Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Pharmacol Res ; 160: 105188, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32919041

RESUMO

There has always been a keen interest of basic and clinical researchers to search for cancer therapeutics having minimum off-target effects and maximum anticancer activities. In accordance with this approach, there has been an explosion in the field of natural products research in the past few decades because of extra-ordinary list of natural extracts and their biologically and pharmacologically active constituents having significant medicinal properties. Apparently, luteolin-mediated anticancer effects have been investigated in different cancers but there is superfluousness of superficial data. Generalized scientific evidence encompassing apoptosis, DNA damage and anti-inflammatory effects has been reported extensively. However, how luteolin modulates deregulated oncogenic pathways in different cancers has not been comprehensively uncovered. In this review we have attempted to focus on cutting-edge research which has unveiled remarkable abilities of luteolin to modulate deregulated oncogenic pathways in different cancers. We have partitioned the review into various sections to separately discuss advancements in therapeutic targeting of oncogenic protein networks. We have provided detailed mechanistic insights related to JAK-STAT signaling and summarized how luteolin inhibited STAT proteins to inhibit STAT-driven gene network. We have also individually analyzed Wnt/ß-catenin and NOTCH pathway and how luteolin effectively targeted these pathways. Mapping of the signaling landscape has revealed that NOTCH pathway can be targeted therapeutically. NOTCH pathway was noted to be targeted by luteolin. We have also conceptually analyzed how luteolin restored TRAIL-induced apoptosis in resistant cancers. Luteolin induced an increase in pro-apoptotic proteins and efficiently inhibited anti-apoptotic proteins to induce apoptosis. Luteolin mediated regulation of non-coding RNAs is an exciting and emerging facet. Excitingly, there is sequential and systematic accumulation of clues which have started to shed light on intricate regulation of microRNAs by luteolin in different cancers. Collectively, sophisticated information will enable us to develop a refined understanding of the multi-layered regulation of signaling pathways and non-coding RNAs by luteolin in different cancers.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Luteolina/farmacologia , MicroRNAs/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Neoplasias/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Animais , Antineoplásicos Fitogênicos/uso terapêutico , Apoptose/efeitos dos fármacos , Marcação de Genes , Humanos , Luteolina/uso terapêutico , Receptores Notch/efeitos dos fármacos , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/efeitos dos fármacos , Fatores de Transcrição STAT/efeitos dos fármacos , Serina-Treonina Quinases TOR/efeitos dos fármacos
2.
Pharmacol Res ; 141: 512-520, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30641278

RESUMO

Caloric restriction (CR) has been shown to promote longevity and ameliorate aging-associated diseases, including cancer. Extensive research over recent decades has revealed that CR reduces IGF-1/PI3K/AKT signaling and increases sirtuin signaling. We recently found that CR also enhances ALDOA/DNA-PK/p53 signaling. In the present review, we summarize the molecular mechanisms underlying the modulation of the IGF-1/PI3K/AKT pathway, sirtuin signaling, and the ALDOA/DNA-PK/p53 pathway by CR. We also summarize the evidence concerning the roles of these signaling pathways in carcinogenesis, and discuss how they are regulated by CR. Finally, we discuss the crosstalk between these signaling pathways.


Assuntos
Restrição Calórica , Carcinogênese/metabolismo , Neoplasias/dietoterapia , Neoplasias/metabolismo , Transdução de Sinais , Animais , Restrição Calórica/métodos , Proteína Quinase Ativada por DNA/metabolismo , Frutose-Bifosfato Aldolase/metabolismo , Humanos , Fator de Crescimento Insulin-Like I/metabolismo , Neoplasias/prevenção & controle , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Sirtuínas/metabolismo , Proteína Supressora de Tumor p53/metabolismo
3.
Carcinogenesis ; 39(12): 1477-1487, 2018 12 31.
Artigo em Inglês | MEDLINE | ID: mdl-30256916

RESUMO

MicroRNA-34a (miR-34a) behaves as a tumor suppressor by decreasing the expression of oncogenes involved in multiple carcinogenic pathways. Intravenous delivery of miR-34a mimics has been investigated in clinical trials as a potential treatment for advanced cancers; however, the effect of miR-34a on cancer immune surveillance is controversial. In the current study, we found that miR-34a plays a dual role in the regulation of major histocompatibility complex class I-related sequence B (MICB) protein, a ligand of the NKG2D receptor. MiR-34a could both induce and reduce MICB expression by upregulating ataxia telangiectasia and Rad3-related (ATR) protein kinase and downregulating the transcription factor E2F1, respectively. The net effect of miR-34a on MICB expression depended on endogenous E2F1 levels. Overexpression of miR-34a promoted MICB expression in hepatocytes and hepatocellular carcinoma (HCC) cells that have low E2F1 levels but not in HCC cells that have high E2F1 levels. In HCC patients, the expression of miR-34a and MICB showed positive correlation in paratumor liver tissues, which have low E2F1 levels, but not in HCC tissues, which have high E2F1 levels. We showed that miR-34a overexpression in non-transformed liver cells enhanced cytolysis and interferon-γ production by NK-92MI cells. Furthermore, higher miR-34a expression in tumor and paratumor tissues was associated with positive and negative outcomes, respectively, in HCC patients. Our findings suggest that miR-34a induces MICB expression in paratumor liver tissues, which may cause liver damage and serious cytokine release syndrome, thus disclosing potential side effects of systemic administration of miR-34a in anticancer therapy.


Assuntos
Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Hepatócitos/patologia , Antígenos de Histocompatibilidade Classe I/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , MicroRNAs/genética , Proteínas Mutadas de Ataxia Telangiectasia/genética , Carcinogênese/genética , Carcinogênese/patologia , Linhagem Celular Tumoral , Regulação para Baixo/genética , Fator de Transcrição E2F1/genética , Regulação Neoplásica da Expressão Gênica/genética , Células Hep G2 , Humanos , Interferon gama/genética , Células Matadoras Naturais , Oncogenes/genética , Regulação para Cima/genética
4.
Nucleic Acids Res ; 44(8): 3629-42, 2016 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-26704979

RESUMO

Dicer participates in heterochromatin formation in fission yeast and plants. However, whether it has a similar role in mammals remains controversial. Here we showed that the human Dicer protein interacts with SIRT7, an NAD(+)-dependent H3K18Ac (acetylated lysine 18 of histone H3) deacetylase, and holds a proportion of SIRT7 in the cytoplasm. Dicer knockdown led to an increase of chromatin-associated SIRT7 and simultaneously a decrease of cytoplasmic SIRT7, while its overexpression induced SIRT7 reduction in the chromatin-associated fraction and increment in the cytoplasm. Furthermore, DNA damaging agents promoted Dicer expression, leading to decreased level of chromatin-associated SIRT7 and increased level of H3K18Ac, which can be alleviated by Dicer knockdown. Taken together with that H3K18Ac was exclusively associated with the chromatin, our findings suggest that Dicer induction by DNA damaging treatments prevents H3K18Ac deacetylation, probably by trapping more SIRT7 in the cytoplasm.


Assuntos
RNA Helicases DEAD-box/metabolismo , Dano ao DNA , Histonas/metabolismo , Ribonuclease III/metabolismo , Sirtuínas/metabolismo , Linhagem Celular , Cromatina/metabolismo , Cisplatino/toxicidade , RNA Helicases DEAD-box/antagonistas & inibidores , Doxorrubicina/toxicidade , Células HEK293 , Humanos , Radiação Ionizante , Ribonuclease III/antagonistas & inibidores
5.
Carcinogenesis ; 38(9): 873-882, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28911000

RESUMO

DNA double-strand break (DSB) repair is an important mechanism underlying chemotherapy resistance in human cancers. Dicer participates in DSB repair by facilitating homologous recombination. However, whether Dicer is involved in non-homologous end joining (NHEJ) remains unknown. Here, we addressed whether Dicer regulates NHEJ and chemosensitivity in colon cancer cells. Using our recently developed NHEJ assay, we found that DSB introduction by I-SceI cleavage leads to Dicer upregulation. Dicer knockdown increased SIRT7 binding and decreased the level of H3K18Ac (acetylated lysine 18 of histone H3) at DSB sites, thereby repressing the recruitment of NHEJ factors to DSB sites and inhibiting NHEJ. Dicer overexpression reduced SIRT7 binding and increased the level of H3K18Ac at DSB sites, promoting the recruitment of NHEJ factors to DSBs and moderately enhancing NHEJ. Dicer knockdown and overexpression increased and decreased, respectively, the chemosensitivity of colon cancer cells. Dicer protein expression in colon cancer tissues of patients was directly correlated with chemoresistance. Our findings revealed a function of Dicer in NHEJ-mediated DSB repair and the association of Dicer expression with chemoresistance in colon cancer patients.


Assuntos
Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/genética , RNA Helicases DEAD-box/fisiologia , Reparo do DNA por Junção de Extremidades/genética , Resistencia a Medicamentos Antineoplásicos/genética , Ribonuclease III/fisiologia , Animais , RNA Helicases DEAD-box/genética , Quebras de DNA de Cadeia Dupla , Desoxirribonucleases de Sítio Específico do Tipo II/metabolismo , Feminino , Técnicas de Silenciamento de Genes , Células HCT116 , Células HEK293 , Histonas/metabolismo , Humanos , Estimativa de Kaplan-Meier , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias , RNA Interferente Pequeno/genética , Ribonuclease III/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Sirtuínas/genética , Sirtuínas/metabolismo
6.
J Biol Chem ; 289(20): 14225-38, 2014 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-24648519

RESUMO

RRAD (Ras-related associated with diabetes) is a small Ras-related GTPase that is frequently inactivated by DNA methylation of the CpG island in its promoter region in cancer tissues. However, the role of the methylation-induced RRAD inactivation in tumorigenesis remains unclear. In this study, the Ras-regulated transcriptome and epigenome were profiled by comparing T29H (a Ras(V12)-transformed human ovarian epithelial cell line) with T29 (an immortalized but non-transformed cell line) through reduced representation bisulfite sequencing and digital gene expression. We found that Ras(V12)-mediated oncogenic transformation was accompanied by RRAD promoter hypermethylation and a concomitant loss of RRAD expression. In addition, we found that the RRAD promoter was hypermethylated, and its transcription was reduced in ovarian cancer versus normal ovarian tissues. Treatment with the DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine resulted in demethylation in the RRAD promoter and restored RRAD expression in T29H cells. Additionally, treatment with farnesyltransferase inhibitor FTI277 resulted in restored RRAD expression and inhibited DNA methytransferase expression and activity in T29H cells. By employing knockdown and overexpression techniques in T29 and T29H, respectively, we found that RRAD inhibited glucose uptake and lactate production by repressing the expression of glucose transporters. Finally, RRAD overexpression in T29H cells inhibited tumor formation in nude mice, suggesting that RRAD is a tumor suppressor gene. Our results indicate that Ras(V12)-mediated oncogenic transformation induces RRAD epigenetic inactivation, which in turn promotes glucose uptake and may contribute to ovarian cancer tumorigenesis.


Assuntos
Inativação Gênica , Glucose/metabolismo , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Proteínas ras/deficiência , Proteínas ras/genética , Adulto , Idoso , Animais , Transporte Biológico/genética , Carcinogênese/genética , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Metilação de DNA/genética , Células Epiteliais/patologia , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Ácido Láctico/biossíntese , Camundongos , Pessoa de Meia-Idade , Neoplasias Ovarianas/patologia
7.
Carcinogenesis ; 35(1): 155-63, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23917076

RESUMO

Hepatitis B surface antigen (HBsAg) seropositivity is an important risk factor for hepatocellular carcinoma (HCC), and HBsAg-transgenic mice have been reported to spontaneously develop HCC. The major histocompatibility complex class I-related molecules A and B (MICA and MICB) are NKG2D ligands that play important roles in tumor immune surveillance. In the present study, we found that HBsAg overexpression in HepG2 cells led to upregulation of 133 and downregulation of 9 microRNAs (miRNAs). Interestingly, several HBsAg-induced miRNAs repressed the expression of MICA and MICB via targeting their 3'-untranslated regions. In addition, the expression of MICA and MICB was significantly reduced upon HBsAg overexpression, which was partially restored by inhibiting the activities of HBsAg-induced miRNAs. Moreover, HBsAg-overexpressing HCC cells exhibited reduced sensitivity to natural killer cell-mediated cytolysis. Taken together, our data suggest that HBsAg supresses the expression of MICA and MICB via induction of cellular miRNAs, thereby preventing NKG2D-mediated elimination of HCC cells.


Assuntos
Carcinoma Hepatocelular/virologia , Antígenos de Superfície da Hepatite B/metabolismo , Antígenos de Histocompatibilidade Classe I/metabolismo , Neoplasias Hepáticas/virologia , MicroRNAs/genética , Carcinoma Hepatocelular/genética , Regulação Neoplásica da Expressão Gênica , Células Hep G2/virologia , Antígenos de Superfície da Hepatite B/genética , Vírus da Hepatite B/patogenicidade , Vírus da Hepatite B/fisiologia , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Células Matadoras Naturais/metabolismo , Células Matadoras Naturais/patologia , Neoplasias Hepáticas/genética
8.
Carcinogenesis ; 35(9): 2127-33, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24913918

RESUMO

Hepatitis B virus surface antigen (HBsAg) is an important risk factor for hepatocellular carcinoma (HCC) and is downregulated during hepatocarcinogenesis. MicroRNAs (miRNAs) are frequently deregulated in HCC tissues. However, whether the deregulation of certain miRNAs in HCC has an impact on HBsAg expression remains unclear. We found here that microRNA-581 (miR-581), which is deregulated during hepatocarcinogenesis, promoted HBsAg expression. Additionally, miR-581 targeted Dicer and endoplasmic reticulum degradation-enhancing alpha-mannosidase-like protein 1 (EDEM1) and repressed their expression. Although Dicer cannot process HBV transcripts, Dicer knockdown led to increased HBsAg secretion, most likely due to a reduction in the levels of Dicer-processed 7SL RNA fragments. Moreover, Dicer-processed 7SL RNA fragments partially inhibited the ability of miR-581 to stimulate HBsAg expression. Furthermore, we found that forced EDEM1 expression inhibited miR-581-mediated induction of HBsAg. Finally, transfection of miR-581 into HepG2.2.15 cells promoted cell proliferation and led to upregulation of genes involved in development, cell proliferation and protein secretion. Altogether, we conclude that miR-581 promotes HBsAg expression by targeting Dicer and EDEM1. Our findings suggest that downregulation of miR-581 during hepatocarcinogenesis may lead to a reduction in HBsAg expression and impede HCC development.


Assuntos
RNA Helicases DEAD-box/genética , Antígenos de Superfície da Hepatite B/metabolismo , Proteínas de Membrana/genética , MicroRNAs/genética , Ribonuclease III/genética , Regiões 3' não Traduzidas , Sítios de Ligação , RNA Helicases DEAD-box/metabolismo , Regulação Neoplásica da Expressão Gênica , Regulação Viral da Expressão Gênica , Células Hep G2 , Humanos , Proteínas de Membrana/metabolismo , Interferência de RNA , Ribonuclease III/metabolismo
9.
J Gastroenterol Hepatol ; 29(6): 1149-58, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24476011

RESUMO

BACKGROUND AND AIM: Controlled attenuation parameter (CAP) is a novel ultrasound-based elastography method for detection of steatosis severity. This meta-analysis aimed to assess the performance of CAP. METHODS: PubMed, the Cochrane Library, and the Web of Knowledge were searched to find studies, published in English, relating to accuracy evaluations of CAP for detecting stage 1 (S1), stage 2 (S2), or stage 3 (S3) hepatic steatosis which was diagnosed by liver biopsy. Sensitivities, specificities, and hierarchical summary receiver operating characteristic (HSROC) curves were used to examine CAP performance. The clinical utility of CAP was also evaluated. RESULTS: Nine studies, with 11 cohorts were analyzed. The summary sensitivities and specificities values were 0.78 (95% confidence interval [CI], 0.69-0.84) and 0.79 (95% CI, 0.68-0.86) for ≥ S1, 0.85 (95% CI, 0.74-0.92) and 0.79 (95% CI, 0.71-0.85) for ≥ S2, and 0.83 (95% CI, 0.76-0.89) and 0.79 (95% CI, 0.68-0.87) for ≥ S3. The HSROCs were 0.85 (95% CI, 0.81-88) for ≥ S1, 0.88 (95% CI, 0.85-0.91) for ≥ S2, and 0.87 (95% CI, 0.84-0.90) for ≥ S3. Following a "positive" measurement (over the threshold value) for ≥ S1, ≥ S2, and ≥ S3, the corresponding post-test probabilities for the presence of steatosis (pretest probability was 50%) were 78%, 80% and 80%, respectively; if the values were below these thresholds ("negative" results), the post-test probabilities were 22%, 16%, and 17%, respectively. CONCLUSIONS: CAP has good sensitivity and specificity for detecting hepatic steatosis; however, based on a meta-analysis, CAP was limited in their accuracy of steatosis, which precluded widespread use in clinical practice.


Assuntos
Bases de Dados Bibliográficas , Técnicas de Imagem por Elasticidade/métodos , Fígado Gorduroso/diagnóstico , Hepatopatias/diagnóstico , Ultrassonografia/métodos , Doença Crônica , Estudos de Coortes , Humanos , Curva ROC , Sensibilidade e Especificidade , Índice de Gravidade de Doença
10.
Ophthalmologica ; 232(2): 110-7, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24516061

RESUMO

PURPOSE: To evaluate the relationship between lumican polymorphisms and high myopia in Chinese populations. METHODS: An electronic search was conducted in Pubmed, Embase, Cochrane Library and the China Biological Medicine Database for articles published prior to September 30, 2012. A meta-analysis was performed to assess heterogeneity, combine results and determine publication bias. RESULTS: This meta-analysis, including 1,545 subjects from 5 studies, indicated that Chinese lumican rs3759223 C allele carriers had a decreased risk of high myopia in comparison to T allele carriers (odds ratio: 0.531; 95% confidence interval, CI: 0.304-0.925; p = 0.025). There was some heterogeneity between studies. A metaregression showed that the mean axial length of controls weakens the effect of rs3759223 on high myopia (slope: -0.914; 95% CI: -1.490 to 0.337; p = 0.002). Sensitivity analysis confirmed the reliability and stability of this meta-analysis. CONCLUSION: Chinese lumican rs3759223 C allele carriers may be at reduced risk of high myopia.


Assuntos
Povo Asiático/genética , Proteoglicanas de Sulfatos de Condroitina/genética , Sulfato de Queratano/genética , Miopia Degenerativa/genética , Polimorfismo de Nucleotídeo Único , Regiões Promotoras Genéticas/genética , Adulto , Estudos de Casos e Controles , China/epidemiologia , Feminino , Genótipo , Humanos , Lumicana , Masculino , Miopia Degenerativa/etnologia , Adulto Jovem
11.
STAR Protoc ; 5(2): 102998, 2024 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-38573862

RESUMO

We recently identified a class of small cytosolic double-stranded DNA (scDNA) approximately 20-40 bp in size in human and mouse cells. Here, we present a protocol for scDNA isolation from cultured murine cells. We describe steps for cytosolic compartment separation, DNA isolation in the cytosolic fraction using phenol-chloroform extraction, and ethanol precipitation. We then detail procedures for denaturing purified cytosolic DNA through urea polyacrylamide gel electrophoresis and obtaining scDNA in the cytosolic DNA fraction via gel purification. For complete details on the use and execution of this protocol, please refer to Liu et al.1.


Assuntos
Citosol , DNA , Animais , Camundongos , DNA/isolamento & purificação , Citosol/metabolismo , Citosol/química , Células Cultivadas , Eletroforese em Gel de Poliacrilamida/métodos
12.
Nat Commun ; 15(1): 6964, 2024 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-39138195

RESUMO

Though RNAi and RNA-splicing machineries are involved in regulating severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) replication, their precise roles in coronavirus disease 2019 (COVID-19) pathogenesis remain unclear. Herein, we show that decreased RNAi component (Dicer and XPO5) and splicing factor (SRSF3 and hnRNPA3) expression correlate with increased COVID-19 severity. SARS-CoV-2 N protein induces the autophagic degradation of Dicer, XPO5, SRSF3, and hnRNPA3, inhibiting miRNA biogenesis and RNA splicing and triggering DNA damage, proteotoxic stress, and pneumonia. Dicer, XPO5, SRSF3, and hnRNPA3 knockdown increases, while their overexpression decreases, N protein-induced pneumonia's severity. Older mice show lower expression of Dicer, XPO5, SRSF3, and hnRNPA3 in their lung tissues and exhibit more severe N protein-induced pneumonia than younger mice. PJ34, a poly(ADP-ribose) polymerase inhibitor, or anastrozole, an aromatase inhibitor, ameliorates N protein- or SARS-CoV-2-induced pneumonia by restoring Dicer, XPO5, SRSF3, and hnRNPA3 expression. These findings will aid in developing improved treatments for SARS-CoV-2-associated pneumonia.


Assuntos
COVID-19 , Carioferinas , Ribonuclease III , SARS-CoV-2 , Fatores de Processamento de Serina-Arginina , Animais , Fatores de Processamento de Serina-Arginina/metabolismo , Fatores de Processamento de Serina-Arginina/genética , Humanos , Ribonuclease III/metabolismo , Ribonuclease III/genética , SARS-CoV-2/genética , COVID-19/metabolismo , COVID-19/virologia , COVID-19/genética , Camundongos , Carioferinas/metabolismo , Carioferinas/genética , RNA Helicases DEAD-box/metabolismo , RNA Helicases DEAD-box/genética , Regulação para Baixo , Pulmão/metabolismo , Pulmão/patologia , Pulmão/virologia , Masculino , Feminino , MicroRNAs/genética , MicroRNAs/metabolismo , Splicing de RNA , Autofagia/genética , Dano ao DNA , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B
13.
J Gastroenterol Hepatol ; 28(9): 1434-43, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23731128

RESUMO

BACKGROUND AND AIM: Endoscopic ultrasound (EUS) elastography is not used for detection but rather for characterization of solid pancreatic masses. A meta-analysis was used to assess the accuracy of EUS elastography for identification of malignant pancreatic masses. METHODS: PubMed, the Cochrane Library, and the ISI Web of Knowledge were searched. The studies relating to evaluation accuracy of qualitative or quantitative EUS elastography for identification of malignant pancreatic masses were collected. Language was limited to English. The sensitivity and specificity were used to examine the accuracy. Clinical utility was evaluated by likelihood ratio scattergram. RESULTS: A total of 10 studies including 893 pancreatic masses (646 malignant, 72.3%) were analyzed. The summary sensitivity and specificity for the diagnosis of malignant pancreatic masses were 0.98 (95% confidence interval [CI] 0.93-1.00) and 0.69 (95% CI 0.52-0.82) for qualitative EUS elastography, and 0.96 (95% CI 0.86-0.99) and 0.76 (95% CI 0.58-0.87) for quantitative EUS elastography, respectively. The hierarchical summary receiver operating characteristic curves were 0.94 and 0.93 for qualitative and quantitative EUS elastography. The accuracy of quantitative methods was similar to qualitative methods. The positive and negative likelihood ratios were 3.15 and 0.03 for qualitative EUS elastography, and 3.94 and 0.05 for quantitative EUS elastography, respectively. Both qualitative and quantitative methods were useful for exclusion of presence of malignant pancreatic masses and not for its confirmation. CONCLUSIONS: EUS elastography could be used as a good identification tool for benign and malignant pancreatic masses, with its good performance for exclusion of presence of malignant pancreatic masses.


Assuntos
Técnicas de Imagem por Elasticidade/métodos , Endossonografia/métodos , Neoplasias Pancreáticas/diagnóstico por imagem , Diagnóstico Diferencial , Humanos , Sensibilidade e Especificidade
14.
Onkologie ; 36(10): 560-5, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24107909

RESUMO

BACKGROUND/AIMS: To accurately evaluate the impact of the C/T polymorphism in microRNA (miRNA)-196a2 on the colorectal cancer (CRC) risk, by meta-analysis. METHODS: An electronic search for articles was conducted in PubMed, EMBASE, ISI Web of Science, and the Cochrane Library. The pooled odds ratio (OR) and its 95% confidence interval (CI) were used to assess the association through meta-analysis. RESULTS: 5 studies were used for analysis. The results showed a significant association between the miRNA-196a2 C/T polymorphism and CRC risk in the genetic models (C vs. T: OR = 1.168, 95% CI = 1.106-1.282, p = 0.001; CC vs. TT: OR = 1.368, 95% CI = 1.132-1.654, p = 0.001; TC/CC vs. TT: OR = 1.206, 95% = CI 1.035-1.405, p = 0.016; CC vs. TC/TT: OR = 1.254, 95% CI = 1.077-1.461, p = 0.004), with the exception of the TC-versus-TT model (TC vs. TT: OR = 1.130, 95% CI = 0.961-1.329, p = 0.138). In a subgroup analysis based on ethnicity, we identified a significant overrepresentation of the polymorphism in individuals of Asian ethnicity. CONCLUSION: This meta-analysis indicates a significant association between the miRNA-196a2 polymorphism and CRC risk.


Assuntos
Neoplasias Colorretais/epidemiologia , Neoplasias Colorretais/genética , Marcadores Genéticos/genética , Predisposição Genética para Doença/epidemiologia , Predisposição Genética para Doença/genética , MicroRNAs/genética , Polimorfismo de Nucleotídeo Único/genética , Feminino , Estudos de Associação Genética , Humanos , Masculino , Prevalência , Fatores de Risco
15.
Autophagy ; : 1-3, 2023 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-38013411

RESUMO

The CGAS (cyclic GMP-AMP synthase)-STING1 (stimulator of interferon response cGAMP interactor 1) pathway is an important innate immune pathway that induces proinflammatory cytokine production following stimulation with dsDNA > 45 bp. We recently identified a class of ~ 20-40 bp small cytosolic dsDNA (scDNA) that blocks CGAS-STING1 activation. In this punctum, we discuss the mechanism underlying the inhibition of CGAS-STING1 activation via scDNA. scDNA binds to CGAS but cannot activate its enzymatic activity. It competes with dsDNA > 45 bp for binding with CGAS to inhibit CGAS-STING1 activation. Moreover, scDNA activates macroautophagy/autophagy and induces the autophagic degradation of STING1 and long dsDNA. Autophagy then increases scDNA levels, driving a feedback loop that accelerates the degradation of STING1 and long cytosolic dsDNA. These findings reveal that mutual communication between scDNA and autophagy inhibits CGAS-STING1 activation following stimulation with dsDNA > 45 bp.

16.
Med Oncol ; 41(1): 10, 2023 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-38070011

RESUMO

Cadherin epidermal growth factor and laminin-G seven-pass G-type receptor 1 (CELSR1) is a planar cell polarity protein involved in the transmission of directional cues to align either individual cells within an epithelial sheet or multicellular clusters. CELSR1 has been suggested to play a role in glioma, breast cancer, and chronic lymphocytic leukemia development; however, whether it has a role in the pathogenesis of ovarian cancer remains unknown. The aim of this study was to determine the role of CELSR1 in ovarian cancer and elucidate its underlying molecular mechanisms. By analyzing gene expression data downloaded from the Cancer Genome Atlas database, we found that CELSR1 expression was upregulated in ovarian cancer tissues compared to that in normal ovarian tissues. High CELSR1 expression levels were associated with poor prognosis in patients with ovarian cancer. Cell proliferation, scratch, and transwell assays revealed that CELSR1 promoted the proliferation, migration, and invasion of ovarian cancer cells in vitro. In addition, transcriptome sequencing analysis revealed that CELSR1 knockdown in T29H cells resulted in the dysregulation of the expression of 1320 genes. Further analysis revealed that genes involved in proliferation- and migration-associated signaling pathways were regulated by CELSR1. Our study demonstrates that CELSR1 is highly expressed in ovarian cancer cells and regulates their proliferation and migration, suggesting its potential as a diagnostic marker and therapeutic target.


Assuntos
Caderinas , Neoplasias Ovarianas , Feminino , Humanos , Caderinas/metabolismo , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Ovarianas/metabolismo , Regulação para Cima
17.
Med Oncol ; 40(8): 217, 2023 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-37393403

RESUMO

FAM50A encodes a nuclear protein involved in mRNA processing; however, its role in cancer development remains unclear. Herein, we conducted an integrative pan-cancer analysis using The Cancer Genome Atlas, Genotype-Tissue Expression, and the Clinical Proteomic Tumor Analysis Consortium databases. Based on the gene expression data from TCGA and GTEx databases, we compared FAM50A mRNA levels in 33 types of human cancer tissues to those in corresponding normal tissues and found that FAM50A mRNA level was upregulated in 20 of the 33 types of common cancer tissues. Then, we compared the DNA methylation status of the FAM50A promoter in tumor tissues to that in corresponding normal tissues. FAM50A upregulation was accompanied by promoter hypomethylation in 8 of the 20 types of tumor tissues, suggesting that promoter hypomethylation contributes to the upregulation of FAM50A in these cancer tissues. Elevated FAM50A expression in 10 types of cancer tissues was associated with poor prognosis in patients with cancer. FAM50A expression was positively correlated with CD4+ T-lymphocyte and dendritic cell infiltration in cancer tissues but was negatively correlated with CD8+ T-cell infiltration in cancer tissues. FAM50A knockdown caused DNA damage, induced interferon beta and interleukin-6 expression, and repressed the proliferation, invasion, and migration of cancer cells. Our findings indicate that FAM50A might be useful in cancer detection, reveal insights into its role in cancer development, and may contribute to the development of cancer diagnostics and treatments.


Assuntos
Neoplasias , Proteômica , Humanos , Regulação para Cima , Ativação Transcricional , Neoplasias/genética , Linfócitos T CD4-Positivos , Proteínas de Ligação a DNA , Proteínas de Ligação a RNA
18.
Med Oncol ; 40(3): 96, 2023 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-36792863

RESUMO

Tubulin γ-1 (TUBG1) is a highly conserved component of the centrosome and its deregulation is involved in the development of several types of cancer. However, the role of TUBG1 in hepatocellular carcinoma (HCC) remains unclear. In this study, we found that TUBG1 was upregulated in human HCC cells and tissues and that TUBG1 upregulation was associated with promoter hypomethylation in HCC tissues. TUBG1 knockdown suppressed the proliferation, invasion, and migration of HCC cells. While TUBG1 expression was positively correlated with CD4 + memory T lymphocyte infiltration, it was negatively correlated with CD4 + regulatory T-cell infiltration in human HCC tissues. Furthermore, TUBG1 expression was positively correlated with the expression of genes involved in cell division. Noticeably, high expression of TUBG1 was associated with poor prognosis in patients with HCC. Overall, our findings revealed that TUBG1 promotes hepatocarcinogenesis by increasing proliferation, invasion, and migration of HCC cells and may regulate T lymphocyte infiltration. The current findings provide important insights into TUBG1 regulation in HCC, which could provide new therapeutic targets for hepatocarcinoma which has a very high incidence and mortality rate worldwide.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/patologia , Regulação para Cima , Tubulina (Proteína)/genética , Proliferação de Células/genética , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Movimento Celular/genética
19.
Cell Death Dis ; 14(12): 846, 2023 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-38114473

RESUMO

Radiotherapy is an important treatment modality for patients with esophageal cancer; however, the response to radiation varies among different tumor subpopulations due to tumor heterogeneity. Cancer cells that survive radiotherapy (i.e., radioresistant) may proliferate, ultimately resulting in cancer relapse. However, the interaction between radiosensitive and radioresistant cancer cells remains to be elucidated. In this study, we found that the mutual communication between radiosensitive and radioresistant esophageal cancer cells modulated their radiosensitivity. Radiosensitive cells secreted more exosomal let-7a and less interleukin-6 (IL-6) than radioresistant cells. Exosomal let-7a secreted by radiosensitive cells increased the radiosensitivity of radioresistant cells, whereas IL-6 secreted by radioresistant cells decreased the radiosensitivity of radiosensitive cells. Although the serum levels of let-7a and IL-6 before radiotherapy did not vary significantly between patients with radioresistant and radiosensitive diseases, radiotherapy induced a more pronounced decrease in serum let-7a levels and a greater increase in serum IL-6 levels in patients with radioresistant cancer compared to those with radiosensitive cancer. The percentage decrease in serum let-7a and the percentage increase in serum IL-6 levels at the early stage of radiotherapy were inversely associated with tumor regression after radiotherapy. Our findings suggest that early changes in serum let-7a and IL-6 levels may be used as a biomarker to predict the response to radiotherapy in patients with esophageal cancer and provide new insights into subsequent treatments.


Assuntos
Neoplasias Esofágicas , Interleucina-6 , Humanos , Recidiva Local de Neoplasia , Tolerância a Radiação/fisiologia , Neoplasias Esofágicas/radioterapia
20.
Cell Rep ; 42(8): 112852, 2023 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-37481718

RESUMO

The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway is a major mediator of inflammation following stimulation with >45 bp double-stranded DNA (dsDNA). Herein, we identify a class of ∼20-40 bp small cytosolic dsDNA (scDNA) molecules that compete with long dsDNA (200-1,500 bp herring testis [HT]-DNA) for binding to cGAS, thus repressing HT-DNA-induced cGAS activation. The scDNA promotes cGAS and Beclin-1 interaction, releasing Rubicon, a negative regulator of phosphatidylinositol 3-kinase class III (PI3KC3), from the Beclin-1-PI3KC3 complex. This leads to PI3KC3 activation and induces autophagy, causing degradation of STING and long cytosolic dsDNA. Moreover, DNA damage decreases, and autophagy inducers increase scDNA levels. scDNA transfection and treatment with autophagy inducers attenuate DNA damage-induced cGAS activation. Thus, scDNA molecules serve as effective brakes for cGAS activation, preventing excessive inflammatory cytokine production following DNA damage. Our findings may have therapeutic implications for cytosolic DNA-associated inflammatory diseases.


Assuntos
DNA , Proteínas de Membrana , Masculino , Humanos , Proteína Beclina-1 , Proteínas de Membrana/metabolismo , DNA/metabolismo , Nucleotidiltransferases/metabolismo , Fosfatidilinositol 3-Quinase , Autofagia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA