Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Theranostics ; 13(14): 4711-4729, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37771768

RESUMO

Background: The introduction of magnetic resonance (MR)-guided radiation treatment planning has opened a new space for theranostic nanoparticles to reduce acute toxicity while improving local control. In this work, second-generation AGuIX® nanoparticles (AGuIX-Bi) are synthesized and validated. AGuIX-Bi are shown to maintain MR positive contrast while further amplifying the radiation dose by the replacement of some Gd3+ cations with higher Z Bi3+. These next-generation nanoparticles are based on the AGuIX® platform, which is currently being evaluated in multiple Phase II clinical trials in combination with radiotherapy. Methods: In this clinically scalable methodology, AGuIX® is used as an initial chelation platform to exchange Gd3+ for Bi3+. AGuIX-Bi nanoparticles are synthesized with three ratios of Gd/Bi, each maintaining MR contrast while further amplifying radiation dose relative to Bi3+. Safety, efficacy, and theranostic potential of the nanoparticles were evaluated in vitro and in vivo in a human non-small cell lung cancer model. Results: We demonstrated that increasing Bi3+ in the nanoparticles is associated with more DNA damage and improves in vivo efficacy with a statistically significant delay in tumor growth and 33% complete regression for the largest Bi/Gd ratio tested. The addition of Bi3+ by our synthetic method leads to nanoparticles that present slightly altered pharmacokinetics and lengthening of the period of high tumor accumulation with no observed evidence of toxicity. Conclusions: We confirmed the safety and enhanced efficacy of AGuIX-Bi with radiation therapy at the selected ratio of 30Gd/70Bi. These results provide crucial evidence towards patient translation.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Nanopartículas , Humanos , Medicina de Precisão , Meios de Contraste , Imageamento por Ressonância Magnética/métodos , Doses de Radiação , Nanomedicina Teranóstica/métodos
2.
Cancer Res ; 83(20): 3442-3461, 2023 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-37470810

RESUMO

Although external beam radiotherapy (xRT) is commonly used to treat central nervous system (CNS) tumors in patients of all ages, young children treated with xRT frequently experience life-altering and dose-limiting neurocognitive impairment (NI) while adults do not. The lack of understanding of mechanisms responsible for these differences has impeded the development of neuroprotective treatments. Using a newly developed mouse model of xRT-induced NI, we found that neurocognitive function is impaired by ionizing radiation in a dose- and age-dependent manner, with the youngest animals being most affected. Histologic analysis revealed xRT-driven neuronal degeneration and cell death in neurogenic brain regions in young animals but not adults. BH3 profiling showed that neural stem and progenitor cells, neurons, and astrocytes in young mice are highly primed for apoptosis, rendering them hypersensitive to genotoxic damage. Analysis of single-cell RNA sequencing data revealed that neural cell vulnerability stems from heightened expression of proapoptotic genes including BAX, which is associated with developmental and mitogenic signaling by MYC. xRT induced apoptosis in primed neural cells by triggering a p53- and PUMA-initiated, proapoptotic feedback loop requiring cleavage of BID and culminating in BAX oligomerization and caspase activation. Notably, loss of BAX protected against apoptosis induced by proapoptotic signaling in vitro and prevented xRT-induced apoptosis in neural cells in vivo as well as neurocognitive sequelae. On the basis of these findings, preventing xRT-induced apoptosis specifically in immature neural cells by blocking BAX, BIM, or BID via direct or upstream mechanisms is expected to ameliorate NI in pediatric patients with CNS tumor. SIGNIFICANCE: Age- and differentiation-dependent apoptotic priming plays a pivotal role in driving radiotherapy-induced neurocognitive impairment and can be targeted for neuroprotection in pediatric patients.


Assuntos
Proteínas Reguladoras de Apoptose , Apoptose , Animais , Criança , Pré-Escolar , Humanos , Camundongos , Apoptose/fisiologia , Proteínas Reguladoras de Apoptose/metabolismo , Proteína X Associada a bcl-2/metabolismo , Morte Celular , Transdução de Sinais , Proteína Supressora de Tumor p53/genética
3.
Front Public Health ; 10: 947068, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36483259

RESUMO

Aircrew (consisting of flight attendants, pilots, or flight engineers/navigators) are exposed to cosmic ionizing radiation (CIR) at flight altitude, which originates from solar activity and galactic sources. These exposures accumulate over time and are considerably higher for aircrew compared to the general population, and even higher compared to U.S. radiation workers. Many epidemiological studies on aircrew have observed higher rates of specific cancers compared to the general population. Despite high levels of CIR exposure and elevated rates of cancer in aircrew, a causal link between CIR and cancer has yet to be established. Many challenges still exist in effectively studying this relationship, not the least of which is evaluating CIR exposure separately from the constellation of factors that occur as part of the flight environment. This review concentrates on cancer incidence and mortality observed among aircrew in epidemiologic studies in relation to CIR exposure and limitation trends observed across the literature. The aim of this review is to provide an updated comprehensive summary of the literature that will support future research by identifying epidemiological challenges and highlighting existing increased cancer concerns in an occupation where CIR exposure is anticipated to increase in the future.


Assuntos
Neoplasias , Exposição à Radiação , Humanos , Exposição à Radiação/efeitos adversos , Neoplasias/epidemiologia , Neoplasias/etiologia
4.
NanoImpact ; 25: 100379, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35559885

RESUMO

Nano-enabled, toner-based printing equipment emit nanoparticles during operation. The bioactivity of these nanoparticles as documented in a plethora of published toxicological studies raises concerns about their potential health effects. These include pro-inflammatory effects that can lead to adverse epigenetic alterations and cardiovascular disorders in rats. At the same time, their potential to alter DNA repair pathways at realistic doses remains unclear. In this study, size-fractionated, airborne particles from a printer center in Singapore were sampled and characterized. The PM0.1 size fraction (particles with an aerodynamic diameter less than 100 nm) of printer center particles (PCP) were then administered to human lung adenocarcinoma (Calu-3) or lymphoblastoid (TK6) cells. We evaluated plasma membrane integrity, mitochondrial activity, and intracellular reactive oxygen species (ROS) generation. Moreover, we quantified DNA damage and alterations in the cells' capacity to repair 6 distinct types of DNA lesions. Results show that PCP altered the ability of Calu-3 cells to repair 8oxoG:C lesions and perform nucleotide excision repair, in the absence of acute cytotoxicity or DNA damage. Alterations in DNA repair capacity have been correlated with the risk of various diseases, including cancer, therefore further genotoxicity studies are needed to assess the potential risks of PCP exposure, at both occupational settings and at the end-consumer level.


Assuntos
Células Epiteliais , Nanopartículas , Animais , Dano ao DNA , Reparo do DNA , Humanos , Nanopartículas/toxicidade , Ratos , Espécies Reativas de Oxigênio/metabolismo
5.
ACS Nano ; 15(3): 4728-4746, 2021 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-33710878

RESUMO

The potential genotoxic effects of engineered nanomaterials (ENMs) may occur through the induction of DNA damage or the disruption of DNA repair processes. Inefficient DNA repair may lead to the accumulation of DNA lesions and has been linked to various diseases, including cancer. Most studies so far have focused on understanding the nanogenotoxicity of ENM-induced damages to DNA, whereas the effects on DNA repair have been widely overlooked. The recently developed fluorescence multiplex-host-cell reactivation (FM-HCR) assay allows for the direct quantification of multiple DNA repair pathways in living cells and offers a great opportunity to address this methodological gap. Herein an FM-HCR-based method is developed to screen the impact of ENMs on six major DNA repair pathways using suspended or adherent cells. The sensitivity and efficiency of this DNA repair screening method were demonstrated in case studies using primary human small airway epithelial cells and TK6 cells exposed to various model ENMs (CuO, ZnO, and Ga2O3) at subcytotoxic doses. It was shown that ENMs may inhibit nucleotide-excision repair, base-excision repair, and the repair of oxidative damage by DNA glycosylases in TK6 cells, even in the absence of significant genomic DNA damage. It is of note that the DNA repair capacity was increased by some ENMs, whereas it was suppressed by others. Overall, this method can be part of a multitier, in vitro hazard assessment of ENMs as a functional, high-throughput platform that provides insights into the interplay of the properties of ENMs, the DNA repair efficiency, and the genomic stability.


Assuntos
Nanopartículas , Nanoestruturas , Dano ao DNA , Reparo do DNA , Ensaios de Triagem em Larga Escala , Humanos
6.
Hematol Oncol Stem Cell Ther ; 14(4): 267-274, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33626329

RESUMO

Increasing trend in oral cancer (0.6% per year) and its related mortality has been reported worldwide since 2010. The United States alone reports an increase of 57% within the past 10 years. This emphasizes the need not only for designing strategies of prevention and planning but also for an effective treatment regime for the various oral cancers. Cancers of the lips, tongue, cheeks, floor of the mouth, and hard palate have been primarily classified under the category of oral cancers. If left undiagnosed, these cancers can be life threatening. Amongst these, the most undesignated and understudied cancer type is the lip carcinoma, which is either categorized under oral cancer or/as well as skin cancer or head and neck cancer. However, lip cancer corresponds to 25-30% of all diagnosed oral cancers. Though the etiology of lip cancer is not yet fully understood, numerous risk factors involved in its development are now being studied. The cells in the lip region are continuously exposed to various DNA damaging agents from endogenous as well as exogenous sources. Flaws in DNA repair mechanisms involved in eliminating these damages may be linked to the origin of carcinogenesis. Accumulation of DNA damage and defect in repair mechanisms may play a role in lip carcinogenesis and progression. This literature review is an exhaustive compilation of the research work performed on the role of DNA damage and repair responses in lip carcinoma which will pave a path for researchers to identify predictive DNA repair biomarker/s for lip cancer, and its diagnosis, prevention, and treatment.


Assuntos
Neoplasias Labiais , Carcinogênese , Dano ao DNA , Humanos , Neoplasias Labiais/genética
7.
Biol Reprod ; 104(1): 58-70, 2021 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-32902600

RESUMO

There has been a significant annual increase in the number of cases of uterine leiomyomas or fibroids (UF) among women of all races and ages across the world. A fortune is usually spent by the healthcare sector for fibroid-related treatments and management. Molecular studies have established the higher mutational heterogeneity in UF as compared to normal myometrial cells. The contribution of DNA damage and defects in repair responses further increases the mutational burden on the cells. This in turn leads to genetic instability, associated with cancer risk and other adverse reproductive health outcomes. Such and many more growing bodies of literature have highlighted the genetic/molecular, biochemical and clinical aspects of UF; none the less there appear to be a lacuna bridging the bench to bed gap in addressing and preventing this disease. Presented here is an exhaustive review of not only the molecular mechanisms underlying the predisposition to the disease but also possible strategies to effectively diagnose, prevent, manage, and treat this disease.


Assuntos
Dano ao DNA , Reparo do DNA , Leiomioma/genética , Neoplasias Uterinas/genética , Feminino , Humanos , Leiomioma/metabolismo , Leiomioma/patologia , Miométrio/patologia , Neoplasias Uterinas/metabolismo , Neoplasias Uterinas/patologia
8.
Oral Oncol ; 108: 104804, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32474390

RESUMO

Ameloblastoma is a rare human disease of benign neoplasm odontogenic tumor with a lower prevalence but higher recurrence rate. Etiology of ameloblastoma is not fully understood thus lacks implementation of curative treatments. One of the proposed models of evolution of ameloblastoma is related to alteration in DNA damage and repair effects. Growing body of literature has associated defect in DNA damage and repair mechanisms with cancer risk and various adverse health outcomes in humans. Persistent defect of repair and escape of these genomic unstable cells from cell death mechanisms can contribute towards accumulation of oncogene driver or tumor suppressor mutations selective for malignant transformations. In addition, growth, progression and survival of tumor depends upon its DNA repair mechanisms too, thus identifying a DNA repair biomarker can be of advantageous to eliminate the tumor. Understanding the interconnection of oral lesion and role of various DNA repair mechanisms in context to ameloblastoma will assist to build up a platform for translational based research. This study is a literature review of research work published up to date in the field of ameloblastoma in regard to DNA damage and repair effects.


Assuntos
Ameloblastoma/genética , Dano ao DNA/genética , Humanos
9.
Environ Mol Mutagen ; 61(5): 551-559, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32324932

RESUMO

Radio-adaptive response (RAR) is a biological mechanism, where cells primed with a low dose exhibit reduced DNA damage with a high challenging dose. Single nucleotide polymorphisms (SNPs) of DNA repair genes including base excision repair (BER) pathway are known to be associated with radio-sensitivity but involvement in RAR is not yet understood. In the present study, attempt was made to correlate genotype frequencies of four BER SNPs [hOGG1(Ser326Cys), XRCC1(Arg399Gln), APE1(Asp148Glu) and LIGASE1(A/C)] with DNA damage, repair and mRNA expression level among 20 healthy donors (12 adaptive and 8 nonadaptive). Our results revealed that LIGASE1 (p = .002) showed significant correlation with DNA damage and mRNA expression level with increasing dose. hOGG1 (Ser326Cys), XRCC1 (Arg399Gln) and LIGASE1(A/C) polymorphisms showed significant difference with DNA damage (%T) and mRNA expression profile in primed cells among adaptive donors. In conclusion, BER gene polymorphisms play important role in identifying donors with radio-sensitivity and RAR in human cells.


Assuntos
DNA Glicosilases/genética , DNA Ligase Dependente de ATP/genética , Reparo do DNA/genética , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/genética , Raios gama , Leucócitos Mononucleares/metabolismo , Tolerância a Radiação/efeitos da radiação , Proteína 1 Complementadora Cruzada de Reparo de Raio-X/genética , Dano ao DNA/genética , DNA Glicosilases/sangue , DNA Ligase Dependente de ATP/sangue , Reparo do DNA/efeitos da radiação , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/sangue , Regulação da Expressão Gênica , Frequência do Gene/genética , Genótipo , Humanos , Leucócitos Mononucleares/efeitos da radiação , Polimorfismo de Nucleotídeo Único/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Tolerância a Radiação/genética , Proteína 1 Complementadora Cruzada de Reparo de Raio-X/sangue
11.
Cell Commun Signal ; 18(1): 36, 2020 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-32138738

RESUMO

BACKGROUND: The current successful clinical use of agents promoting robust anti-tumor immunity in cancer patients warrants noting that radiation therapy (RT) induces immunogenic cell death (ICD) of tumor cells, which can generate anti-tumor immune responses. However, breast cancer stem cells (BCSCs) are resistant to RT and RT alone usually failed to mount an anti-tumor immune response. METHODS: High aldehyde dehydrogenase activity (ALDH)bright and CD44+/CD24-/ESA+ cancer cells, previously shown to have BCSC properties, were isolated from human MDA-MB-231 and UACC-812 breast cancer cell lines by flow cytometer. Flow sorted BCSCs and non-BCSCs were further tested for their characteristic of stemness by mammosphere formation assay. Induction of ICD in BCSCs vs. non-BCSCs in response to different in vitro treatments was determined by assessing cell apoptosis and a panel of damage-associated molecular pattern molecules (DAMPs) by flow and enzyme-linked immunosorbent assay (ELISA). RESULTS: We found that ionizing radiation (IR) triggered a lower level of ICD in BCSCs than non-BCSCs. We then investigated the ability of disulfiram/cooper (DSF/Cu) which is known to preferentially induce cancer stem cells (CSCs) apoptosis to enhance IR-induced ICD of BCSCs. The results indicate that DSF/Cu induced a similar extent of IDC in both BCSCs and non-BCSCs and rendered IR-resistant BCSCs as sensitive as non-BCSCs to IR-induced ICD. IR and DSF/Cu induced ICD of BCSCs could be partly reversed by pre-treatment of BCSCs with a reactive oxygen species (ROS) scavenger and XBP1s inhibitors. CONCLUSION: DSF/Cu rendered IR-resistant BCSCs as sensitive as non-BCSCs to IR-induced ICD. Our data demonstrate the potential of IR and DSF/Cu to induce ICD in BCSCs and non-BCSCs leading to robust immune responses against not only differentiated/differentiating breast cancer cells but also BCSCs, the root cause of cancer formation, progression and metastasis.


Assuntos
Antineoplásicos , Neoplasias da Mama/tratamento farmacológico , Dissulfiram , Reposicionamento de Medicamentos , Morte Celular Imunogênica/efeitos dos fármacos , Tolerância a Radiação/efeitos dos fármacos , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacologia , Neoplasias da Mama/patologia , Neoplasias da Mama/radioterapia , Linhagem Celular Tumoral , Dissulfiram/administração & dosagem , Dissulfiram/farmacologia , Feminino , Humanos , Células-Tronco Neoplásicas
12.
Front Public Health ; 8: 590412, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33392133

RESUMO

During air travel, flight crew (flight attendants, pilots) can be exposed to numerous flight-related environmental DNA damaging agents that may be at the root of an excess risk of cancer and other diseases. This already complex mix of exposures is now joined by SARS-CoV-2, the virus that causes COVID-19. The complex exposures experienced during air travel present a challenge to public health research, but also provide an opportunity to consider new strategies for understanding and countering their health effects. In this article, we focus on threats to genomic integrity that occur during air travel and discuss how these threats and our ability to respond to them may influence the risk of SARS-CoV-2 infection and the development of range of severity of the symptoms. We also discuss how the virus itself may lead to compromised genome integrity. We argue that dauntingly complex public health problems, such as the challenge of protecting flight crews from COVID-19, must be met with interdisciplinary research teams that include epidemiologists, engineers, and mechanistic biologists.


Assuntos
Viagem Aérea/estatística & dados numéricos , COVID-19/genética , COVID-19/transmissão , Dano ao DNA , Resistência à Doença/genética , Genoma , Exposição Ocupacional/estatística & dados numéricos , Adulto , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Fatores de Risco , SARS-CoV-2
13.
Mutagenesis ; 30(5): 663-76, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25958388

RESUMO

Radio-adaptive response is a mechanism whereby a low-dose exposure (priming dose) induces resistance to a higher dose (challenging dose) thus significantly reducing its detrimental effects. Radiation-induced DNA damage gets repaired through various DNA repair pathways in human cells depending upon the type of lesion. The base excision repair (BER) pathway repairs radiation-induced base damage, abasic sites and single-strand breaks in cellular DNA. In the present study, an attempt has been made to investigate the involvement of BER genes and proteins in the radio-adaptive response in human resting peripheral blood mononuclear cells (PBMC). Venous blood samples were collected from 20 randomly selected healthy male individuals with written informed consent. PBMC were isolated and irradiated at a priming dose of 0.1 Gy followed 4h later with a challenging dose of 2.0 Gy (primed cells). Quantitation of DNA damage was done using the alkaline comet assay immediately and expression profile of BER genes and proteins were studied 30 min after the challenging dose using real-time quantitative polymerase chain reaction and western blot, respectively. The overall result showed significant (P ≤ 0.05) reduction of DNA damage in terms of percentage of DNA in tail (%T) with a priming dose of 0.1 Gy followed by a challenging dose of 2.0 Gy after 4 h. Twelve individuals showed significant (P ≤ 0.05) reduction in %T whereas eight individuals showed marginal reduction in DNA damage that was not statistically significant. However, at the transcriptional level, BER genes such as APE1, FEN1 and LIGASE1 showed significant (P ≤ 0.05) up-regulation in both groups. Significant (P ≤ 0.05) up-regulation was also observed at the protein level for OGG1, APE1, MBD4, FEN1 and LIGASE1 in primed cells. Up-regulation of some BER genes and proteins such as APE1, FEN1 and LIGASE1 in primed cells of resting PBMC is suggestive of active involvement of the BER pathway in radio-adaptive response.


Assuntos
Dano ao DNA , Enzimas Reparadoras do DNA/metabolismo , Reparo do DNA , Raios gama , Leucócitos Mononucleares/efeitos da radiação , Adaptação Fisiológica , Adulto , Ensaio Cometa , DNA/metabolismo , DNA/efeitos da radiação , Enzimas Reparadoras do DNA/genética , Humanos , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/fisiologia , Masculino , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA