Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Bioengineered ; 13(2): 2878-2888, 2022 02.
Article in English | MEDLINE | ID: mdl-35040754

ABSTRACT

Lung adenocarcinoma is the main cause of the excessive mortality for patients who lives with lung cancers. According to the GEPIA database analysis, GATA5 and ARHGAP9 were found to be low expressed in lung adenocarcinoma, and they were positively correlated, and in addition ARHGAP9 low expression was associated with poor prognosis in lung adenocarcinoma. Therefore, the present study focused on the effect of promoting GATA5 to induce ARHGAP9 on the malignant process of lung adenocarcinoma cells. The expressions of GATA5 and ARHGAP9 were measured with Western blot and RT-qPCR. With the adoption of CCK-8, EDU staining, transwell and colony formation, the cell viability, proliferation, invasion and tumorigenesis ability were detected, respectively. In addition, the wound healing and Western blot were employed to evaluate migration and metastasis-related proteins individually. Moreover, the luciferase activity as well as the binding of GATA5 and ARHGAP9 promoters were detected by luciferase report and ChIP. After further comprehensive assessments, the results confirmed that GATA5 could successfully activate ARHGAP9. Moreover, ARHGAP9 upregulation remarkably inhibited lung adenocarcinoma cell proliferation, invasion and migration as compared to the control group. More importantly, GATA5 silencing reversed the inhibitory effect of ARHGAP9 upregulation on the malignant progression of lung adenocarcinoma cells. To conclude, the present study successfully demonstrated for the first time that GATA5-induced ARHGAP9 upregulation has a protective effect on lung adenocarcinoma cells.


Subject(s)
Adenocarcinoma of Lung/pathology , GATA5 Transcription Factor/physiology , GTPase-Activating Proteins/genetics , Lung Neoplasms/pathology , A549 Cells , Adenocarcinoma of Lung/diagnosis , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/mortality , Biomarkers, Tumor/genetics , Cells, Cultured , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Prognosis , Promoter Regions, Genetic , Up-Regulation/genetics
2.
Front Immunol ; 12: 704965, 2021.
Article in English | MEDLINE | ID: mdl-34456915

ABSTRACT

Interferon-induced transmembrane protein 3 (IFITM3) is an interferon-induced membrane protein, which has been identified as a functional gene in multiple human cancers. The role of IFITM3 in cancer has been preliminarily summarized, but its relationship to antitumor immunity is still unclear. A pancancer analysis was conducted to investigate the expression pattern and immunological role of IFITM3 based on transcriptomic data downloaded from The Cancer Genome Atlas (TCGA) database. Next, correlations between IFITM3 and immunological features in the bladder cancer (BLCA) tumor microenvironment (TME) were assessed. In addition, the role of IFITM3 in estimating the clinical characteristics and the response to various therapies in BLCA was also evaluated. These results were next confirmed in the IMvigor210 cohort and a recruited cohort. In addition, correlations between IFITM3 and emerging immunobiomarkers, such as microbiota and N6-methyladenosine (m6A) genes, were assessed. IFITM3 was enhanced in most tumor tissues in comparison with adjacent tissues. IFITM3 was positively correlated with immunomodulators, tumor-infiltrating immune cells (TIICs), cancer immunity cycles, and inhibitory immune checkpoints. In addition, IFITM3 was associated with an inflamed phenotype and several established molecular subtypes. IFITM3 expression also predicted a notably higher response to chemotherapy, anti-EGFR therapy, and immunotherapy but a low response to anti-ERBB2, anti-ERBB4, and antiangiogenic therapy. In addition, IFITM3 was correlated with immune-related microbiota and m6A genes. In addition to BLCA, IFITM3 is expected to be a marker of high immunogenicity in most human cancers. In conclusion, IFITM3 expression can be used to identify immuno-hot tumors in most cancers, and IFITM3 may be a promising pancancer biomarker to estimate the immunological features of tumors.


Subject(s)
Biomarkers, Tumor/immunology , Databases, Nucleic Acid , Gene Expression Regulation, Neoplastic/immunology , Transcriptome/immunology , Tumor Microenvironment/immunology , Urinary Bladder Neoplasms/immunology , Humans , Inflammation/immunology , Membrane Proteins , RNA-Binding Proteins
3.
Cell Death Dis ; 11(9): 760, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32934206

ABSTRACT

Palbociclib, a CDK4/6 inhibitor, has been granted accelerated approval by US FDA for hormone receptor-positive HER2-negative metastatic breast cancer. To determine potential biomarkers of palbociclib sensitivity to assist in patient selection and clinical development, we investigated the effects of palbociclib in a panel of molecularly characterized breast cancer cell lines. We quantified palbociclib sensitivity and c-myc expression in 11 breast cancer cell lines, 124 breast cancer samples, and The Cancer Genome Atlas database. We found non-TNBC subtypes were more sensitive to palbociclib than TNBC. Activation of c-myc led to differential palbociclib sensitivities, and further inhibition of c-myc enhanced palbociclib sensitivity. Moreover, we identified for the first time a c-myc/miR-29b-3p/CDK6 axis in breast cancer that could be responsible for c-myc-induced palbociclib insensitivity, in which c-myc activation resulted in downregulation of miR-29b-3p, further activated CDK6 and inhibited cell-cycle arrest at G1 phase. Moreover, downregulated (inactived) c-myc-induced oncogenic addiction could increase palbociclib efficacy, using both Xenograft model and patient-derived tumor xenograft (PDTX) model. Our finding extends the concept of combined blockade of the CDK4/6 and c-myc signaling pathways to increase palbociclib sensitivity, making c-myc a promising biomarker for palbociclib sensitivity in breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , Cyclin-Dependent Kinase 6/metabolism , Piperazines/pharmacology , Proto-Oncogene Proteins c-myc/metabolism , Pyridines/pharmacology , Antineoplastic Agents/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cyclin-Dependent Kinase 6/genetics , Female , Genes, myc , Humans , Prognosis , Proto-Oncogene Proteins c-myc/genetics
4.
Carbohydr Polym ; 233: 115841, 2020 Apr 01.
Article in English | MEDLINE | ID: mdl-32059893

ABSTRACT

A novel bio-based flame retardant, ammonium phosphate starch carbamates (APSC) was synthesized from starch, phosphoric acid and urea to achieve better thermal stability and char formation. Flame retardant expandable polystyrene foams (EPS) were prepared by coating method with APSC. The fire resistance evaluation of EPS composites by limiting oxygen index (LOI), vertical burning (UL-94) and cone calorimeter tests indicated that the addition of 47 wt.% of APSC enhanced the LOI from 17.6%-35.2%, with V-0 rating in UL-94, and decreased the peak heat release rate from 666 kW/m2 to 316 kW/m2. Moreover, the total smoke production was also sharply decreased from 70 m2 of pure EPS to 17 m2. By the presence of APSC, the substantial char formation prevented the heat and oxygen transfer, and interrupted the releasing of flammable products, thus protecting the EPS foam from burning.

5.
Neoplasia ; 21(6): 533-544, 2019 06.
Article in English | MEDLINE | ID: mdl-31029033

ABSTRACT

Treatment options are limited for patients with triple negative breast cancer (TNBC). Understanding genes that participate in cancer progression and DNA damage response (DDR) may improve therapeutic strategies for TNBC. DAXX, a death domain-associated protein, has been reported to be critically involved in cancer progression and drug sensitivity in multiple cancer types. However, its role in breast cancer, especially for TNBC, remains unclear. Here, we demonstrated a tumor suppressor function of DAXX in TNBC proliferation, colony formation, and migration. In Mouse Xenograft Models, DAXX remarkably inhibited tumorigenicity of TNBC cells. Mechanistically, DAXX could directly bind to the promoter region of RAD51 and impede DNA damage repair, which impacted the protection mechanism of tumor cells that much depended on remaining DDR pathways for cell growth. Furthermore, DAXX-mediated inefficient DNA damage repair could sensitize BRCA-proficient TNBC cells to PARP inhibitors. Additionally, we identified that dual RAD51 and PARP inhibition with RI-1 and ABT888 significantly reduced TNBC growth both in vitro and in vivo, which provided the first evidence of combining RAD51 and PARP inhibition in BRCA-proficient TNBC. In conclusion, our data support DAXX as a modulator of DNA damage repair and suppressor of TNBC progression to sensitize tumors to the PARP inhibitor by repressing RAD51 functions. These provide an effective strategy for a better application of PARP inhibition in the treatment of TNBC.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Nuclear Proteins/genetics , Poly (ADP-Ribose) Polymerase-1/genetics , Rad51 Recombinase/genetics , Triple Negative Breast Neoplasms/drug therapy , Tumor Suppressor Proteins/genetics , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Animals , Benzimidazoles/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Co-Repressor Proteins , DNA Damage/drug effects , DNA Repair/drug effects , Female , Heterografts , Humans , Mice , Molecular Chaperones , Morpholines/pharmacology , Nuclear Proteins/antagonists & inhibitors , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Promoter Regions, Genetic/drug effects , Pyrroles/pharmacology , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Tumor Suppressor Proteins/antagonists & inhibitors
6.
Int J Biol Sci ; 15(3): 522-532, 2019.
Article in English | MEDLINE | ID: mdl-30745839

ABSTRACT

The dysregulation of cyclin D -Cyclin-dependent kinase 4/6 (CDK4/6)-Rb axis has been implicated in breast cancer progression and the selective CDK4/6 inhibitors have shown effective activity in advanced breast cancer, especially in tumors driven by the estrogen receptor (ER). However, resistance to these small molecular inhibitors has become an inevitable clinical issue after their initial use. Here, we investigated the potential mechanism of resistance by establishing a CDK4/6 inhibitor palbociclib-resistant breast cancer cell line (MCF-7pR). After prolonged exposure to palbociclib, we detected the loss of the ER signaling and an increase in androgen receptor (AR). Moreover, we demonstrated more localization of AR in the cell nucleus of MCF-7pR compared to the parental cell (MCF-7). We also reported that AR could promote the progression of the cell cycle. Blockade of AR signaling could reduce the level of the relative G1-S cyclins, abolish Rb phosphorylation and inhibit the activation of transcriptional programs in S phase. Furthermore, dual inhibition of AR and CDK4/6 could reverse the resistance of palbociclib both in vitro and in vivo. In sum, our studies provide evidence that AR activation promotes cell cycle progression and cell proliferation in CDK4/6 inhibitor resistance, and identify AR inhibition as a putative novel therapeutic strategy to treat CDK4/6 inhibitor resistance in cancer.


Subject(s)
Breast Neoplasms/metabolism , Cell Cycle/drug effects , Cell Survival/drug effects , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , Piperazines/pharmacology , Pyridines/pharmacology , Receptors, Androgen/isolation & purification , Animals , Benzamides , Blotting, Western , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 6/genetics , Female , Humans , Immunohistochemistry , Immunoprecipitation , MCF-7 Cells , Mice , Nitriles , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/pharmacology , Receptors, Androgen/genetics
7.
Mol Cancer ; 17(1): 99, 2018 07 14.
Article in English | MEDLINE | ID: mdl-30007403

ABSTRACT

Triple negative breast cancer (TNBC) is a heterogeneous disease with aggressive behavior and poor prognosis. Genomic sequencing has detected a distinctive mutational portrait of both the germline and somatic alterations in TNBC, which is staggeringly different from other breast cancer subtypes. The clinical utility of sequencing germline BRCA1/2 genes has been well established in TNBC. However, for other predisposition genes, studies concerning the risk and penetrance to TNBC are relatively scarce. Very few recurrent mutations, including TP53 and PI3KCA mutations, together with a long tail of individually rare mutations occur in TNBC. These combined effects of genomic alterations drive TNBC progression. Given the complexity and heterogeneity of TNBC, clinical interpretation of the genomic alterations in TNBC may pave a new way for the treatment of TNBC. In this review, we summarized the germline and somatic mutation profiles of TNBC and discussed the current and upcoming therapeutic strategies targeting the mutant proteins or pathways to enable tailored-therapeutics.


Subject(s)
Antineoplastic Agents/therapeutic use , DNA Mutational Analysis/methods , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Adult , Antineoplastic Agents/pharmacology , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Clinical Trials as Topic , Female , Germ-Line Mutation , Humans , Middle Aged , Molecular Targeted Therapy/methods , Nuclear Proteins/genetics , Transcription Factors/genetics , Tumor Suppressor Protein p53/genetics
8.
Cell Death Differ ; 25(12): 2209-2220, 2018 12.
Article in English | MEDLINE | ID: mdl-29844570

ABSTRACT

Androgen receptor (AR) is emerging as a novel prognostic biomarker in triple-negative breast cancer (TNBC), but the underlying mechanisms remain unknown. As accumulating evidence has shown that long non-coding RNAs (lncRNAs) regulate important cancer hallmarks, we hypothesised that AR-regulated lncRNAs might play roles in TNBC progression. Here, we performed experiments with or without DHT treatment in three TNBC cell lines, and we identified an AR negatively induced lncRNA (ARNILA), which correlated with poor progression-free survival (PFS) in TNBC patients and promoted epithelial-mesenchymal transition (EMT), invasion and metastasis in vitro and in vivo. Subsequently, we demonstrated that ARNILA functioned as a competing endogenous RNA (ceRNA) for miR-204 to facilitate expression of its target gene Sox4, which is known to induce EMT and contribute to breast cancer progression, thereby promoting EMT, invasion and metastasis of TNBC. Our findings not only provide new insights into the mechanisms of lncRNA in regulating AR but also suggest ARNILA as an alternative therapeutic target to suppress metastasis of TNBC patients.


Subject(s)
Mammary Neoplasms, Experimental/metabolism , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Receptors, Androgen/metabolism , Triple Negative Breast Neoplasms/metabolism , Animals , Cell Movement , Female , Humans , Mammary Neoplasms, Experimental/diagnostic imaging , Mice , Mice, Inbred BALB C , Mice, Nude , Oligonucleotide Array Sequence Analysis , Positron Emission Tomography Computed Tomography , RNA, Long Noncoding/genetics , Triple Negative Breast Neoplasms/diagnostic imaging , Tumor Cells, Cultured
9.
Neoplasia ; 20(5): 478-488, 2018 05.
Article in English | MEDLINE | ID: mdl-29605721

ABSTRACT

Triple negative breast cancer (TNBC) is a highly aggressive subtype of breast cancer that poses a clinical challenge. Thus, new therapy strategies are urgently needed. The selective WEE1 inhibitor, AZD1775, has shown strong anti-proliferative effects on a variety of tumors. Here, we first demonstrate that inhibition of ATR by selective inhibitor AZD6738 can enhance AZD1775-caused growth inhibition in TNBC. Our results show that the enhanced cell death is attributed to repressed DNA damage repair and excessive replication stress, thereby causing increased DNA damage reflected by accumulation of the DNA double-strand-break marker γH2AX. On the other hand, combined treatment with AZD6738 and AZD1775 forces mitotic entry of cells with DNA damages by activating CDK1 activity, inducing severely aberrant mitosis and mitotic catastrophe, ultimately resulting in cell death. Dual inhibition of WEE1 and ATR also inactivated RAD51-mediated homologous recombination, which sensitized TNBC cells to cisplatin and PARP inhibitor. Here, based on the preclinical results that ATR inhibition synergizes with WEE1 inhibition in TNBC, we propose that this combination therapy alone, or in parallel with chemotherapy, represents an innovative and potent targeted therapy in TNBC.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , Nuclear Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Triple Negative Breast Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Damage/drug effects , DNA Repair/drug effects , Female , Humans , Indoles , MCF-7 Cells , Mice , Mitosis/drug effects , Morpholines , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Pyrimidinones , Sulfonamides , Sulfoxides/pharmacology , Xenograft Model Antitumor Assays/methods
10.
Cancer Biol Ther ; 18(6): 369-378, 2017 06 03.
Article in English | MEDLINE | ID: mdl-28494179

ABSTRACT

Treatment of triple negative breast cancer (TNBC) has been a big challenge since it is defined. To date, platinum-based chemotherapy has played a significant role in the treatment of TNBC patients. However, some patients do not respond to platinum salts or gradually develop chemoresistance, resulting in little effect, or even some adverse effects. Here, we review numerous preclinical and clinical investigations to summarize possible mechanisms and potential predictive biomarkers of platinum in TNBC. The homologous recombination deficiency (HRD) resulting from the loss of BRCA function is the main rationale of platinum efficacy in TNBC. BRCA mutation and methylation have been demonstrated to be important potential biomarkers. Based on genome-wide effects, BRCA-like classifier can identify the functional loss of BRCA and work as the predictor. HRD score that is able to identify the "BRCAness" and predict the sensitivity of platinum is increasingly considered. Taken together, all findings suggest that HR deficiency profile encompassed by BRCA mutation and high HRD score could predict response to platinum, even to other DNA-damage inducing agents. p53 family members and molecular subtypes of TNBC are also important alternative considerations for predicting platinum response based on the preclinical trials. Currently, tumor infiltrating lymphocyte level and thrombocytopenia are emerging as predictive biomarkers.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/genetics , Platinum Compounds/therapeutic use , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Antineoplastic Agents/pharmacology , DNA End-Joining Repair , Female , Genomic Instability , Humans , Mutation , Platinum Compounds/pharmacology , Treatment Outcome , Triple Negative Breast Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...