Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.312
Filtrar
1.
Theranostics ; 13(12): 4121-4137, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37554266

RESUMO

Background: Due to the immunosuppressive tumor microenvironment (TME), radiation therapy (RT)-mediated immune response is far from satisfactory. How to improve the efficacy of immunogenic RT by priming strong immunogenic cell death (ICD) is an interesting and urgent challenge. Methods: A polyacrylic acid-coated core-shell UiO@Mn3O4 (denoted as UMP) nanocomposite is constructed for immunogenic RT via multiple strategies. Results: Reshaping the TME via Mn3O4-mediated integration of O2 production, GSH depletion, ROS generation and cell cycle arrest, accompanied by Hf-based UiO-mediated radiation absorption, eventually amplifies UMP-mediated RT to induce intense ICD. With the potent ICD induction and reprogrammed tumor-associated macrophages, this synergetic strategy can promote dendritic cells maturation and CD8+ T cells infiltration, and potentiate anti-tumor immunity against primary, distant, and metastatic tumors. Conclusion: This work is expected to shed light on the immunosuppressive TME-reshaping via multiple strategies to reinforce the immunogenic RT outcome and facilitate the development of effective cancer nanomedicine.


Assuntos
Morte Celular , Nanomedicina , Nanoestruturas , Neoplasias , Animais , Humanos , Camundongos , Linfócitos T CD8-Positivos/imunologia , Pontos de Checagem do Ciclo Celular , Morte Celular/imunologia , Morte Celular/efeitos da radiação , Linhagem Celular Tumoral , Células Dendríticas/imunologia , Glutationa/metabolismo , Camundongos Endogâmicos BALB C , Nanomedicina/métodos , Nanoestruturas/química , Nanoestruturas/uso terapêutico , Metástase Neoplásica/imunologia , Neoplasias/imunologia , Neoplasias/patologia , Neoplasias/radioterapia , Oxigênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Microambiente Tumoral , Macrófagos Associados a Tumor/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Nature ; 618(7966): 834-841, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37286599

RESUMO

Tumours most often arise from progression of precursor clones within a single anatomical niche. In the bone marrow, clonal progenitors can undergo malignant transformation to acute leukaemia, or differentiate into immune cells that contribute to disease pathology in peripheral tissues1-4. Outside the marrow, these clones are potentially exposed to a variety of tissue-specific mutational processes, although the consequences of this are unclear. Here we investigate the development of blastic plasmacytoid dendritic cell neoplasm (BPDCN)-an unusual form of acute leukaemia that often presents with malignant cells isolated to the skin5. Using tumour phylogenomics and single-cell transcriptomics with genotyping, we find that BPDCN arises from clonal (premalignant) haematopoietic precursors in the bone marrow. We observe that BPDCN skin tumours first develop at sun-exposed anatomical sites and are distinguished by clonally expanded mutations induced by ultraviolet (UV) radiation. A reconstruction of tumour phylogenies reveals that UV damage can precede the acquisition of alterations associated with malignant transformation, implicating sun exposure of plasmacytoid dendritic cells or committed precursors during BPDCN pathogenesis. Functionally, we find that loss-of-function mutations in Tet2, the most common premalignant alteration in BPDCN, confer resistance to UV-induced cell death in plasmacytoid, but not conventional, dendritic cells, suggesting a context-dependent tumour-suppressive role for TET2. These findings demonstrate how tissue-specific environmental exposures at distant anatomical sites can shape the evolution of premalignant clones to disseminated cancer.


Assuntos
Transformação Celular Neoplásica , Células Dendríticas , Leucemia Mieloide Aguda , Neoplasias Cutâneas , Pele , Raios Ultravioleta , Humanos , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Células da Medula Óssea/efeitos da radiação , Morte Celular/efeitos da radiação , Linhagem da Célula/genética , Linhagem da Célula/efeitos da radiação , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Transformação Celular Neoplásica/efeitos da radiação , Células Clonais/metabolismo , Células Clonais/patologia , Células Clonais/efeitos da radiação , Células Dendríticas/metabolismo , Células Dendríticas/patologia , Células Dendríticas/efeitos da radiação , Leucemia Mieloide Aguda/etiologia , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Mutação/efeitos da radiação , Especificidade de Órgãos , Análise da Expressão Gênica de Célula Única , Neoplasias Cutâneas/etiologia , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Raios Ultravioleta/efeitos adversos , Pele/patologia , Pele/efeitos da radiação
3.
PLoS One ; 18(5): e0277759, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37130114

RESUMO

Ultrasound-stimulated microbubbles (USMB) cause localized vascular effects and sensitize tumors to radiation therapy (XRT). We investigated acoustic parameter optimization for combining USMB and XRT. We treated breast cancer xenograft tumors with 500 kHz pulsed ultrasound at varying pressures (570 or 740 kPa), durations (1 to 10 minutes), and microbubble concentrations (0.01 to 1% (v/v)). Radiation therapy (2 Gy) was administered immediately or after a 6-hour delay. Histological staining of tumors 24 hours after treatment detected changes in cell morphology, cell death, and microvascular density. Significant cell death resulted at 570 kPa after a 1-minute exposure with 1% (v/v) microbubbles with or without XRT. However, significant microvascular disruption required higher ultrasound pressure and exposure duration greater than 5 minutes. Introducing a 6-hour delay between treatments (USMB and XRT) showed a similar tumor effect with no further improvement in response as compared to when XRT was delivered immediately after USMB.


Assuntos
Neoplasias da Mama , Neoplasias Mamárias Animais , Terapia por Ultrassom , Animais , Humanos , Feminino , Neoplasias da Mama/radioterapia , Neoplasias da Mama/patologia , Terapia por Ultrassom/métodos , Microbolhas , Morte Celular/efeitos da radiação , Ultrassonografia
4.
Health Phys ; 123(5): 376-386, 2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-36069830

RESUMO

ABSTRACT: With rapid technical advances, ionizing radiation has been put into wider application in ordinary living, with the worst cytological effect on the human body being cell death. Moreover, according to the Nomenclature Committee on Cell Death, the method of radiation-induced cell death, usually classified as interphase and proliferative death, undergoes more detailed classifications oriented by its molecular mechanism. Elaborating its mode and molecular mechanism is crucial for the protection and treatment of radiation injury, as well as the radiotherapy and recovery of tumors. Varying with the changes of the radiation dose and the environment, the diverse targets and pathways of ionizing radiation result in various cell deaths. This review focuses on classifications of radiation-induced cell death and its molecular mechanism. We also examine the main characteristics of ionizing radiation-induced cell death. The modes of radiation-induced cell death can be classified as apoptosis, necrosis, autophagy-dependent cell death, pyroptosis, ferroptosis, immunogenic cell death, and non-lethal processes. Once the dose is high enough, radiation effects mostly appear as destructiveness ("destructiveness" is used to describe a situation in which cells do not have the opportunity to undergo a routine death process, in which case high-dose radiation works like a physical attack). This breaks up or even shatters cells, making it difficult to find responses of the cell itself. Due to diversities concerning cell phenotypes, phases of cell cycle, radiation dose, and even cellular subregions, various methods of cell death occur, which are difficult to identify and classify. Additionally, the existence of common initial activation and signaling molecules among all kinds of cell deaths, as well as sophisticated crossways in cellular molecules, makes it more laborious to distinguish and classify various cell deaths.


Assuntos
Apoptose , Neoplasias , Apoptose/efeitos da radiação , Morte Celular/efeitos da radiação , Humanos , Neoplasias/radioterapia , Radiação Ionizante , Transdução de Sinais/efeitos da radiação
5.
Nature ; 606(7913): 351-357, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35545677

RESUMO

Death is defined as the irreversible cessation of circulatory, respiratory or brain activity. Many peripheral human organs can be transplanted from deceased donors using protocols to optimize viability. However, tissues from the central nervous system rapidly lose viability after circulation ceases1,2, impeding their potential for transplantation. The time course and mechanisms causing neuronal death and the potential for revival remain poorly defined. Here, using the retina as a model of the central nervous system, we systemically examine the kinetics of death and neuronal revival. We demonstrate the swift decline of neuronal signalling and identify conditions for reviving synchronous in vivo-like trans-synaptic transmission in postmortem mouse and human retina. We measure light-evoked responses in human macular photoreceptors in eyes removed up to 5 h after death and identify modifiable factors that drive reversible and irreversible loss of light signalling after death. Finally, we quantify the rate-limiting deactivation reaction of phototransduction, a model G protein signalling cascade, in peripheral and macular human and macaque retina. Our approach will have broad applications and impact by enabling transformative studies in the human central nervous system, raising questions about the irreversibility of neuronal cell death, and providing new avenues for visual rehabilitation.


Assuntos
Transdução de Sinal Luminoso , Reabilitação Neurológica , Mudanças Depois da Morte , Retina , Animais , Autopsia , Morte Celular/efeitos da radiação , Sistema Nervoso Central/efeitos da radiação , Humanos , Transdução de Sinal Luminoso/efeitos da radiação , Macaca , Camundongos , Retina/metabolismo , Retina/efeitos da radiação , Fatores de Tempo
6.
J Biol Chem ; 298(4): 101776, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35227760

RESUMO

Zyxin is a zinc-binding phosphoprotein known to regulate cell migration, adhesion, and cell survival. Zyxin also plays a role in signal transduction between focal adhesions and the nuclear compartment. However, the mechanism of Zyxin shuttling to nucleus is still unclear. Here, we identify that the GlcNAc transferase (O-linked GlcNAc [O-GlcNAc] transferase) can O-GlcNAcylate Zyxin and regulate its nuclear localization. We show that O-GlcNAc transferase O-GlcNAcylates Zyxin at two residues, serine 169 (Ser-169) and Ser-246. In addition, O-GlcNAcylation of Ser-169, but not Ser-246, enhances its interaction with 14-3-3γ, which is a phosphoserine/threonine-binding protein and is reported to bind with phosphorylated Zyxin. Furthermore, we found that 14-3-3γ could promote the nuclear localization of Zyxin after Ser-169 O-GlcNAcylation by affecting the function of the N-terminal nuclear export signal sequence; functionally, UV treatment increases the O-GlcNAcylation of Zyxin, which may enhance the nuclear location of Zyxin. Finally, Zyxin in the nucleus maintains homeodomain-interacting protein kinase 2 stability and promotes UV-induced cell death. In conclusion, we uncover that the nuclear localization of Zyxin can be regulated by its O-GlcNAcylation, and that this protein may regulate UV-induced cell death.


Assuntos
Morte Celular , Adesões Focais , N-Acetilglucosaminiltransferases/metabolismo , Transporte Proteico , Zixina , Morte Celular/genética , Morte Celular/efeitos da radiação , Adesões Focais/metabolismo , N-Acetilglucosaminiltransferases/genética , Serina , Zixina/genética , Zixina/metabolismo
7.
Exp Cell Res ; 410(1): 112946, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34826424

RESUMO

The principle underlying radiotherapy is to kill cancer cells while minimizing the harmful effects on non-cancer cells, which has still remained as a major challenge. In relation, ferroptosis has recently been proposed as a novel mechanism of radiation-induced cell death. In this study, we investigated and demonstrated the role of Hemin as an iron overloading agent in the generation of reactive oxygen species (ROS) induced by ionizing radiation in lung cancer and non-cancer cells. It was found that the presence of Hemin in irradiated lung cancer cells enhanced the productivity of initial ROS, resulting in lipid peroxidation and subsequent ferroptosis. We observed that application of Hemin as a co-treatment increased the activity of GPx4 degradation in both cancer and normal lung cells. Furthermore, Hemin protected normal lung cells against radiation-induced cell death, in that it suppressed ROS after radiation, and boosted the production of bilirubin which was a lipophilic ROS antioxidant. In addition, we demonstrated significant FTH1 expression in normal lung cells when compared to lung cancer cells, which prevented iron from playing a role in increasing IR-induced cell death. Our findings demonstrated that Hemin had a dual function in enhancing the radiosensitivity of ferroptosis in lung cancer cells while promoting cell survival in normal lung cells.


Assuntos
Ferritinas/genética , Hemina/farmacologia , Neoplasias Pulmonares/radioterapia , Oxirredutases/genética , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/genética , Células A549 , Animais , Morte Celular/efeitos dos fármacos , Morte Celular/efeitos da radiação , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Ferroptose/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Hemina/química , Xenoenxertos , Humanos , Ferro/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Camundongos , Tolerância a Radiação/efeitos dos fármacos , Radiação Ionizante , Espécies Reativas de Oxigênio/metabolismo
8.
Bull Exp Biol Med ; 172(1): 22-25, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34792714

RESUMO

We performed a comparative study of the colony-forming ability and the number of residual foci of DNA repair proteins in cultured human lung fibroblasts (MRC-5 cell line) after exposure to subpicosecond beams of accelerated electrons with an energy of 3.6 MeV and quasi-continuous radiation (accelerated electrons with an energy of 4 MeV and X-rays). The yield of damages causing reproductive cell death after pulsed subpicosecond radiation exposure was higher by ~1.8 times than after quasi-continuous radiation exposure. The quantitative yield of residual γH2AX foci (phosphorylated H2AX histone, a protein marker of DNA double breaks) in cells irradiated with subpicosecond beams of accelerated electrons was shown to be ~2.0- 2.5-fold higher than in cells irradiated with quasi-continuous beams of accelerated electrons.


Assuntos
Morte Celular/efeitos da radiação , Proliferação de Células/efeitos da radiação , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Enzimas Reparadoras do DNA/metabolismo , Fibroblastos/efeitos da radiação , Linhagem Celular , Elétrons , Histonas/metabolismo , Humanos , Pulmão/citologia , Pulmão/efeitos da radiação
9.
Int J Mol Sci ; 22(21)2021 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-34769035

RESUMO

Background: The invention of non-ionizing emission devices revolutionized science, medicine, industry, and the military. Currently, different laser systems are commonly used, generating the potential threat of excessive radiation exposure, which can lead to adverse health effects. Skin is the organ most exposed to laser irradiation; therefore, this study aims to evaluate the effects of 445 nm, 520 nm, and 638 nm non-ionizing irradiation on keratinocytes and fibroblasts. Methods: Keratinocytes and fibroblasts were exposed to a different fluency of 445 nm, 520 nm, and 638 nm laser irradiation. In addition, viability, type of cell death, cell cycle distribution, and proliferation rates were investigated. Results: The 445 nm irradiation was cytotoxic to BJ-5ta (≥58.7 J/cm2) but not to Ker-CT cells. Exposure influenced the cell cycle distribution of Ker-CT (≥61.2 J/cm2) and BJ-5ta (≥27.6 J/cm2) cells, as well as the Bj-5ta proliferation rate (≥50.5 J/cm2). The 520 nm irradiation was cytotoxic to BJ-5ta (≥468.4 J/cm2) and Ker-CT (≥385.7 J/cm2) cells. Cell cycle distribution (≥27.6 J/cm2) of Ker-CT cells was also affected. The 638 nm irradiation was cytotoxic to BJ-5ta and Ker-CT cells (≥151.5 J/cm2). The proliferation rate and cell cycle distribution of BJ-5ta (≥192.9 J/cm2) and Ker-CT (13.8 and 41.3 J/cm2) cells were also affected. Conclusions: At high fluences, 455 nm, 520 nm, and 638 nm irradiation, representing blue, green, and red light spectra, are hazardous to keratinocytes and fibroblasts. However, laser irradiation may benefit the cells at low fluences by modulating the cell cycle and proliferation rate.


Assuntos
Fibroblastos/efeitos da radiação , Pele/efeitos da radiação , Ciclo Celular/efeitos da radiação , Morte Celular/efeitos da radiação , Proliferação de Células/efeitos da radiação , Sobrevivência Celular/efeitos da radiação , Células Cultivadas , Humanos , Lasers , Luz , Terapia com Luz de Baixa Intensidade/métodos
10.
Int J Mol Sci ; 22(22)2021 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-34830482

RESUMO

Radiotherapy promotes tumor cell death and senescence through the induction of oxidative damage. Recent work has highlighted the importance of lipid peroxidation for radiotherapy efficacy. Excessive lipid peroxidation can promote ferroptosis, a regulated form of cell death. In this review, we address the evidence supporting a role of ferroptosis in response to radiotherapy and discuss the molecular regulators that underlie this interaction. Finally, we postulate on the clinical implications for the intersection of ferroptosis and radiotherapy.


Assuntos
Metabolismo dos Lipídeos/efeitos da radiação , Peroxidação de Lipídeos/efeitos da radiação , Neoplasias/radioterapia , Morte Celular/efeitos da radiação , Senescência Celular/genética , Senescência Celular/efeitos da radiação , Ferroptose/genética , Ferroptose/efeitos da radiação , Humanos , Metabolismo dos Lipídeos/genética , Neoplasias/genética , Neoplasias/patologia , Estresse Oxidativo/efeitos da radiação
11.
Cells ; 10(11)2021 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-34831123

RESUMO

Fluoroquinolones cause phototoxic reactions, manifested as different types of skin lesions, including hyperpigmentation. The disturbances of melanogenesis indicate that fluoroquinolones may affect cellular processes in melanocytes. It has been reported that these antibiotics may bind with melanin and accumulate in pigmented cells. The study aimed to examine the changes in melanogenesis in human normal melanocytes exposed to UVA radiation and treated with lomefloxacin and moxifloxacin, the most and the least fluoroquinolone, respectively. The obtained results demonstrated that both tested fluoroquinolones inhibited melanogenesis through a decrease in tyrosinase activity and down-regulation of tyrosinase and microphthalmia-associated transcription factor production. Only lomefloxacin potentiated UVA-induced melanogenesis. Under UVA irradiation lomefloxacin significantly enhanced melanin content and tyrosinase activity in melanocytes, although the drug did not cause an increased expression of tyrosinase or microphthalmia-associated transcription factor. The current studies revealed that phototoxic activity of fluoroquinolones is associated with alterations in the melanogenesis process. The difference in phototoxic potential of fluoroquinolones derivatives may be connected with various effects on UVA-induced events at a cellular level.


Assuntos
Fluoroquinolonas/farmacologia , Melaninas/biossíntese , Melanócitos/metabolismo , Raios Ultravioleta , Morte Celular/efeitos dos fármacos , Morte Celular/efeitos da radiação , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Fluoroquinolonas/química , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos da radiação , Humanos , Melanócitos/efeitos dos fármacos , Melanócitos/efeitos da radiação , Fator de Transcrição Associado à Microftalmia/genética , Fator de Transcrição Associado à Microftalmia/metabolismo , Monofenol Mono-Oxigenase/genética , Monofenol Mono-Oxigenase/metabolismo , Moxifloxacina/química , Moxifloxacina/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
12.
Genes (Basel) ; 12(6)2021 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-34204447

RESUMO

(1) Kinase inhibitors (KI) targeting components of the DNA damage repair pathway are a promising new type of drug. Combining them with ionizing radiation therapy (IR), which is commonly used for treatment of head and neck tumors, could improve tumor control, but could also increase negative side effects on surrounding normal tissue. (2) The effect of KI of the DDR (ATMi: AZD0156; ATRi: VE-822, dual DNA-PKi/mTORi: CC-115) in combination with IR on HPV-positive and HPV-negative HNSCC and healthy skin cells was analyzed. Cell death and cell cycle arrest were determined using flow cytometry. Additionally, clonogenic survival and migration were analyzed. (3) Studied HNSCC cell lines reacted differently to DDRi. An increase in cell death for all of the malignant cells could be observed when combining IR and KI. Healthy fibroblasts were not affected by simultaneous treatment. Migration was partially impaired. Influence on the cell cycle varied between the cell lines and inhibitors; (4) In conclusion, a combination of DDRi with IR could be feasible for patients with HNSCC. Side effects on healthy cells are expected to be limited to normal radiation-induced response. Formation of metastases could be decreased because cell migration is impaired partially. The treatment outcome for HPV-negative tumors tends to be improved by combined treatment.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Morte Celular/efeitos dos fármacos , Neoplasias de Cabeça e Pescoço/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Raios X , Proteínas Mutadas de Ataxia Telangiectasia/antagonistas & inibidores , Morte Celular/efeitos da radiação , Linhagem Celular Tumoral , Células Cultivadas , Reparo do DNA/efeitos dos fármacos , Proteína Quinase Ativada por DNA/antagonistas & inibidores , Humanos , Isoxazóis/farmacologia , Pirazinas/farmacologia , Piridinas/farmacologia , Quinolinas/farmacologia , Triazóis/farmacologia
13.
Cells ; 10(6)2021 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-34201238

RESUMO

Multimodal tumor treatment settings consisting of radiotherapy and immunomodulating agents such as immune checkpoint inhibitors are more and more commonly applied in clinics. In this context, the immune phenotype of tumor cells has a major influence on the anti-tumor immune response as well as the composition of the tumor microenvironment. A promising approach to further boost anti-tumor immune responses is to add hyperthermia (HT), i.e., heating the tumor tissue between 39 °C to 45 °C for 60 min. One key technique is the use of radiative hyperthermia systems. However, knowledge is limited as to how the frequency of the used radiative systems affects the immune phenotype of the treated tumor cells. By using our self-designed in vitro hyperthermia system, we compared cell death induction and expression of immune checkpoint molecules (ICM) on the tumor cell surface of murine B16 melanoma and human MDA-MB-231 and MCF-7 breast cancer cells following HT treatment with clinically relevant microwaves at 915 MHz or 2.45 GHz alone, radiotherapy (RT; 2 × 5 Gy or 5 × 2 Gy) alone or in combination (RHT). At 44 °C, HT alone was the dominant cell death inductor with inactivation rates of around 70% for B16, 45% for MDA-MB-231 and 35% for MCF-7 at 915 MHz and 80%, 60% and 50% at 2.45 GHz, respectively. Additional RT resulted in 5-15% higher levels of dead cells. The expression of ICM on tumor cells showed time-, treatment-, cell line- and frequency-dependent effects and was highest for RHT. Computer simulations of an exemplary spherical cell revealed frequency-dependent local energy absorption. The frequency of hyperthermia systems is a newly identified parameter that could also affect the immune phenotype of tumor cells and consequently the immunogenicity of tumors.


Assuntos
Morte Celular/efeitos da radiação , Hipertermia Induzida/métodos , Micro-Ondas/uso terapêutico , Neoplasias/radioterapia , Animais , Terapia Combinada , Humanos , Células MCF-7 , Melanoma Experimental , Camundongos
14.
Photochem Photobiol Sci ; 20(5): 639-652, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33978941

RESUMO

Solar UV radiation consists of both UVA and UVB. The wavelength-specific molecular responses to UV radiation have been studied, but the interaction between UVA and UVB has not been well understood. In this study, we found that long-wavelength UVA, UVA1, augmented UVB-induced cell death, and examined the underlying mechanisms. Human keratinocytes HaCaT were exposed to UVA1, followed by UVB irradiation. Irradiation by UVA1 alone showed no effect on cell survival, whereas the UVA1 pre-irradiation remarkably enhanced UVB-induced cell death. UVA1 delayed the repair of pyrimidine dimers formed by UVB and the accumulation of nucleotide excision repair (NER) proteins to damaged sites. Gap synthesis during NER was also decreased, suggesting that UVA1 delayed NER, and unrepaired pyrimidine dimers and single-strand breaks generated in the process of NER were left behind. Accumulation of this unrepaired DNA damage might have led to the formation of DNA double-strand breaks (DSBs), as was detected using gel electrophoresis analysis and phosphorylated histone H2AX assay. Combined exposure enhanced the ATM-Chk2 signaling pathway, but not the ATR-Chk1 pathway, confirming the enhanced formation of DSBs. Moreover, UVA1 suppressed the UVB-induced phosphorylation of Akt, a survival signal pathway. These results indicated that UVA1 influenced the repair of UVB-induced DNA damage, which resulted in the formation of DSBs and enhanced cell death, suggesting the risk of simultaneous exposure to high doses of UVA1 and UVB.


Assuntos
Queratinócitos/patologia , Raios Ultravioleta , Morte Celular/efeitos da radiação , Sobrevivência Celular/efeitos da radiação , Células Cultivadas , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Humanos , Queratinócitos/efeitos da radiação
15.
Genes Cells ; 26(6): 447-454, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33848395

RESUMO

UHRF1 (Ubiquitin-like with PHD and ring finger domains 1) regulates DNA methylation and histone modifications and plays a key role in cell proliferation and the DNA damage response. However, the function of UHRF2, a paralog of UHRF1, in the DNA damage response remains largely unknown. Here, we show that UHRF2 is essential for maintaining cell viability after UV irradiation, as well as for the proliferation of cancer cells. UHRF2 was found to physically interact with ATR in a DNA damage-dependent manner through UHRF2's TTD domain. In addition, phosphorylation of threonine at position 1989, which is required for UV-induced activation of ATR, was impaired in cells depleted of UHRF2, suggesting that UHRF2 is essential in ATR activation. In conclusion, these results suggest a new regulatory mechanism of ATR activation mediated by UHRF2.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Raios Ultravioleta , Morte Celular/efeitos da radiação , Linhagem Celular Tumoral , Proliferação de Células/efeitos da radiação , Quinase 1 do Ponto de Checagem/metabolismo , Dano ao DNA , Humanos , Ligação Proteica/efeitos da radiação
16.
Radiat Res ; 195(5): 441-451, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33721021

RESUMO

We examined lethal damages of X rays induced by direct and indirect actions, in terms of double-strand break (DSB) repair susceptibility using two kinds of repair-deficient Chinese hamster ovary (CHO) cell lines. These CHO mutants (51D1 and xrs6) are genetically deficient in one of the two important DNA repair pathways after genotoxic injury [homologous recombination (HR) and non-homologous end binding (NHEJ) pathways, respectively]. The contribution of indirect action on cell killing can be estimated by applying the maximum level of dimethylsulfoxide (DMSO) to get rid of OH radicals. To control the proportion of direct and indirect actions in lethal damage, we irradiated CHO mutant cells under aerobic and anoxic conditions. The contributions of indirect action on HR-defective 51D1 cells were 76% and 57% under aerobic and anoxic conditions, respectively. Interestingly, these percentages were similar to those of the wild-type cells even if the radiosensitivity was different. However, the contributions of indirect action to cell killing on NHEJ-defective xrs6 cells were 52% and 33% under aerobic and anoxic conditions, respectively. Cell killing by indirect action was significantly affected by the oxygen concentration and the DSB repair pathways but was not correlated with radiosensitivity. These results suggest that the lethal damage induced by direct action is mostly repaired by NHEJ repair pathway since killing of NHEJ-defective cells has significantly higher contribution by the direct action. In other words, the HR repair pathway may not effectively repair the DSB by direct action in place of the NHEJ repair pathway. We conclude that the type of DSB produced by direct action is different from that of DSB induced by indirect action.


Assuntos
Dano ao DNA , Oxigênio/metabolismo , Aerobiose/genética , Aerobiose/efeitos da radiação , Animais , Células CHO , Morte Celular/genética , Morte Celular/efeitos da radiação , Cricetulus , Reparo do DNA por Junção de Extremidades/efeitos da radiação , Recombinação Homóloga/efeitos da radiação , Raios X/efeitos adversos
17.
Oncol Rep ; 45(4)2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33655336

RESUMO

Radiation therapy is an effective treatment against various types of cancer, but some radiation­resistant cancer cells remain a major therapeutic obstacle; thus, understanding radiation resistance mechanisms is essential for cancer treatment. In this study, we established radiation­resistant colon cancer cell lines and examined the radiation­induced genetic changes associated with radiation resistance. Using RNA­sequencing analysis, collapsin response mediator protein 4 (CRMP4) was identified as the candidate gene associated with radiation sensitivity. When cells were exposed to radiation, intracellular Ca2+ influx, collapse of mitochondrial membrane potential, and cytochrome c release into the cytosol were increased, followed by apoptosis induction. Radiation treatment­ or Ca2+ ionophore A23187­induced apoptosis was significantly inhibited in CRMP4­deficient cells, including radiation­resistant or CRMP4­shRNA cell lines. Furthermore, treatment of CRMP4­deficient cells with low levels (<5 µM) of BAPTA­AM, a Ca2+ chelator, resulted in radiation resistance. Conversely, Ca2+ deficiency induced by a high BAPTA­AM concentration (>10 µM) resulted in higher cell death in the CRMP4­depleted cells compared to CRMP4­expressing control cells. Our results suggest that CRMP4 plays an important role in Ca2+­mediated cell death pathways under radiation exposure and that CRMP4 may be a therapeutical target for colon cancer treatment.


Assuntos
Cálcio/metabolismo , Neoplasias do Colo/metabolismo , Neoplasias do Colo/radioterapia , Proteínas Musculares/metabolismo , Morte Celular/efeitos da radiação , Linhagem Celular Tumoral , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Humanos , Proteínas Musculares/efeitos da radiação , Tolerância a Radiação , Análise de Sequência de RNA , Transdução de Sinais/efeitos da radiação
18.
Int J Mol Sci ; 22(4)2021 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-33669634

RESUMO

Little is known about the effects on hyaluronan (HA) metabolism of UVA radiation. This study demonstrates that the secretion of HA by human dermal fibroblasts (HDFs) is downregulated by UVA, accompanied by the down- and upregulation of mRNA and protein levels of the HA-synthesizing enzyme (HAS2) and the HA-degrading protein, HYaluronan Binding protein Involved in HA Depolymerization(HYBID), respectively. Signaling analysis revealed that the exposure distinctly elicits activation of the p38/MSK1/CREB/c-Fos/AP-1 axis, the JNK/c-Jun axis, and the p38/ATF-2 axis, but downregulates the phosphorylation of NF-kB and JAK/STAT3. A signal inhibition study demonstrated that the inhibition of p38 significantly abrogates the UVA-accentuated mRNA level of HYBID. Furthermore, the inhibition of STAT3 significantly downregulates the level of HAS2 mRNA in non-UVA exposed HDFs. Analysis using siRNAs demonstrated that transfection of ATF-2 siRNA but not c-Fos siRNA abrogates the increased protein level of HYBID in UVA-exposed HDFs. An inhibitor of protein tyrosine phosphatase but not of protein serine/threonine phosphatase restored the diminished phosphorylation level of STAT3 at Tyr 705, accompanied by a significant abolishing effect on the decreased mRNA expression level of HAS2. Silencing with a protein tyrosine phosphatase PTP-Meg2 siRNA revealed that it abrogates the decreased phosphorylation of STAT3 at Tyr 705 in UVA-exposed HDFs. These findings suggest that the UVA-induced decrease in HA secretion by HDFs is attributable to the down- and upregulation of HAS2 and HYBID expression, respectively, changes that are mainly ascribed to the inactivated signaling of the STAT3 axis due to the activated tyrosine protein phosphatase PTP-Meg2 and the activated signaling of the p38/ATF2 axis, respectively.


Assuntos
Regulação para Baixo/efeitos da radiação , Fibroblastos/efeitos da radiação , Hialuronan Sintases/metabolismo , Ácido Hialurônico/metabolismo , Hialuronoglucosaminidase/metabolismo , Transdução de Sinais/efeitos da radiação , Raios Ultravioleta , Regulação para Cima/efeitos da radiação , Fator 2 Ativador da Transcrição/metabolismo , Morte Celular/efeitos dos fármacos , Morte Celular/efeitos da radiação , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Derme/citologia , Regulação para Baixo/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Janus Quinase 2/metabolismo , Masculino , Modelos Biológicos , Peso Molecular , Fosforilação/efeitos dos fármacos , Fosforilação/efeitos da radiação , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fator de Transcrição STAT3/metabolismo , Regulação para Cima/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
19.
Toxicol In Vitro ; 72: 105108, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33545343

RESUMO

Malignant melanoma is the cause of 80% of deaths in skin cancer patients. Treatment of melanoma in the 4th stage of clinical advancement, in which inoperable metastasis occur, does not provide sufficient effects. Ketoprofen has phototoxic properties and it can be used as a new treatment option for skin cancers as a part of photochemotherapy. The present study was designed to investigate whether ketoprofen in combination with UVA induces cytotoxic, anti-proliferative and pro-apoptotic effects on melanoma cells. It was stated that co-treatment with 1.0 mM ketoprofen and UVA irradiation disturbed homeostasis of C32 melanoma cells by lowering its vitality (decrease of GSH level). Contrary to C32 cells, melanocytes showed low sensitivity to ketoprofen and UVA radiation, pointing selectivity in the mode of action towards melanoma cells. Co-treatment with ketoprofen and UVA irradiation has cytotoxic and anti-proliferative and pro-apoptotic effect on C32. The co-treatment triggered the DNA fragmentation and changed the cell cycle in C32 cells. In conclusion, it could be stated that local application of ketoprofen in combination with UVA irradiation may be used to support the treatment of melanoma and creates the possibility of reducing the risk of cancer recurrence and metastasis.


Assuntos
Antineoplásicos/administração & dosagem , Dermatite Fototóxica , Cetoprofeno/administração & dosagem , Melanoma Amelanótico/tratamento farmacológico , Fotoquimioterapia , Neoplasias Cutâneas/tratamento farmacológico , Raios Ultravioleta , Morte Celular/efeitos dos fármacos , Morte Celular/efeitos da radiação , Linhagem Celular Tumoral , Glutationa/metabolismo , Humanos , Melanócitos/efeitos dos fármacos , Melanócitos/efeitos da radiação , Melanoma Amelanótico/genética , Mutação , Proteínas Proto-Oncogênicas B-raf/genética , Neoplasias Cutâneas/genética
20.
Cells ; 10(2)2021 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-33498294

RESUMO

Phototropins are plasma membrane-associated photoreceptors of blue light and UV-A/B radiation. The Arabidopsis thaliana genome encodes two phototropins, PHOT1 and PHOT2, that mediate phototropism, chloroplast positioning, and stomatal opening. They are well characterized in terms of photomorphogenetic processes, but so far, little was known about their involvement in photosynthesis, oxidative stress responses, and cell death. By analyzing phot1, phot2 single, and phot1phot2 double mutants, we demonstrated that both phototropins influence the photochemical and non-photochemical reactions, photosynthetic pigments composition, stomata conductance, and water-use efficiency. After oxidative stress caused by UV-C treatment, phot1 and phot2 single and double mutants showed a significantly reduced accumulation of H2O2 and more efficient photosynthetic electron transport compared to the wild type. However, all phot mutants exhibited higher levels of cell death four days after UV-C treatment, as well as deregulated gene expression. Taken together, our results reveal that on the one hand, both phot1 and phot2 contribute to the inhibition of UV-C-induced foliar cell death, but on the other hand, they also contribute to the maintenance of foliar H2O2 levels and optimal intensity of photochemical reactions and non-photochemical quenching after an exposure to UV-C stress. Our data indicate a novel role for phototropins in the condition-dependent optimization of photosynthesis, growth, and water-use efficiency as well as oxidative stress and cell death response after UV-C exposure.


Assuntos
Proteínas de Arabidopsis/metabolismo , Arabidopsis/fisiologia , Arabidopsis/efeitos da radiação , Estresse Oxidativo/efeitos da radiação , Fotossíntese/efeitos da radiação , Proteínas Serina-Treonina Quinases/metabolismo , Raios Ultravioleta , Antioxidantes/metabolismo , Arabidopsis/genética , Proteínas de Arabidopsis/genética , Biomassa , Morte Celular/efeitos da radiação , Fluorescência , Perfilação da Expressão Gênica , Regulação da Expressão Gênica de Plantas/efeitos da radiação , Homeostase/efeitos da radiação , Peróxido de Hidrogênio/metabolismo , Mutação/genética , Estresse Oxidativo/genética , Pigmentos Biológicos/metabolismo , Folhas de Planta/anatomia & histologia , Folhas de Planta/efeitos da radiação , Estômatos de Plantas/fisiologia , Estômatos de Plantas/efeitos da radiação , Proteínas Serina-Treonina Quinases/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transcriptoma/genética , Transcriptoma/efeitos da radiação , Água
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...