Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 85
Filtrar
1.
OMICS ; 27(5): 215-226, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37196148

RESUMO

Glioblastoma multiforme (GBM) is a highly malignant brain tumor where new biomarkers and drug targets are much needed in the oncology clinic. miR-433 was identified as a tumor-suppressing miRNA in several different types of human cancer. However, the integrative biology of miR-433 in GBM is still largely unknown. By analyzing the expression profiles of miR-433 in 198 patients with glioma at The Cancer Genome Atlas, we found that the miR-433 expression was decreased in glioma whereas the low expression of miR-433 was significantly associated with shorter overall survival. We then conducted in vitro studies and demonstrated that increased expression of miR-433 suppressed the proliferation, migration, and invasion of LN229 and T98G cells, two representative glioma cell lines. Further, using in vivo mouse model, we found that upregulation of miR-433 inhibited the tumor growth of glioma cells. To situate the integrative biology understanding of the action of miR-433 in glioma, we identified ERBB4 as a gene targeted directly by miR-433 in LN229 and T98G cells. Overexpressed ERBB4 rescued the phenotype caused by overexpression of miR-433. Finally, we showed that miR-433 suppressed the PI3K/Akt pathway in glioma cells. In conclusion, our study demonstrated that miR-433 could potentially act as a tumor suppressor for GBM and may serve as a potential therapeutic target for GBM. Further integrative biology and clinical translational research are warranted to evaluate miR-433 in GBM.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Glioma , MicroRNAs , Humanos , Animais , Camundongos , Glioblastoma/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Linhagem Celular Tumoral , Transdução de Sinais/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Glioma/genética , Neoplasias Encefálicas/metabolismo , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica/genética , Movimento Celular/genética , Receptor ErbB-4/genética , Receptor ErbB-4/metabolismo
2.
OMICS ; 26(8): 415-421, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35925812

RESUMO

Chronic liver disease (CLD) is a significant planetary health burden. CLD includes a broad range of liver pathologies from different causes, for example, hepatitis B virus infection, fatty liver disease, hepatocellular carcinoma, and nonalcoholic fatty liver disease or the metabolic associated fatty liver disease. Biomarker and diagnostic discovery, and new molecular targets for precision treatments are timely and sorely needed in CLD. In this context, multi-omics data integration is increasingly being facilitated by artificial intelligence (AI) and attendant digital transformation of systems science. While the digital transformation of multi-omics integrative analyses is still in its infancy, there are noteworthy prospects, hope, and challenges for diagnostic and therapeutic innovation in CLD. This expert review aims at the emerging knowledge frontiers as well as gaps in multi-omics data integration at bulk tissue levels, and those including single cell-level data, gut microbiome data, and finally, those incorporating tissue-specific information. We refer to AI and related digital transformation of the CLD research and development field whenever possible. This review of the emerging frontiers at the intersection of systems science and digital transformation informs future roadmaps to bridge digital technology discovery and clinical omics applications to benefit planetary health and patients with CLD.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Inteligência Artificial , Carcinoma Hepatocelular/patologia , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/terapia , Medicina de Precisão
3.
Cancer Med ; 10(16): 5681-5695, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34227253

RESUMO

In patients with prostate cancer (PCa), there is a high rate of overdiagnosis and frequent overtreatment. Therefore, there is an urgent need for more accurate prediction of biochemical recurrence (BCR). DNA methylation regulation patterns play crucial roles in tumorigenicity, progression, and treatment efficacy in PCa. However, the global relationship between epigenetic alterations, changes in mRNA levels, and pathologic phenotypes of PCa remain largely undefined. Here, we conducted a systematic analysis to identify global coexpression and comethylation modules in PCa. We identified coregulated methylation and expression modules and the relationships between epigenetic modifications, tumor progression, and the corresponding immune microenvironment in PCa. Our results show that DNA methyltransferases (DNMTs) are strongly associated with pathologic phenotypes and immune infiltration patterns in PCa. We built a two-factor predictive model using the expression features of DNMT3B and DNMT1. The model was used to predict the BCR status of patients with PCa and achieved area under the receiver operating characteristic curve values of 0.70 and 0.88 in the training and independent testing datasets, respectively.


Assuntos
5-Metilcitosina/metabolismo , DNA (Citosina-5-)-Metiltransferase 1/genética , DNA (Citosina-5-)-Metiltransferases/genética , Recidiva Local de Neoplasia/epidemiologia , Neoplasias da Próstata/genética , DNA (Citosina-5-)-Metiltransferase 1/metabolismo , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA/imunologia , Conjuntos de Dados como Assunto , Epigênese Genética/imunologia , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Calicreínas/sangue , Masculino , Modelos Genéticos , Recidiva Local de Neoplasia/diagnóstico , Recidiva Local de Neoplasia/genética , Antígeno Prostático Específico/sangue , Neoplasias da Próstata/sangue , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/terapia , Curva ROC , Medição de Risco/métodos , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia , DNA Metiltransferase 3B
4.
Transl Cancer Res ; 9(4): 2231-2242, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35117583

RESUMO

BACKGROUND: Prostate cancer (PCa) is a common type of malignancy, which represents one of the leading causes of death among men worldwide. Copy number variations (CNVs) and gene fusions play important roles in PCa and may serve as markers for the prognosis of this condition. METHODS: We have presently conducted an analysis of CNVs and gene fusions in PCa, using whole exome sequencing (WES) data of primary tumors. For this, a cohort of 74 PCa patients, including 30 recurrent and 44 non-recurrent cases, were assessed during 5 years of follow-up. RESULTS: We have identified 66 CNVs that were specific to the primary tumor tissues from the recurrent PCa group. Most of duplicated genomic regions were located in 8q2, suggesting that this chromosomal region could be important for the prognosis of PCa. Meanwhile, we have developed a random forest model, using six selected CNVs, with an accuracy near 90% for predicting PCa recurrence according to a 10-fold cross validation. In addition, we have detected 16 recurrent oncogenic gene fusions in PCa. Among these, ALK (ALK receptor tyrosine kinase)-involved fusions were the most common type of gene fusion (n=7). Four of these fusions (i.e., EML4-ALK, STRN-ALK, CLTC-ALK, ETV6-ALK) were previously identified in other cancer types, while the remaining three gene fusions (FRYL-ALK, ABL1-ALK, and BCR-ALK) were here identified. CONCLUSIONS: Our findings expand the current understanding in regard to prostate carcinogenesis. Current data might be further used for assay development as well as to predict PCa recurrence, using primary tissues.

5.
OMICS ; 23(8): 380-388, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31194651

RESUMO

Prostate cancer (PCa) is a highly common cancer among men but lacks robust diagnostics that can predict disease recurrence after initial treatment, for example, with radical prostatectomy. Recent advances in genomics and next-generation sequencing heralded the discovery of biomarkers such as the androgen receptor gene (AR) splice events, the TMPRSS2:EGR gene fusion, long noncoding RNA MALAT-1 and SCHLAP1 for PCa prognosis. Still, the question of why some patients experience recurrence, whereas others do not introduce marked uncertainty for both patients and physicians. We report here the whole exome sequencing of 30 recurrent and 44 nonrecurrent PCa patients. We identified 72 and 34 specific somatic single nucleotide variations in the recurrent and the nonrecurrent group, respectively, and developed a classification model to forecast PCa recurrence using a random forest model. The model displayed a sensitivity and specificity of 87.8% and 94.4%, respectively, for identifying the patients with recurrent PCa. These observations warrant further research in independent and larger clinical samples so as to inform future diagnostics innovation for PCa prognosis and recurrence.


Assuntos
Biomarcadores Tumorais , Sequenciamento do Exoma , Neoplasias da Próstata/diagnóstico , Neoplasias da Próstata/genética , Idoso , Biologia Computacional/métodos , Humanos , Masculino , Anotação de Sequência Molecular , Mutação , Gradação de Tumores , Recidiva Local de Neoplasia , Polimorfismo de Nucleotídeo Único , Prognóstico
6.
OMICS ; 23(4): 181-189, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30932742

RESUMO

Nonalcoholic fatty liver disease (NAFLD) is a growing global public health concern and becoming the leading cause of liver disease worldwide. The estimated global prevalence of NAFLD is ∼25% depending on the country and the assessment method used to establish the diagnosis. Meta-analyses suggest that the highest prevalence is in the Middle East (31.8%) and South America (30.4%), and the lowest in Africa (13.5%). In the United States, between 75 and 100 million individuals were estimated to have NAFLD. This important disease is associated with increased incidence of liver-related deaths, hepatocarcinoma, and overall mortality. Fibrosis stage, among other histological characteristics, is the most critical factor in predicting all-cause and disease-specific mortality in NAFLD. The ability to detect fibrosis early in NAFLD patients is critical in controlling mortality associated with this highly prevalent disease. We present here an expert review on recent advances in novel blood biomarkers, for example, the Wisteria floribunda agglutinin-positive mac-2 binding protein (WFA+-M2BP), type IV collagen 7S, chitinase 3 like 1 (CHI3L1; YKL-40), and insulin-like growth factor-1 (IGF-1). Algorithms using multiple biomarkers such as alpha-2-macroglobulin, mir34a, YKL-40, and hemoglobin A1c (HbA1c; NIS4), enhanced liver fibrosis (ELF), Hepascore, FibroMeter, FibroTest, FIBROSpect, FIB-C3, and ADPAPT are highlighted. Novel technologies such as tandem mass spectrometry to directly measure protein turnover rate of the key proteins involved in hepatic fibrosis, as an indicator of fibrogenesis, are also discussed. In conclusion, NAFLD is a growing global health problem that warrants long-term funding, research, and training of scholars across the fields of public health diagnostics, systems sciences, nutrition, hepatology, and clinical oncology.


Assuntos
Biomarcadores/sangue , Cirrose Hepática/sangue , Cirrose Hepática/diagnóstico , Neoplasias Hepáticas/sangue , Neoplasias Hepáticas/diagnóstico , Carcinoma Hepatocelular/sangue , Carcinoma Hepatocelular/diagnóstico , Carcinoma Hepatocelular/metabolismo , Saúde Global , Humanos , Hepatopatias , Neoplasias Hepáticas/metabolismo , Hepatopatia Gordurosa não Alcoólica/sangue , Hepatopatia Gordurosa não Alcoólica/diagnóstico , Hepatopatia Gordurosa não Alcoólica/metabolismo , Lectinas de Plantas/metabolismo , Saúde Pública , Receptores de N-Acetilglucosamina/metabolismo
7.
Sci Rep ; 8(1): 15029, 2018 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-30301907

RESUMO

CHI3L1 (YKL40) is a secreted glycoprotein and elevated serum CHI3L1 level has been proved to be associated with poor prognosis in many human cancers. However, the mechanism of how CHI3L1 causes poor prognosis in cancers is still unknown. Here, considering that CHI3L1 is a liver specific/enriched protein, we use hepatocellular carcinoma as a model to study the function of CHI3L1. We showed that, both in vivo and in vitro, overexpression of CHI3L1 could promote liver cancer cells growth, migration and invasion. We then used RNA-seq to analyze the expression profiles of CHI3L1 overexpressed in two HCC cell lines and found that CHI3L1 overexpression affected genes that were involved in cell-cell adhesion, extracellular exosome and adherens junction. Western blot analysis further revealed that CHI3L1 could activate TGF-ß signal pathways. Our data added new understanding of the mechanism of CHI3L1's action. 1) CHI3L1 promoted cancer cell proliferation by regulating cell cycles; 2) CHI3L1 promoted cancer cell invasion and metastasis; 3) CHI3L1 regulate liver cancer potentially by regulating the TGF-ß signaling pathways; 4) CHI3L1 has direct kinase activities or activate kinase to phosphorylate SMAD2, SMAD3.


Assuntos
Carcinoma Hepatocelular/genética , Proteína 1 Semelhante à Quitinase-3/genética , Neoplasias Hepáticas/genética , Fator de Crescimento Transformador beta/genética , Animais , Carcinogênese/genética , Carcinoma Hepatocelular/patologia , Proliferação de Células/genética , Transição Epitelial-Mesenquimal/genética , Regulação Neoplásica da Expressão Gênica/genética , Células Hep G2 , Humanos , Neoplasias Hepáticas/patologia , Camundongos , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Fosforilação , Transdução de Sinais , Proteína Smad2/genética , Proteína Smad3/genética , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Colloids Surf B Biointerfaces ; 172: 665-673, 2018 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-30243220

RESUMO

GBM, the most common and aggressive malignant primary brain tumors which needs new research approach to reveal the underline molecular mechanism of tumor progression. The 3D in vitro tumor model can be a simple and effective way to study tumor characteristics with ability to replicate of the tumor milieu. In the current study, we adopted the DNA microarray to analyze the gene expression of GBM tumor cells cultured under 2D cell culture flasks and 3D PLA porous scaffolds for 4,7 and 14 days. For 14 day old cultures, 8117 and 3060 genes expression were upregulated and downregulated respectively. Further KEGG pathway analysis revealed, the upregulated genes were mainly enriched/involved in PPAR and PI3K-Akt signaling pathways whereas the downregulated genes were mainly contributed in metabolism, ECM related and TGF-beta pathways. Thus, our approach of establishing 3D in vitro tumor model provides realistic results and proves itself a powerful tool for understanding the inner nature of GBM and can be considered as potential platform for drug screening.


Assuntos
Técnicas de Cultura de Células/métodos , Genômica/métodos , Glioblastoma/genética , Glioblastoma/patologia , Regulação Neoplásica da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Poliésteres/química , Células Tumorais Cultivadas
9.
PLoS One ; 12(6): e0177861, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28570625

RESUMO

The androgen receptor (AR) transcription factor plays a key role in the development and progression of prostate cancer, as is evident from the efficacy of androgen-deprivation therapy, AR is also the most frequently mutated gene, in castration resistant prostate cancer (CRPC). AR has therefore become an even more attractive therapeutic target in aggressive and disseminated prostate cancer. To investigate mechanisms of AR and AR target gene activation in different subpopulations of prostate cancer cells, a toolkit of AR expressor and androgen response element (ARE) reporter vectors were developed. Three ARE reporter vectors were constructed with different ARE consensus sequences in promoters linked to either fluorescence or luciferase reporter genes in lentiviral vector backbones. Cell lines transduced with the different vectors expressed the reporters in an androgen-dependent way according to fluorescence microscopy, flow cytometry and multi-well fluorescent and luminescence assays. Interestingly, the background reporter activity in androgen-depleted medium was significantly higher in LNCaP cells compared to the prostate transit amplifying epithelial cell lines, EP156T-AR and 957E/hTERT-AR with exogenous AR. The androgen-induced signal to background was much higher in the latter benign prostate cells than in LNCaP cells. Androgen-independent nuclear localization of AR was seen in LNCaP cells and reduced ARE-signaling was seen following treatment with abiraterone, an androgen synthesis inhibitor. The ARE reporter activity was significantly stronger when stimulated by androgens than by ß-estradiol, progesterone and dexamethasone in all tested cell types. Finally, no androgen-induced ARE reporter activity was observed in tumorigenic mesenchymal progeny cells of EP156T cells following epithelial to mesenchymal transition. This underscores the observation that expression of the classical luminal differentiation transcriptome is restricted in mesenchymal type cells with or without AR expression, and presence of androgen.


Assuntos
Genes Reporter , Próstata/fisiologia , Receptores Androgênicos/fisiologia , Linhagem Celular , Proteínas de Fluorescência Verde/genética , Humanos , Masculino , Próstata/citologia , Receptores Androgênicos/genética , Transfecção
10.
Oncotarget ; 8(16): 26185-26199, 2017 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-28412734

RESUMO

Hepatocellular carcinoma (HCC) has become the third most deadly disease worldwide and HBV is the major factor in Asia and Africa. We conducted 9 WGS (whole genome sequencing) analyses for matched samples of tumor, adjacent non-tumor tissues and normal blood samples of HCC patients from three HBV positive patients. We then validated the mutations identified in a larger cohort of 177 HCC patients. We found that the number of the unique somatic mutations (average of 59,136) in tumor samples is significantly less than that in adjacent non-tumor tissues (average 83, 633). We discovered that the TP53 R249S mutation occurred in 7.7% of the HCC patients, and it was significantly associated with poor diagnosis. In addition, we found that the L104P mutation in the VCX gene (Variable charge, X-linked) was absent in white blood cell samples, but present at 11.1% frequency in the adjacent tissues and increased to 14.6% in HCC tissues, suggesting that this mutation might be a tumor driver gene driving HCC carcinogenesis. Finally, we identified a TK1-RNU7 fusion, which would result in a deletion of 103 amino acids from its C-terminal. The frequencies of this fusion event decreased from the adjacent tissues (29.2%) to the tumors (16.7%), suggesting that a truncated thymidine Kinase1 (TK1) caused by the fusion event might be deleterious and be selected against during tumor progression. The three-way comparisons allow the identification of potential driver mutations of carcinogenesis. Furthermore, our dataset provides the research community a valuable dataset for identifying dynamic changes of mutation profiles and driver mutations for HCC.


Assuntos
Carcinoma Hepatocelular/genética , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Neoplasias Hepáticas/genética , Mutação , Carcinoma Hepatocelular/patologia , Biologia Computacional/métodos , Variações do Número de Cópias de DNA , Genômica/métodos , Humanos , Estimativa de Kaplan-Meier , Neoplasias Hepáticas/patologia , Polimorfismo de Nucleotídeo Único , Reprodutibilidade dos Testes , Sequenciamento Completo do Genoma
11.
Proc Natl Acad Sci U S A ; 113(33): 9339-44, 2016 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-27482107

RESUMO

Oncogenic mutations of the Wnt (wingless)/ß-catenin pathway are frequently observed in major cancer types. Thus far, however, no therapeutic agent targeting Wnt/ß-catenin signaling is available for clinical use. Here we demonstrate that axitinib, a clinically approved drug, strikingly blocks Wnt/ß-catenin signaling in cancer cells, zebrafish, and Apc(min/+) mice. Notably, axitinib dramatically induces Wnt asymmetry and nonrandom DNA segregation in cancer cells by promoting nuclear ß-catenin degradation independent of the GSK3ß (glycogen synthase kinase3ß)/APC (adenomatous polyposis coli) complex. Using a DARTS (drug affinity-responsive target stability) assay coupled to 2D-DIGE (2D difference in gel electrophoresis) and mass spectrometry, we have identified the E3 ubiquitin ligase SHPRH (SNF2, histone-linker, PHD and RING finger domain-containing helicase) as the direct target of axitinib in blocking Wnt/ß-catenin signaling. Treatment with axitinib stabilizes SHPRH and thereby increases the ubiquitination and degradation of ß-catenin. Our findings suggest a previously unreported mechanism of nuclear ß-catenin regulation and indicate that axitinib, a clinically approved drug, would provide therapeutic benefits for cancer patients with aberrant nuclear ß-catenin activation.


Assuntos
Divisão Celular/efeitos dos fármacos , Imidazóis/farmacologia , Indazóis/farmacologia , Neoplasias/patologia , Inibidores de Proteínas Quinases/farmacologia , Via de Sinalização Wnt/efeitos dos fármacos , beta Catenina/fisiologia , Animais , Axitinibe , DNA Helicases/fisiologia , Glicogênio Sintase Quinase 3 beta/fisiologia , Células HCT116 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Regeneração/efeitos dos fármacos , Ubiquitina-Proteína Ligases/fisiologia , Peixe-Zebra
12.
BMC Cancer ; 16: 377, 2016 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-27378372

RESUMO

BACKGROUND: Expression of the androgen receptor (AR) is associated with androgen-dependent proliferation arrest and terminal differentiation of normal prostate epithelial cells. Additionally, activation of the AR is required for survival of benign luminal epithelial cells and primary cancer cells, thus androgen deprivation therapy (ADT) leads to apoptosis in both benign and cancerous tissue. Escape from ADT is known as castration-resistant prostate cancer (CRPC). In the course of CRPC development the AR typically switches from being a cell-intrinsic inhibitor of normal prostate epithelial cell proliferation to becoming an oncogene that is critical for prostate cancer cell proliferation. A clearer understanding of the context dependent activation of the AR and its target genes is therefore desirable. METHODS: Immortalized human prostate basal epithelial EP156T cells and progeny cells that underwent epithelial to mesenchymal transition (EMT), primary prostate epithelial cells (PrECs) and prostate cancer cell lines LNCaP, VCaP and 22Rv1 were used to examine context dependent restriction and activation of the AR and classical target genes, such as KLK3. Genome-wide gene expression analyses and single cell protein analyses were applied to study the effect of different contexts. RESULTS: A variety of growth conditions were tested and found unable to activate AR expression and transcription of classical androgen-dependent AR target genes, such as KLK3, in prostate epithelial cells with basal cell features or in mesenchymal type prostate cells. The restriction of androgen- and AR-dependent transcription of classical target genes in prostate basal epithelial cells was at the level of AR expression. Exogenous AR expression was sufficient for androgen-dependent transcription of AR target genes in prostate basal epithelial cells, but did not exert a positive feedback on endogenous AR expression. Treatment of basal prostate epithelial cells with inhibitors of epigenetic gene silencing was not efficient in inducing androgen-dependent transcription of AR target genes, suggesting the importance of missing cofactor(s). CONCLUSIONS: Regulatory mechanisms of AR and androgen-dependent AR target gene transcription are insufficiently understood and may be critical for prostate cancer initiation, progression and escape from standard therapy. The present model is useful for the study of context dependent activation of the AR and its transcriptome.


Assuntos
Redes Reguladoras de Genes , Neoplasias da Próstata/metabolismo , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Neoplasias da Próstata/genética , Ligação Proteica , Análise de Sequência de RNA/métodos , Análise de Célula Única
13.
OMICS ; 19(6): 339-45, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26415140

RESUMO

Liver fibrosis is a major disease that is primarily caused by hepatitis virus infections, toxins, and alcohol abuse. Diagnosing and staging liver fibrosis are critical in guiding the treatment of chronic liver diseases, according to several international and Chinese guidelines. Liver biopsy is the gold standard for diagnosing and staging liver fibrosis, but it is invasive and suffers from several limitations. Consequently, much research has focused on the search for a noninvasive serum biomarker of fibrosis. In this study, we determined that Chitinase 3-like 1 (CHI3L1) is an abundantly expressed liver gene whose expression is highly enriched in the liver. We then compared serum levels of CHI3L1 among patients with various stages of liver fibrosis, as determined by liver biopsies, and found that the CHI3L1 levels were able to differentiate early stages of liver fibrosis (S0-S2) from late stages of liver fibrosis (S3-S4). We further showed that CHI3L1 is a good marker of substantial fibrosis, with areas under the ROC curves (AUCs) of 0.94 for substantial (S2, S3, S4) fibrosis and 0.96 for advanced (S3, S4) fibrosis. Finally, we showed that CHI3L1 is superior to hyaluronic acid (HA), type III procollagen (PCIII), laminin (LN), and type IV collagen (CIV), which are also serum biomarkers of liver fibrosis, in identifying advanced liver fibrosis in patients with HBV-related liver fibrosis in China.


Assuntos
Adipocinas/genética , Adipocinas/metabolismo , Biomarcadores/metabolismo , Lectinas/genética , Lectinas/metabolismo , Cirrose Hepática/diagnóstico , Cirrose Hepática/genética , Fígado/metabolismo , Fígado/patologia , Adipocinas/sangue , Biomarcadores/sangue , Proteína 1 Semelhante à Quitinase-3 , Ensaio de Imunoadsorção Enzimática , Humanos , Lectinas/sangue
14.
Cancer Lett ; 365(2): 240-50, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26052095

RESUMO

MMP14 (MT1-MMP) is a cell membrane-associated proteinase of the extracellular matrix, whose biological roles vary from angiogenesis to cell proliferation and survival. We recently found a direct correlation between MMP14 expression levels in brain tumors of glioma patients and the disease progression. By using gene silencing as an experimental approach we found that MMP14 knockdown decreases production of pro-angiogenic factors such as VEGF and IL8 and thereby suppresses angiogenesis in glioma tumors. Although the clinical relevance of MMP14 down-regulation and its possible implications for glioma therapy in humans remain unclear, we observed a significant improvement in animal survival upon down-regulation of MMP14 in murine intracranial glioma xenografts infected with MMP14 shRNA-expressing CRAd. We further found that down-regulation of MMP14 in gliomas by combinational treatment with CRAd-S-5/3 and Marimastat, a chemical inhibitor of metalloproteinases, augments suppression of pro-angiogenic factors, caused by the replication-competent adenovirus. We also demonstrated that delivery of MMP14-targeting shRNA by a fiber-modified adenoviral vector to the glioma cells effectively suppresses their proliferation in vitro and in vivo. Thus our data indicate that inhibition of MMP14 expression in tumors in combination with glioma virotherapy could be effectively utilized to suppress angiogenesis and neovascularization of glioma tumors by decreasing production of pro-angiogenic factors.


Assuntos
Adenoviridae/genética , Neoplasias Encefálicas/genética , Glioma/genética , Metaloproteinase 14 da Matriz/genética , Terapia Viral Oncolítica , Idoso , Animais , Linhagem Celular Tumoral , Proliferação de Células/genética , Progressão da Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Terapia Genética , Vetores Genéticos/genética , Humanos , Ácidos Hidroxâmicos/farmacologia , Interleucina-8/biossíntese , Masculino , Camundongos , Pessoa de Meia-Idade , Transplante de Neoplasias , Neovascularização Patológica/genética , Interferência de RNA , RNA Interferente Pequeno , Transplante Heterólogo , Fator A de Crescimento do Endotélio Vascular/biossíntese
15.
Oncotarget ; 6(10): 7686-700, 2015 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-25797256

RESUMO

Androgen deprivation therapy in prostate cancer (PCa) causes neuroendocrine differentiation (NED) of prostatic adenocarcinomas (PAC) cells, leading to recurrence of PCa. Androgen-responsive genes involved in PCa progression including NED remain largely unknown. Here we demonstrated the importance of androgen receptor (AR)-microRNA-204 (miR-204)-XRN1 axis in PCa cell lines and the rat ventral prostate. Androgens downregulate miR-204, resulting in induction of XRN1 (5'-3' exoribonuclease 1), which we identified as a miR-204 target. miR-204 acts as a tumor suppressor in two PAC cell lines (LNCaP and 22Rv1) and as an oncomiR in two neuroendocrine-like prostate cancer (NEPC) cell lines (PC-3 and CL1). Importantly, overexpression of miR-204 and knockdown of XRN1 inhibited AR expression in PCa cells. Repression of miR-34a, a known AR-targeting miRNA, contributes AR expression by XRN1. Thus we revealed the AR-miR-204-XRN1-miR-34a positive feedback loop and a dual function of miR-204/XRN1 axis in prostate cancer.


Assuntos
Adenocarcinoma/genética , Adenocarcinoma/patologia , Exorribonucleases/genética , MicroRNAs/genética , Proteínas Associadas aos Microtúbulos/genética , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Adenocarcinoma/metabolismo , Animais , Castração , Processos de Crescimento Celular/genética , Linhagem Celular Tumoral , Regulação para Baixo , Exorribonucleases/metabolismo , Xenoenxertos , Humanos , Masculino , MicroRNAs/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Neoplasias da Próstata/metabolismo , Ratos , Transfecção
16.
Oncotarget ; 6(6): 3977-87, 2015 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-25738357

RESUMO

Oncolytic gene therapy using viral vectors may provide an attractive therapeutic option for malignant gliomas. These viral vectors are designed in a way to selectively target tumor cells and spare healthy cells. To determine the translational impact, it is imperative to assess the factors that interfere with the anti-glioma effects of the oncolytic adenoviral vectors. In the current study, we evaluated the efficacy of survivin-driven oncolytic adenoviruses pseudotyping with adenoviral fiber knob belonging to the adenoviral serotype 3, 11 and 35 in their ability to kill glioblastoma (GBM) cells selectively without affecting normal cells. Our results indicate that all recombinant vectors used in the study can effectively target GBM in vitro with high specificity, especially the 3 knob-modified vector. Using intracranial U87 and U251 GBM xenograft models we have also demonstrated that treatment with Conditionally Replicative Adenovirus (CRAd-S-5/3) vectors can effectively regress tumor. However, in several patient-derived GBM cell lines, cells exhibited resistance to the CRAd infection as evident from the diminishing effects of autophagy. To improve therapeutic response, tumor cells were pretreated with tamoxifen. Our preliminary data suggest that tamoxifen sensitizes glioblastoma cells towards oncolytic treatment with CRAd-S-5/3, which may prove useful for GBM in future experimental therapy.


Assuntos
Adenoviridae/fisiologia , Antineoplásicos Hormonais/farmacologia , Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Terapia Viral Oncolítica/métodos , Tamoxifeno/farmacologia , Adenoviridae/genética , Animais , Autofagia/efeitos dos fármacos , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/virologia , Linhagem Celular Tumoral , Terapia Combinada , Feminino , Glioblastoma/genética , Glioblastoma/patologia , Glioblastoma/virologia , Humanos , Camundongos , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Oncotarget ; 6(7): 5369-81, 2015 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-25749033

RESUMO

Epigenetic changes, including H3K4me3 and H3K27me3 histone modification, play an important role in carcinogenesis. However, no genome-wide histone modification map has been generated for gliomas. Here, we report a genome-wide map of H3K4me3 and H3K27me3 histone modifications for 8 glioma stem cell (GSC) lines, together with the associated gene activation or repression patterns. In addition, we compared the genome-wide histone modification maps of GSC lines to those of astrocytes to identify unique gene activation or repression profiles in GSCs and astrocytes. We also identified a set of bivalent genes, which are genes that are associated with both H3K4me3 and H3K27me3 marks and are poised for action in embryonic stem cells. These bivalent genes are potential targets for inducing differentiation in glioblastoma (GBM) as a therapeutic approach. Finally, we identified SLC17A7 as a bivalent tumor suppressor gene in GBM, as it is down-regulated at both the protein and RNA levels in GBM tissues compared with normal brain tissues, and it inhibits GBM cell proliferation, migration and invasion.


Assuntos
Cromatina/genética , Genes Supressores de Tumor , Glioblastoma/genética , Glioblastoma/patologia , Histonas/genética , Células-Tronco Neoplásicas/patologia , Proteína Vesicular 1 de Transporte de Glutamato/genética , Apoptose , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Western Blotting , Diferenciação Celular , Movimento Celular , Proliferação de Células , Imunoprecipitação da Cromatina , Epigênese Genética , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Genoma Humano , Glioblastoma/metabolismo , Humanos , Células-Tronco Neoplásicas/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas , Proteína Vesicular 1 de Transporte de Glutamato/metabolismo
18.
OMICS ; 19(1): 1-11, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25562196

RESUMO

Early detection is vital to improve the overall survival rate of bladder cancer (BCa) patients, yet there is a lack of a reliable urine-based assay for early detection of BCa. Urine metabolites represented a potential rich source of biomarkers for BCa. This study aimed to develop a metabolomics approach for high coverage discovery and identification of metabolites in urine samples. Urine samples from 23 early stage BCa patients and 21 healthy volunteers with minimum sample preparations were analyzed by a short 30 min UPLC-HRMS method. We detected and quantified over 9000 unique UPLC-HRMS features, which is more than four times than about 2000 features detected in previous urine metabolomic studies. Furthermore, multivariate OPLS-DA classification models were established to differentiate urine samples from bladder cancer cohort and normal health cohort. We identified three BCa-upregulated metabolites: nicotinuric acid, trehalose, AspAspGlyTrp, and three BCa-downregulated metabolites: inosinic acid, ureidosuccinic acid, GlyCysAlaLys. Finally, analysis of six post-surgery BCa urine samples showed that these BCa-metabolomic features reverted to normal state after tumor removal, suggesting that they reflected metabolomic features associated with BCa. ROC analyses using two linear regression models to combine the identified markers showed a high diagnostic performance for detecting BCa with AUC (area under the ROC curve) values of 0.919 to 0.934. In summary, we developed a high coverage metabolomic approach that has potential for biomarker discovery in cancers.


Assuntos
Biomarcadores Tumorais/urina , Neoplasias da Bexiga Urinária/diagnóstico , Neoplasias da Bexiga Urinária/urina , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
19.
Zhonghua Zhong Liu Za Zhi ; 37(10): 759-63, 2015 Oct.
Artigo em Chinês | MEDLINE | ID: mdl-26813595

RESUMO

OBJECTIVE: To identify SNPs in the miRNA genes in colorectal cancer (CRC) patients and to investigate their association with CRC. METHODS: DNAs were isolated from 30 CRC tumor tissues and 30 tumor-adjacent tissues, and subjected to target capture using a custom miRNA chip covering 685 miRNA genes from NimbleGen. The captured DNAs were then sequenced using the Illumina's sequencing technology, and the data were analyzed. RESULTS: We identified 64 SNPs in 43 miRNA genes and most of these SNPs are novel SNPs not reported previously. Prediction of functional consequences of the SNPs using TargetScan and miRSNP showed that SNPs of hsa-mir-1273-G/A, hsa-mir-548h-3-C/U, hsa-mir-1290-A/G, and hsa-mir-1273-C/U resulted in reduction of their mature miRNA abundance. SNPs of hsa-mir-376b-C/G, hsa-mir-604-T/C, hsa-mir-1268-T/G and hsa-mir-146a-C/G resulted in changes in their targeted genes. Finally, we focused on the analysis of SNPs in mir-146a and we found that mir-146a rs1052918 C>G was predicted to promote tumorigenesis via the Wnt signaling pathway. CONCLUSIONS: SNPs in the miRNA genes are important for tumorigenesis. The changes by hsa-mir-146a rs1052918 C>G may result in loss of Wnt, constant activation of the Wnt signaling pathway, and uncontrolled cell proliferation and tumor progression.


Assuntos
Neoplasias Colorretais/genética , MicroRNAs/fisiologia , Polimorfismo de Nucleotídeo Único , Análise de Sequência de DNA , Sequência de Bases , Proliferação de Células , Neoplasias Colorretais/patologia , Humanos , Análise de Sequência com Séries de Oligonucleotídeos
20.
BMC Neurol ; 14: 207, 2014 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-25366337

RESUMO

BACKGROUND: SOX4 is a transcription factor required for tissue development and differentiation in vertebrates. Overexpression of SOX4 has been reported in many cancers including glioblastoma multiforme (GBM), however, the underlying mechanism of actions has not been studied. In this study, we investigated the role of SOX4 in GBM. METHODS: Kaplan-Meier analysis was performed to assess the association between SOX4 expression levels and survival times in primary GBM samples. Cre/lox P system was used to generate gain or loss of SOX4 in GBM cells, and microarray analysis uncovered the regulation network of SOX4 in GBM cells. RESULTS: High SOX4 expression was significantly associated with good prognosis of primary GBMs. SOX4 inhibited the growth of GBM cell line LN229, A172G and U87MG, partly via the activation of p53-p21 signaling and down-regulation of phosphorylated AKT1. Gene expression profiling and subsequent gene ontology analysis showed that SOX4 influenced several key pathways including the Wnt/ beta-catenin and TGF-beta signaling pathways. CONCLUSIONS: Our study found that SOX4 acts as a tumor suppressor in GBM cells by induce cell cycle arrest and inhibiting cell growth.


Assuntos
Glioblastoma/patologia , Fatores de Transcrição SOXC/genética , Linhagem Celular Tumoral , Proliferação de Células , Regulação para Baixo , Pontos de Checagem da Fase G1 do Ciclo Celular/fisiologia , Glioblastoma/genética , Humanos , Estimativa de Kaplan-Meier , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA