Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Sci Rep ; 12(1): 13691, 2022 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-35953697

RESUMO

Localization in underwater wireless sensor network (UWSN) faces an imminent threat when the triangulating anchor node starts to malfunction. Traditional geometric approaches are insufficient to cope with the survivability of UWSN topology. To address these issues, this paper presents a symplectic geometry for identification of the malicious anchor node. Consequently, a geodesic search algorithm (GSA) based Target localization is proposed which reduces the positioning error by exploiting the phase-space constancy of the underwater acoustic sensor network topology to effectively triangulate the target node despite its mobility. First, a malicious anchor node model is presented. The node movement is expressed in the form of "ripple region". GSA is then proposed which effectively frees the node metastasis from anchor node geometry, thereby making the underwater system more survivable and resilient. Simulation results evaluate the survivability of the geodesic formalism in terms of the reduced penalty incurred by node movement, as well as the reduced impact of anchor node malfunction. An improvement of 13.46% and 9.26% reveals the utility of the geodesic technique in aquamarine sensor deployments, which would be beneficial in underwater resource exploration and defense planning.

2.
Mol Cancer Ther ; 15(12): 2863-2874, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27765854

RESUMO

Although treatment of prostate cancer has improved over the past several years, taxanes, such as cabazitaxel, remain the only form of effective chemotherapy that improves survival in patients with metastatic castration-resistant prostate cancer. However, the effectiveness of this class of drugs has been associated with various side effects and drug resistance. We previously reported that fisetin, a hydroxyflavone, is a microtubule-stabilizing agent and inhibits prostate cancer cell proliferation, migration, and invasion and suggested its use as an adjuvant for treatment of prostate and other cancer types. In this study, we investigated the effect of fisetin in combination with cabazitaxel with the objective to achieve maximum therapeutic benefit, reduce dose and toxicity, and minimize or delay the induction of drug resistance and metastasis. Our data show for the first time that a combination of fisetin (20 µmol/L) enhances cabazitaxel (5 nmol/L) and synergistically reduces 22Rν1, PC-3M-luc-6, and C4-2 cell viability and metastatic properties with minimal adverse effects on normal prostate epithelial cells. In addition, the combination of fisetin with cabazitaxel was associated with inhibition of proliferation and enhancement of apoptosis. Furthermore, combination treatment resulted in the inhibition of tumor growth, invasion, and metastasis when assessed in two in vivo xenograft mouse models. These results provide evidence that fisetin may have therapeutic benefit for patients with advanced prostate cancer through enhancing the efficacy of cabazitaxel under both androgen-dependent and androgen-independent conditions. This study underscores the benefit of the combination of fisetin with cabazitaxel for the treatment of advanced and resistant prostate cancer and possibly other cancer types. Mol Cancer Ther; 15(12); 2863-74. ©2016 AACR.


Assuntos
Antineoplásicos/farmacologia , Flavonoides/farmacologia , Taxoides/farmacologia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Sinergismo Farmacológico , Quimioterapia Combinada , Flavonóis , Humanos , Masculino , Camundongos , Camundongos Nus , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Neoplasias da Próstata/diagnóstico , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Resultado do Tratamento , Ensaio Tumoral de Célula-Tronco , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Oncotarget ; 7(14): 17945-56, 2016 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-26918454

RESUMO

Chronic exposure to ultraviolet radiation (UVR) is linked to the development of cutaneous squamous cell carcinoma (SCC), a non-melanoma form of skin cancer that can metastasize. Tumor necrosis factor-alpha (TNFα), a pro-inflammatory cytokine, is linked to UVR-induced development of SCC. To find clues about the mechanisms by which TNFα may promote UVR-induced development of SCC, we investigated changes in the expression profiling of microRNAs (miRNA), a novel class of short noncoding RNAs, which affects translation and stability of mRNAs. In this experiment, TNFα knockout (TNFα KO) mice and their wild type (WT) littermates were exposed to acute UVR (2.0 kJ/m2) and the expression profiling of epidermal miRNA was determined 4hr post UVR exposure. TNFα deletion in untreated WT mice resulted in differential expression (log fold change>1) of seventeen miRNA. UVR exposure in WT mice induced differential expression of 22 miRNA. However, UVR exposure in TNFα KO mice altered only two miRNAs. Four miRNA, were differentially expressed between WT+UVR and TNFα KO+UVR groups. Differentially expressed selected miRNAs were further validated using real time PCR. Few of the differentially expressed miRNAs (miR-31-5p, miR-196a-5p, miR-127-3p, miR-206-3p, miR-411-5p, miR-709, and miR-322-5p) were also observed in UVR-induced SCC. Finally, bio-informatics analysis using DIANA, MIRANDA, Target Scan, and miRDB algorithms revealed a link with major UVR-induced pathways (MAPK, PI3K-Akt, transcriptional mis-regulation, Wnt, and TGF-beta).


Assuntos
Carcinoma de Células Escamosas/etiologia , Epiderme/metabolismo , MicroRNAs/biossíntese , Lesões Experimentais por Radiação/etiologia , Lesões Experimentais por Radiação/metabolismo , Neoplasias Cutâneas/etiologia , Fator de Necrose Tumoral alfa/biossíntese , Raios Ultravioleta/efeitos adversos , Animais , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Epiderme/efeitos da radiação , Camundongos , Camundongos Knockout , MicroRNAs/genética , Lesões Experimentais por Radiação/genética , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo
4.
Cancer Prev Res (Phila) ; 8(5): 375-86, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25627799

RESUMO

Prostate cancer continues to remain the most common cancer and the second leading cause of cancer-related deaths in American males. The Pten deletions and/or mutations are frequently observed in both primary prostate cancers and metastatic prostate tissue samples. Pten deletion in prostate epithelium in mice results in prostatic intraepithelial neoplasia (PIN), followed by progression to invasive adenocarcinoma. The Pten conditional knockout mice [(Pten-loxp/loxp:PB-Cre4(+)) (Pten-KO)] provide a unique preclinical model to evaluate agents for efficacy for both the prevention and treatment of prostate cancer. We present here for the first time that dietary plumbagin, a medicinal plant-derived naphthoquinone (200 or 500 ppm) inhibits tumor development in intact as well as castrated Pten-KO mice. Plumbagin has shown no signs of toxicity at either of these doses. Plumbagin treatment resulted in a decrease expression of PKCε, AKT, Stat3, and COX2 compared with the control mice. Plumbagin treatment also inhibited the expression of vimentin and slug, the markers of epithelial-to-mesenchymal transition (EMT) in prostate tumors. In summary, the results indicate that dietary plumbagin inhibits growth of both primary and castration-resistant prostate cancer (CRPC) in Pten-KO mice, possibly via inhibition of PKCε, Stat3, AKT, and EMT markers (vimentin and slug), which are linked to the induction and progression of prostate cancer.


Assuntos
Adenocarcinoma/prevenção & controle , Antineoplásicos Fitogênicos/farmacologia , Carcinogênese/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Naftoquinonas/farmacologia , Neoplasias da Próstata/prevenção & controle , Adenocarcinoma/patologia , Animais , Antineoplásicos Fitogênicos/uso terapêutico , Biomarcadores/metabolismo , Carcinogênese/genética , Carcinogênese/metabolismo , Transição Epitelial-Mesenquimal/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Naftoquinonas/uso terapêutico , Orquiectomia , PTEN Fosfo-Hidrolase/genética , Neoplasia Prostática Intraepitelial/tratamento farmacológico , Neoplasia Prostática Intraepitelial/patologia , Neoplasias da Próstata/patologia , Proteína Quinase C-épsilon/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
5.
Antioxid Redox Signal ; 21(5): 682-99, 2014 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-24295217

RESUMO

AIMS: Pancreatic cancer (PC) is the most aggressive malignant disease, ranking as the fourth most leading cause of cancer-related death among men and women in the United States. In this study, we provide evidence of chemotherapeutic effects of α-mangostin, a dietary antioxidant isolated from the pericarp of Garcinia mangostana L. against human PC. RESULTS: The chemotherapeutic effect of α-mangostin was determined using four human PC cells (PL-45, PANC1, BxPC3, and ASPC1). α-Mangostin resulted in a significant inhibition of PC cells viability without having any effects on normal human pancreatic duct epithelial cells. α-Mangostin showed a dose-dependent increase of apoptosis in PC cells. Also, α-mangostin inhibited the expression levels of pNF-κB/p65Ser552, pStat3Ser727, and pStat3Tyr705. α-Mangostin inhibited DNA binding activity of nuclear factor kappa B (NF-κB) and signal transducer and activator 3 (Stat3). α-Mangostin inhibited the expression levels of matrix metallopeptidase 9 (MMP9), cyclin D1, and gp130; however, increased expression of tissue inhibitor of metalloproteinase 1 (TIMP1) was observed in PC cells. In addition, i.p. administration of α-mangostin (6 mg/kg body weight, 5 days a week) resulted in a significant inhibition of both primary (PL-45) and secondary (ASPC1) human PC cell-derived orthotopic and ectopic xenograft tumors in athymic nude mice. No sign of toxicity was observed in any of the mice administered with α-mangostin. α-Mangostin treatment inhibited the biomarkers of cell proliferation (Ki-67 and proliferating cell nuclear antigen [PCNA]) in the xenograft tumor tissues. INNOVATION: We present, for the first time, that dietary antioxidant α-mangostin inhibits the growth of PC cells in vitro and in vivo. CONCLUSION: These results suggest the potential therapeutic efficacy of α-mangostin against human PC.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Antioxidantes/farmacologia , Modelos Animais de Doenças , Garcinia mangostana/química , Neoplasias Pancreáticas/tratamento farmacológico , Xantonas/farmacologia , Animais , Antineoplásicos Fitogênicos/administração & dosagem , Antineoplásicos Fitogênicos/isolamento & purificação , Antioxidantes/administração & dosagem , Antioxidantes/isolamento & purificação , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Injeções Intraperitoneais , Camundongos , Camundongos Nus , Neoplasias Pancreáticas/patologia , Relação Estrutura-Atividade , Xantonas/administração & dosagem , Xantonas/isolamento & purificação , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Int J Cancer ; 131(9): 2175-86, 2012 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-22322442

RESUMO

Pancreatic cancer (PC) is the most aggressive malignant disease, ranks as the fourth most leading cause of cancer-related death among men and women in the United States. We present here that plumbagin (PL), a quinoid constituent isolated from the roots of the medicinal plant Plumbago zeylanica L, inhibits the growth of PC cells both in vitro and in vivo model systems. PL treatment induces apoptosis and inhibits cell viability of PC cells (PANC1, BxPC3 and ASPC1). In addition, i.p. administration of PL (2 mg/kg body weight, 5 days a week) in severe combined immunodeficiency (SCID) mice beginning 3 days after ectopic implantation of PANC1 cells resulted in a significant (P < 0.01) inhibition of both tumor weight and volume. PL treatment inhibited (1) constitutive expression of epidermal growth factor receptor (EGFR), pStat3Tyr705 and pStat3Ser727, (2) DNA binding of Stat3 and (3) physical interaction of EGFR with Stat3, in both cultured PANC1 cells and their xenograft tumors. PL treatment also inhibited phosphorylation and DNA-binding activity of NF-κB in both cultured PC cells (PANC1 and ASPC1) and in PANC1 cells xenograft tumors. Downstream target genes (cyclin D1, MMP9 and Survivin) of Stat3 and NF-κB were similarly inhibited. These results suggest that PL may be used as a novel therapeutic agent against human PC. Published 2012 Wiley-Liss, Inc. This article is a US Government work, and, as such, is in the public domain in the United States of America.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Receptores ErbB/metabolismo , NF-kappa B/metabolismo , Naftoquinonas/farmacologia , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/patologia , Fator de Transcrição STAT3/metabolismo , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ciclina D1/antagonistas & inibidores , Humanos , Proteínas Inibidoras de Apoptose/antagonistas & inibidores , Masculino , Inibidores de Metaloproteinases de Matriz , Camundongos , Camundongos SCID , Fosforilação , Extratos Vegetais/farmacologia , Transdução de Sinais/efeitos dos fármacos , Survivina
7.
Mol Carcinog ; 51(4): 291-302, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21480396

RESUMO

We have reported that protein kinase C epsilon (PKCε) expression level in epidermis dictates the susceptibility of mice to the development of squamous cell carcinomas (SCC) elicited either by repeated exposure to ultraviolet radiation (UVR) or by the DMBA-TPA tumor promotion protocol. To find clues about the mechanism by which PKCε mediates susceptibility to UVR-induced development of SCC, we found that PKCε-over-expressing transgenic mice, as compared to their wild-type littermates, when exposed to UVR, elicit enhanced phosphorylation of Stat3 at Ser727 residues. Stat3 is constitutively activated in SCC and UVR fails to induce SCC in Stat3 mutant mice. Stat3Ser727 phosphorylation is essential for Stat3 transcriptional activity (Cancer Res. 67: 1385, 2007). We now present several novel findings including that PKCε integrates with its downstream partner ERK1/2 to phosphorylate Stat3Ser727. In these experiments, mice were either exposed to UVR (2 kJ/m(2)/dose) emitted by Kodacel-filtered FS-40 sun lamps or treated with TPA (5 nmol). Both UVR and TPA treatment stimulated PKCε-Stat3 interaction, Stat3Ser727 phosphorylation and Stat3-regulated gene COX-2 expression. PKCε-Stat3 interaction and Stat3Ser727 phosphorylation was also observed in SCC elicited by repeated UVR exposures of mice. PKCε-Stat3 interaction was PKCε specific. UVR or TPA-stimulated Stat3Ser727 phosphorylation accompanied interaction of PKCε with ERK1/2 in intact mouse skin in vivo. Deletion of PKCε in wild-type mice attenuated both TPA and UVR-induced expression of phosphoforms of ERK1/2 and Stat3Ser727. These results indicate that PKCε integrates with ERK1/2 to mediate both TPA and UVR-induced epidermal Stat3Ser727 phosphorylation. PKCε and Stat3 may be potential molecular targets for SCC prevention.


Assuntos
Epiderme/enzimologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteína Quinase C-épsilon/metabolismo , Fator de Transcrição STAT3/metabolismo , Acetato de Tetradecanoilforbol/química , Animais , Proliferação de Células , Deleção de Genes , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fosforilação , Ligação Proteica , Proteínas Recombinantes/metabolismo , Raios Ultravioleta
8.
Risk Anal ; 29(5): 662-75, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19302279

RESUMO

A wide range of uncertainties will be introduced inevitably during the process of performing a safety assessment of engineering systems. The impact of all these uncertainties must be addressed if the analysis is to serve as a tool in the decision-making process. Uncertainties present in the components (input parameters of model or basic events) of model output are propagated to quantify its impact in the final results. There are several methods available in the literature, namely, method of moments, discrete probability analysis, Monte Carlo simulation, fuzzy arithmetic, and Dempster-Shafer theory. All the methods are different in terms of characterizing at the component level and also in propagating to the system level. All these methods have different desirable and undesirable features, making them more or less useful in different situations. In the probabilistic framework, which is most widely used, probability distribution is used to characterize uncertainty. However, in situations in which one cannot specify (1) parameter values for input distributions, (2) precise probability distributions (shape), and (3) dependencies between input parameters, these methods have limitations and are found to be not effective. In order to address some of these limitations, the article presents uncertainty analysis in the context of level-1 probabilistic safety assessment (PSA) based on a probability bounds (PB) approach. PB analysis combines probability theory and interval arithmetic to produce probability boxes (p-boxes), structures that allow the comprehensive propagation through calculation in a rigorous way. A practical case study is also carried out with the developed code based on the PB approach and compared with the two-phase Monte Carlo simulation results.


Assuntos
Probabilidade , Segurança , Incerteza , Medição de Risco
9.
J Invest Dermatol ; 129(8): 2011-21, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19194472

RESUMO

Protein kinase C (PKC)epsilon overexpression in FVB/N transgenic mice sensitized skin to UVR-induced development of squamous cell carcinomas and suppressed formation of sunburn cells, which are DNA-damaged keratinocytes undergoing apoptosis. Here, we elucidated the mechanisms associated with the inhibition of UVR-induced appearance of sunburn cells in PKCepsilon transgenic mice. We found that the inhibition of UVR-induced sunburn cell formation in PKCepsilon transgenic mice may be the result of the inhibition of the expression of Fas, Fas ligand, and the mammalian death adaptor protein termed Fas-associated with death domain (FADD). The adaptor protein FADD is the key component of the death-inducing signaling complex of both Fas and tumor necrosis factor receptor 1. A decreased expression of epidermal FADD was observed after a single UVR exposure. However, a complete loss of FADD expression was found after four (Monday, Wednesday, Friday, and Monday) repeated UVR exposures. FADD transmits apoptotic signals from death receptors to the downstream initiator caspase-8 and connects to the mitochondrial intrinsic apoptotic signal transduction pathway by the cleavage of Bid, a Bcl-2 family member. PKCepsilon-mediated loss of FADD expression inhibited UVR signals to the activation of both extrinsic and intrinsic apoptotic pathways.


Assuntos
Apoptose , Proteína de Domínio de Morte Associada a Fas/genética , Proteína Quinase C-épsilon/fisiologia , Receptores Tipo I de Fatores de Necrose Tumoral/fisiologia , Raios Ultravioleta , Receptor fas/fisiologia , Animais , Proteína Agonista de Morte Celular de Domínio Interatuante com BH3/antagonistas & inibidores , Camundongos , Camundongos Transgênicos , Poli(ADP-Ribose) Polimerases/metabolismo , RNA Mensageiro/análise , Transdução de Sinais , Fator de Necrose Tumoral alfa/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA