinogenesis. We postulate that, based on epidemiological and experimental data amassed over the last few decades, whole- or half-body irradiations with LLR should be systematically examined for its potential to be a viable immunotherapeutic treatment option for patients with systemic cancer.


Assuntos
Carcinogênese/efeitos da radiação , Sistema Imunitário/efeitos da radiação , Tolerância Imunológica/efeitos da radiação , Terapia de Imunossupressão/métodos , Neoplasias/imunologia , Neoplasias/radioterapia , Animais , Carcinogênese/imunologia , Relação Dose-Resposta à Radiação , Humanos , Sistema Imunitário/imunologia , Inflamação/patologia , Camundongos , Neoplasias/patologia , Ratos , Microambiente Tumoral/imunologia , Microambiente Tumoral/efeitos da radiação , Irradiação Corporal Total
17.
J Immunol ; 194(3): 1178-89, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25539818

RESUMO

Radiation exposure induces cell and tissue damage, causing local and systemic inflammatory responses. Because the inflammasome pathway is triggered by cell death and danger-associated molecular patterns, we hypothesized that the inflammasome may signal acute and chronic immune responses to radiation. Using a mouse radiation model, we show that radiation induces a dose-dependent increase in inflammasome activation in macrophages, dendritic cells, NK cells, T cells, and B cells as judged by cleaved caspase-1 detection in cells. Time course analysis showed the appearance of cleaved caspase-1 in cells by day 1 and sustained expression until day 7 after radiation. Also, cells showing inflammasome activation coexpressed the cell surface apoptosis marker annexin V. The role of caspase-1 as a trigger for hematopoietic cell losses after radiation was studied in caspase-1(-/-) mice. We found less radiation-induced cell apoptosis and immune cell loss in caspase-1(-/-) mice than in control mice. Next, we tested whether uric acid might mediate inflammasome activation in cells by treating mice with allopurinol and discovered that allopurinol treatment completely blocked caspase-1 activation in cells. Finally, we demonstrate that radiation-induced caspase-1 activation occurs by a Nod-like receptor family protein 3-independent mechanism because radiation-exposed Nlrp3(-/-) mice showed caspase-1 activation profiles that were indistinguishable from those of wild-type mice. In summary, our data demonstrate that inflammasome activation occurs in many immune cell types following radiation exposure and that allopurinol prevented radiation-induced inflammasome activation. These results suggest that targeting the inflammasome may help control radiation-induced inflammation.


Assuntos
Sistema Imunitário/fisiologia , Sistema Imunitário/efeitos da radiação , Inflamassomos/metabolismo , Transdução de Sinais/efeitos da radiação , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Caspase 1/deficiência , Caspase 1/genética , Morte Celular/imunologia , Morte Celular/efeitos da radiação , Sobrevivência Celular/genética , Sobrevivência Celular/imunologia , Sobrevivência Celular/efeitos da radiação , Citocinas/sangue , Relação Dose-Resposta à Radiação , Ativação Enzimática/efeitos da radiação , Masculino , Camundongos , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR , Lesões por Radiação/imunologia , Lesões por Radiação/metabolismo , Baço/citologia , Baço/imunologia , Baço/efeitos da radiação , Ácido Úrico/metabolismo
19.
Can J Physiol Pharmacol ; 95(2): 151-156, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27901344

RESUMO

There are considerable public concerns about the relationship between mobile phone radiation and human health. The present study assesses the effect of electromagnetic field (EMF) emitted from a mobile phone on the immune system in rats and the possible protective role of vitamin D. Rats were randomly divided into six groups: Group I: control group; Group II: received vitamin D (1000 IU/kg/day) orally; Group III: exposed to EMF 1 h/day; Group IV: exposed to EMF 2 h/day; Group V: exposed to EMF 1 h/day and received vitamin D (1000 IU/kg/day); Group VI: exposed to EMF 2 h/day and received vitamin D (1000 IU/kg/day). After 30 days of exposure time, 1 h/day EMF exposure resulted in significant decrease in immunoglobulin levels (IgA, IgE, IgM, and IgG); total leukocyte, lymphocyte, eosinophil and basophil counts; and a significant increase in neutrophil and monocyte counts. These changes were more increased in the group exposed to 2 h/day EMF. Vitamin D supplementation in EMF-exposed rats reversed these results when compared with EMF-exposed groups. In contrast, 7, 14, and 21 days of EMF exposure produced nonsignificant differences in these parameters among all experimental groups. We concluded that exposure to mobile phone radiation compromises the immune system of rats, and vitamin D appears to have a protective effect.


Assuntos
Telefone Celular , Radiação Eletromagnética , Sistema Imunitário/efeitos dos fármacos , Sistema Imunitário/efeitos da radiação , Vitamina D/farmacologia , Animais , Basófilos/citologia , Basófilos/efeitos dos fármacos , Eosinófilos/citologia , Eosinófilos/efeitos dos fármacos , Sistema Imunitário/citologia , Imunoglobulinas/sangue , Imunoglobulinas/efeitos da radiação , Contagem de Leucócitos , Leucócitos/citologia , Leucócitos/efeitos dos fármacos , Contagem de Linfócitos , Masculino , Monócitos/citologia , Monócitos/efeitos dos fármacos , Neutrófilos/citologia , Neutrófilos/efeitos dos fármacos , Ratos
20.
Int J Mol Sci ; 18(2)2017 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-28134809

RESUMO

The interrelationship between ionizing radiation and the immune system is complex, multifactorial, and dependent on radiation dose/quality and immune cell type. High-dose radiation usually results in immune suppression. On the contrary, low-dose radiation (LDR) modulates a variety of immune responses that have exhibited the properties of immune hormesis. Although the underlying molecular mechanism is not fully understood yet, LDR has been used clinically for the treatment of autoimmune diseases and malignant tumors. These advancements in preclinical and clinical studies suggest that LDR-mediated immune modulation is a well-orchestrated phenomenon with clinical potential. We summarize recent developments in the understanding of LDR-mediated immune modulation, with an emphasis on its potential clinical applications.


Assuntos
Hormese/efeitos da radiação , Sistema Imunitário/efeitos da radiação , Animais , Relação Dose-Resposta à Radiação , Humanos , Modelos Biológicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA