Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Eur J Radiol ; 135: 109512, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33429302

ABSTRACT

PURPOSE: To develop a combined nomogram by incorporating the Memorial Sloan Kettering Cancer Center (MSKCC) nomogram and ultrasound (US)-based radiomics score (Radscore) for predicting sentinel lymph node (SLN) metastasis in invasive breast cancer. MATERIALS AND METHODS: This retrospective study was approved by the ethics committee of our institution, and written informed consent was waived. A total of 452 patients with invasive breast cancer who received SLN Biopsy in a single center were included between January 2016 and December 2019. The patients were divided into a training set (n = 318) and a validation set (n = 134). A total of 1216 features were extracted from the regions of interest (ROIs) of the tumors on conventional ultrasound. The maximum relevance minimum redundancy (mRMR) and the least absolute shrinkage and selection operator (LASSO) algorithm were used to build the Radscore. Afterward, the diagnostic performance was assessed and validated. Comparison of receiver operating characteristic (ROC) curves and decision curve analysis (DCA) were performed to evaluate the incremental value of the combined model. RESULTS: Obtained from 18 features, the Radscore indicated a favorable discriminatory capability in the training set with an area under the curve (AUC) of 0.834, whereas a value of 0.770 was observed in the validation set. The AUC of the combined model was 0.901 (95 % confidence interval (95 % CI): 0.865-0.938) in the training set and 0.833 (95 % CI: 0.788-0.878) in the validation set. Both of them were superior to MSKCC or imaging Radscore alone (P < 0.05). DCA demonstrated that the combined model was superior to the others in terms of clinical practicability. CONCLUSION: Preoperative US-based Radscore can improve the accuracy of clinical MSKCC nomogram for SLN metastasis prediction in breast cancer.


Subject(s)
Breast Neoplasms , Nomograms , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/surgery , Humans , Lymph Nodes/diagnostic imaging , Lymphatic Metastasis/diagnostic imaging , ROC Curve , Retrospective Studies , Sentinel Lymph Node Biopsy
2.
J Exp Clin Cancer Res ; 39(1): 21, 2020 Jan 27.
Article in English | MEDLINE | ID: mdl-31987045

ABSTRACT

In the original publication of this article [1], the molecular weight of RBMS3 was incorrectly noted as 38 KDa within Fig 1A, Fig 2A and Fig 2B. The figures have been updated to list the correct molecular weight of RBMS3 as 41 KDa.

3.
Oncogene ; 38(33): 6123-6141, 2019 08.
Article in English | MEDLINE | ID: mdl-31285549

ABSTRACT

Most N6-methyladenosine (m6A) associated regulatory proteins (i.e., m6A writer, eraser, and reader proteins) are involved in the pathogenesis of various cancers, mostly in m6A-dependent manners. As a component in the m6A 'writers', KIAA1429 is reported to be an RNA-binding protein and involved in the m6A modification, mRNA splicing and processing. Till now, the functions of KIAA1429 in tumorigenesis and related mechanism have not been reported. In the present study, we found KIAA1429 was highly expressed in breast cancer tissues, but frequently down-regulated in non-cancerous breast tissues. The overall survival of breast cancer patients with high-expression KIAA1429 was significantly shorter than those with low-expression KIAA1429. Then, we demonstrated that KIAA1429 was associated with breast cancer proliferation and metastasis in vivo and in vitro. The potential targeting genes of KIAA1429 in breast cancer were identified by RNA immunoprecipitation sequencing. One of these genes is cyclin-dependent kinase 1 (CDK1), which plays an oncogenic role in cancers. Furthermore, we confirmed that KIAA1429 played its oncogenic role in breast cancer by regulating CDK1 by an m6A-independent manner. 5'-fluorouracil was found to be very effective in reducing the expression of KIAA1429 and CDK1 in breast cancer. These findings indicated that KIAA1429 could promote breast cancer progression and was correlated with pathogenesis. It may represent a promising therapeutic strategy on breast cancer, especially in combination with CDK1 treatment.


Subject(s)
Adenosine/analogs & derivatives , Breast Neoplasms/genetics , CDC2 Protein Kinase/genetics , Oncogenes/physiology , RNA-Binding Proteins/physiology , Adenosine/metabolism , Adenosine/physiology , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , CDC2 Protein Kinase/metabolism , Carcinogenesis/genetics , Carcinogenesis/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Mice , Mice, Nude , RNA-Binding Proteins/genetics , Tumor Cells, Cultured
4.
J Exp Clin Cancer Res ; 38(1): 105, 2019 Feb 28.
Article in English | MEDLINE | ID: mdl-30819235

ABSTRACT

BACKGROUND: Metastasis remains the biggest obstacle for breast cancer treatment. Therefore, identification of specific biomarker of metastasis is very necessary. The RNA binding protein 3 (RBMS3) acts as a tumor suppressor in various cancers. Whereas, its role and underlying molecular mechanism in breast cancer is far from elucidated. METHODS: Quantitative real-time PCR and western blots were carried out to determine the expression of RBMS3 in breast cancer cells and tissues. Transwell and in vivo metastasis assay were conducted to investigate the effects of RBMS3 on migration, invasion and metastasis of breast cancer cells. Transcriptome sequencing was applied to screen out the differential gene expression affected by RBMS3. RNA immunoprecipitation assay combined with luciferase reporter assay were performed to explore the direct correlation between RBMS3 and Twist1 mRNA. RESULTS: RBMS3 was downregulated in breast cancer and ectopic expression of RBMS3 contributed to inhibition of cell migration, invasion in vitro and lung metastasis in vivo. Furthermore, RBMS3 negatively regulated Twsit1 expression via directly binding to 3'-UTR of Twist1 mRNA, and thereby decreased Twist1-induced expression of matrix metalloproteinase 2 (MMP2). Additionally, Twist1-induced cell migration, invasion and lung metastasis could be reversed by the upregulation of RBMS3. CONCLUSIONS: In summary, our study revealed a novel mechanism of the RBMS3/Twsit1/MMP2 axis in the regulation of invasion and metastasis of breast cancer, which may become a potential molecular marker for breast cancer treatment.


Subject(s)
Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic/physiology , Nuclear Proteins/biosynthesis , RNA-Binding Proteins/metabolism , Trans-Activators/metabolism , Twist-Related Protein 1/biosynthesis , Animals , Breast Neoplasms/metabolism , Cell Movement/physiology , Female , Heterografts , Humans , Matrix Metalloproteinase 2/biosynthesis , Mice, Inbred BALB C , Neoplasm Invasiveness/pathology
5.
Oncol Rep ; 41(1): 455-464, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30542717

ABSTRACT

A total of 70% of breast cancers express the estrogen receptor (ER)α; therefore, targeting the ER may be an effective endocrine therapy with which to inhibit breast cancer growth. Tamoxifen is the most common­used clinically used drug for the treatment of advanced or metastatic ER­positive (ER+) breast cancer. However, a substantial proportion of patients become resistant to endocrine therapies. To overcome this limitation, in this stud, we sought to maximize the benefits associated with tamoxifen therapy via drug combination strategies. We demonstrated that rapamycin, an FDA­approved mammalian target of rapamycin (mTOR) inhibitor, enhanced the effects of endocrine therapy with tamoxifen, and the concentration of tamoxifen required for ER+ breast cancer cell growth inhibition was substantially reduced. Moreover, treatment with rapamycin plus tamoxifen significantly inhibited tumor growth in vivo. In addition, this synergistic effect may be mediated by the induction of p73. We revealed a novel mechanism in which p73 increases ERα expression by directly binding to the promoter region of the ERα gene. Taken together, the findings of this study indicate that combination therapy with rapamycin and tamoxifen underlying p73­mediated ERα expression may provide new insight into the drug combination for the treatment of ER+ breast cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Estrogen Receptor alpha/genetics , Sirolimus/pharmacology , Tamoxifen/pharmacology , Tumor Protein p73/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , Mice , Mice, Nude , RNA, Small Interfering/metabolism , Sirolimus/therapeutic use , Tamoxifen/therapeutic use , Tumor Protein p73/genetics , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
6.
Oncol Lett ; 15(2): 1737-1743, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29434869

ABSTRACT

Natural plants offer a treasure trove of resources for anticancer drug development. Clematis are widely used in Traditional Chinese Medicine. However, studies on the active substances in Clematis are scarce. In the present study, four monomer compounds were successfully extracted from this species and their inhibitory effects on the growth of breast cancer cells were investigated using bioactivity tests. Among them, Clematis hederagenin saponin (CHS) belongs to the class of triterpenoid saponins. CHS showed cytotoxic effects on breast cancer cells in a dose- and time-dependent manner. The compound also induced apoptosis in breast cancer cells in a time-dependent manner. Further investigation into the underlying mechanisms of apoptosis induction in breast cancer cells showed that the compound significantly reduced mitochondrial Apaf-1 and cytochrome c proteins in breast cancer cells. In addition, it upregulated the activities of caspase-3 and -9. In conclusion, CHS induced apoptosis in breast cancer cells through regulation of the mitochondrial apoptosis pathway. The results suggest that the hederagenin saponin extracted from Clematis ganpiniana offers great potential as a novel anti-breast cancer drug.

7.
Biochem Biophys Res Commun ; 495(1): 775-780, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29146183

ABSTRACT

Many natural products have been shown to have inhibitory effects on the metastatic process of various cancers including breast cancer. An active triterpenoid saponin D Rhamnose ß-hederin (DRß-H) from Clematis ganpiniana, has been known induce the apoptosis of breast cancer cells, but the effect of DRß-H on the metastasis of breast cancer cells is largely unknown. In this study, we demonstrated that a non-cytotoxic concentration of DRß-H markedly suppressed wound healing migration, migration through the chamber and invasion through the matrigel. In addition, DRß-H regulated expression of RNPC1, E-cadherin proteins of MDA-MB-231 cells. Furthermore, RNPC1 knockdown decreased the DRß-H-induced up-regulation of RNPC1 and E-Cadherin in MDA-MB-231 cells. RNPC1 knockdown reduced the anti-metastasis activities of DRß-H, meaning that the up-rugulation of RNPC1 by DRß-H is essential for its anti-metastatis activities. These results suggest that DRß-H might be a potential therapeutic candidate for the treatment of breast cancer metastasis.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Movement , Neoplasm Metastasis/prevention & control , Oleanolic Acid/analogs & derivatives , Saponins/administration & dosage , Antineoplastic Agents/administration & dosage , Breast Neoplasms/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Female , Humans , Neoplasm Invasiveness , Neoplasm Metastasis/physiopathology , Oleanolic Acid/administration & dosage , Treatment Outcome
8.
Br J Cancer ; 117(5): 675-684, 2017 Aug 22.
Article in English | MEDLINE | ID: mdl-28683467

ABSTRACT

BACKGROUND: The transforming growth factor-ß (TGF-ß) pathway plays a vital role in driving cancer cell epithelial-mesenchymal transition (EMT). Zonula occludens-1 (ZO-1), which is downregulated in response to TGF-ß, is able to control endothelial cell-cell tension, cell migration, and barrier formation. However, the molecular mechanism of how TGF-ß regulates ZO-1 expression remains unclear. METHODS: Breast cancer cells were treated with TGF-ß to induce an EMT progress. Chromatin immunoprecipitation and dual-luciferase reporter assay were performed to investigate direct relationship between Snail and RNA binding motif protein 38 (RBM38). The RNA immunoprecipitation combined with RNA electrophoretic mobility shift assay and dual-luciferase reporter assay were conducted to testify direct relationship between RBM38 and ZO-1. The ZO-1 siRNA was transfected to breast cancer cells that overexpress RBM38 and the control, followed by transwell and Matrigel invasion assays to examine cell migratory and invasive ability. RESULTS: Transforming growth factor-ß induced a remarkable downregulation of RBM38 in breast cancer that was directly regulated by transcription repressor Snail targeting the E-box elements in promoter region of RBM38 gene. Additionally, RBM38 positively regulated ZO-1 transcript via directly binding to AU/U-rich elements in its mRNA 3'-UTR. Moreover, by magnifying RBM38 expression, cell migration and invasion mediated by knockdown of ZO-1 in breast cancer were reversed. CONCLUSIONS: All the results clarified a linear regulation relationship among Snail, RBM38, and ZO-1, implicating RBM38 as a pivotal mediator in TGF-ß-induced EMT in breast cancer.


Subject(s)
Epithelial-Mesenchymal Transition , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Zonula Occludens-1 Protein/genetics , Zonula Occludens-1 Protein/metabolism , 3' Untranslated Regions , Benzamides/pharmacology , Breast Neoplasms/chemistry , Cell Movement/drug effects , Cell Movement/genetics , Dioxoles/pharmacology , Down-Regulation , E-Box Elements/genetics , Epithelial-Mesenchymal Transition/drug effects , Female , Gene Expression , Humans , MCF-7 Cells , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/antagonists & inhibitors , RNA, Small Interfering/genetics , RNA-Binding Proteins/analysis , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Snail Family Transcription Factors/genetics , Transfection , Transforming Growth Factor beta/pharmacology , Zonula Occludens-1 Protein/analysis
9.
J Exp Clin Cancer Res ; 36(1): 49, 2017 04 11.
Article in English | MEDLINE | ID: mdl-28399911

ABSTRACT

BACKGROUND: RNA-binding protein 38 (RBM38) is a member of the RNA recognition motif (RRM) family of RNA-binding proteins (RBPs). RBM38 often exerts its function by forming regulatory loops with relevant genes. c-Myc is an oncogenic transcription factor that is upregulated in one-third of breast cancers and involved in many cellular processes in this malignancy. In our previous study, RBM38 was identified as a tumor suppressor in breast cancer. In the present study, we investigated the mechanisms underlying the regulation of this tumor suppressor. METHODS: Lentivirus transfections, Western blotting analysis, qRT-PCR and immunohistochemistry were employed to study the expression of c-Myc and RBM38. Chromatin immunoprecipitation and dual-luciferase reporter assays were performed to investigate the direct relationship between c-Myc protein and the RBM38 gene. RNA immunoprecipitation combined with dual-luciferase reporter assays was conducted to confirm the direct relationship between the RBM38 protein and the c-Myc transcript. RESULTS: Knockdown of c-Myc increased RBM38 expression by binding directly to specific DNA sequences (5'-CACGTG-3'), known as the E-box motif, in the promoter region of RBM38 gene. Additionally, RBM38 destabilized the c-Myc transcript by directly targeting AU-rich elements (AREs) in the 3'-untranslated region (3'-UTR) of c-Myc mRNA to suppress c-Myc expression. Moreover, specific inhibitors of c-Myc transcriptional activity inhibited RBM38-induced suppression of growth, implying that RBM38 acts as a tumor suppressor via a mechanism that depends, at least partially, on the reduction of c-Myc expression in breast cancer. CONCLUSIONS: RBM38 and c-Myc form a unique mutually antagonistic RBM38-c-Myc loop in breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , 3' Untranslated Regions , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Neoplasm Staging , Promoter Regions, Genetic
10.
Oncotarget ; 8(10): 16387-16400, 2017 Mar 07.
Article in English | MEDLINE | ID: mdl-27634883

ABSTRACT

Progesterone receptor (PR) could activate transcriptional process involved in normal mammary gland proliferation and breast cancer development. Moreover, PR expression is an important marker of luminal breast cancer, which is associated with good prognosis and indicates better responding to endocrine therapies. The regulation of PR expression was studied mainly on its post-translational levels. In this study, we found PR was positively regulated by RNA-binding region-containing protein 1 (RNPC1), a RNA-binding protein, in PR positive breast cancer. Overexpression of RNPC1 increased, whereas knockdown of RNPC1 decreased, the level of PR protein and transcripts. Additionally, we demonstrated that RNPC1 could bind to PR mRNA via AU-rich elements (AREs) within PR 3'-untranslated region (3'-UTR) and then enhance PR mRNA stability. Moreover, we proved that progesterone-dependent PR functions which could induce breast cancer proliferation were enhanced by RNPC1, both in vitro and in vivo. Conclusively, we revealed a novel mechanism by which PR could be regulated by RNPC1 via stabilizing its mRNA.


Subject(s)
Breast Neoplasms/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Receptors, Progesterone/metabolism , 3' Untranslated Regions/genetics , AU Rich Elements/genetics , Animals , Blotting, Western , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Mice, Inbred NOD , Mice, SCID , Microscopy, Fluorescence , Progesterone/pharmacology , Progestins/pharmacology , Protein Binding , RNA Interference , RNA Stability/genetics , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Receptors, Progesterone/genetics , Reverse Transcriptase Polymerase Chain Reaction , Xenograft Model Antitumor Assays/methods
11.
Oncotarget ; 7(9): 9925-38, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26848530

ABSTRACT

PURPOSE: This work aims to identify differential metabolites and predicting molecular subtypes of breast cancer (BC). METHODS: Plasma samples were collected from 96 BC patients and 79 normal participants. Metabolic profiles were determined by liquid chromatography-mass spectrometry and gas chromatography-mass spectrometry based on multivariate statistical data analysis. RESULTS: We observed 64 differential metabolites between BC and normal group. Compared to human epidermal growth factor receptor 2 (HER2)-negative patients, HER2-positive group showed elevated aerobic glycolysis, gluconeogenesis, and increased fatty acid biosynthesis with reduced Krebs cycle. Compared with estrogen receptor (ER)-negative group, ER-positive patients showed elevated alanine, aspartate and glutamate metabolism, decreased glycerolipid catabolism, and enhanced purine metabolism. A panel of 8 differential metabolites, including carnitine, lysophosphatidylcholine (20:4), proline, alanine, lysophosphatidylcholine (16:1), glycochenodeoxycholic acid, valine, and 2-octenedioic acid, was identified for the classification of BC subtypes. These markers showed potential diagnostic value with average area under the curve at 0.925 (95% CI 0.867-0.983) for the training set (n=51) and 0.893 (95% CI 0.847-0.939) for the test set (n=45). CONCLUSION: Human plasma metabolomics is useful in identifying differential metabolites and predicting breast cancer subtypes.


Subject(s)
Breast Neoplasms/blood , Metabolome , Metabolomics/methods , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/blood , Biomarkers, Tumor/metabolism , Breast Neoplasms/classification , Breast Neoplasms/metabolism , Chromatography, Liquid , Female , Gas Chromatography-Mass Spectrometry , Humans , Mass Spectrometry , Middle Aged , Multivariate Analysis , Prognosis , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism
12.
Int J Mol Med ; 36(2): 473-84, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26046131

ABSTRACT

The RNA binding motif protein 38 (RBM38, also known as RNPC1) plays a pivotal role in regulating a wide range of biological processes, from cell proliferation and cell cycle arrest to cell myogenic differentiation. It was originally recognized as an oncogene, and was frequently found to be amplified in prostate, ovarian and colorectal cancer, chronic lymphocytic leukemia, colon carcinoma, esophageal cancer, dog lymphomas and breast cancer. In the present study, the complete RNPC1 gene was identified in a number of vertebrate genomes, suggesting that RNPC1 exists in all types of vertebrates, including fish, amphibians, birds and mammals. In the different genomes, the gene had a similar 4 exon/3 intron organization, and all the genetic loci were syntenically conserved. The phylogenetic tree demonstrated that the RNPC1 gene from the mammalian, bird, reptile and teleost lineage formed a species-specific cluster. A total of 34 functionally relevant single nucleotide polymorphisms (SNPs), including 14 SNPs causing missense mutations, 8 exonic splicing enhancer SNPs and 12 SNPs causing nonsense mutations, were identified in the human RNPC1 gene. RNPC1 was found to be expressed in bladder, blood, brain, breast, colorectal, eye, head and neck, lung, ovarian, skin and soft tissue cancer. In 14 of the 94 tests, an association between RNPC1 gene expression and cancer prognosis was observed. We found that the association between the expression of RNPC1 and prognosis varied in different types of cancer, and even in the same type of cancer from the different databases used. This suggests that the function of RNPC1 in these tumors may be multidimensional. The sex determining region Y (SRY)-box 5 (Sox5), runt-related transcription factor 3 (RUNX3), CCAAT displacement protein 1 (CUTL1), v-rel avian reticuloendotheliosis viral oncogene homolog (Rel)A, peroxisome proliferator-activated receptor γ isoform 2 (PPARγ2) and activating transcription factor 6 (ATF6) regulatory transcription factor binding sites were identified in the upstream (promoter) region of the RNPC1 gene, and may thus be involved in the effects of RNPC1 in tumors.


Subject(s)
Gene Expression Regulation, Neoplastic , Genomics/methods , Neoplasms/diagnosis , Neoplasms/genetics , RNA-Binding Proteins/genetics , Amino Acid Sequence , Animals , Humans , Molecular Sequence Data , Phylogeny , Polymorphism, Single Nucleotide , Prognosis , RNA-Binding Proteins/chemistry , Sequence Alignment , Vertebrates/genetics
13.
Oncotarget ; 6(14): 12264-78, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25881544

ABSTRACT

Estrogen receptors (ERs), including ERα and ERß, mainly mediate the genotype effect of estrogen. ERα is highly expressed in most breast cancers. Endocrine therapy is the most effective and safety adjunctive therapy for ER positive breast cancers. RNPC1, an RNA binding protein (RBP), post-transcriptionally regulating gene expression, is emerging as a critical mechanism for gene regulation in mammalian cells. In this study, we revealed RNPC1's capability of regulating ERα expression. There was a significant correlation between RNPC1 and ERα expression in breast cancer tissues. Ectopic expression of RNPC1 could increase ERα transcript and expression in breast cancer cells, and vice versa. Consistent with this, RNPC1 was able to bind to ERα transcript to increase its stability. Furthermore, overexpression of ERα could decrease the level of RNPC1 transcript and protein. It suggested a novel mechanism by which ERα expression was regulated via stabilizing mRNA. A regulatory feedback loop between RNPC1 and ERα was proved. It indicated that RNPC1 played a crucial role in ERα regulation in ER-positive breast cancers via binding to ERα mRNA. These findings might provide new insights into breast cancer endocrine therapy and ERα research.


Subject(s)
Breast Neoplasms/genetics , RNA, Messenger/genetics , RNA-Binding Proteins/metabolism , Receptors, Estrogen/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Humans
14.
PLoS One ; 10(3): e0120303, 2015.
Article in English | MEDLINE | ID: mdl-25799220

ABSTRACT

PURPOSE: Combination of percutaneous microwave ablation (PMWA) and intravenous injection of 131I-hypericin(IIIH) may bear potential as a mini-invasive treatment for tumor. The objective of this study was to assess the effect of PMWA and IIIH in breast tumor growth. METHODS: Ten New Zealand White rabbits bearing VX2 breast carcinomas were randomly divided into two groups (each 5 examples) and processed using PMWA followed by IIIH and IIIH alone. The IIIH activity was evaluated using planar scintigraphy, autoradiography and biodistribution analysis. The maximum effective safe dose of IIIH was found through 48 rabbits with VX2 breast tumor, which were randomized into six groups (n=8 per group). Subsequently, a further 75 rabbits bearing VX2 breast solid tumors were randomly divided into five groups (each 15 examples) and treated as follows: A, no treatment group; B, PMWA alone; C, IIIH alone; D, PMWA+IIIH×1 (at 8 h post-PMWA); and E, PMWA+IIIH×2 (at 8 h and at 8 days post-PMWA). The therapeutic effect was assessed by measurement of tumor size and performation of positron emission tomography/computed tomograph (PET/CT) scans, liver and renal function tests and Kaplan-Meier survival analysis. RESULTS: The planar scintigraphy findings suggested a significant uptake of 131I in necrotic tumor tissue. The autoradiography gray scales indicated higher selective uptake of IIIH by necrotic tissue, with significant differences between the groups with and those without necrotic tumor tissue (P<0.05). The maximum effective safe dose of IIIH was 1 mCi/kg. The PET/CT scans and tumor size measurement suggested improvements in treatment groups at all time points (P<0.01). Significant differences were detected among Groups A, B, D and E (P<0.05). Lower levels of lung metastasis were detected in Groups D and E (P<0.05). There were no abnormalities in liver and renal functions tests or other reported side effects. CONCLUSION: IIIH exhibited selective uptake by necrotic tumor tissue. Sequential therapy involving PMWA+IIIH was successfully inhibiting tumor growth and prolonging survival.


Subject(s)
Ablation Techniques , Antineoplastic Agents/therapeutic use , Carcinoma/surgery , Mammary Neoplasms, Experimental/surgery , Microwaves/therapeutic use , Perylene/analogs & derivatives , Radiosurgery , Animals , Anthracenes , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Carcinoma/drug therapy , Drug Evaluation, Preclinical , Female , Mammary Neoplasms, Experimental/drug therapy , Microwaves/adverse effects , Perylene/adverse effects , Perylene/pharmacokinetics , Perylene/therapeutic use , Rabbits , Tissue Distribution
15.
Int J Mol Med ; 35(1): 92-102, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25395235

ABSTRACT

Interleukin-34 (IL-34) is a novel cytokine, which is composed of 222 amino acids and forms homodimers. It binds to the macrophage colony-stimulating factor (M-CSF) receptor and plays an important role in innate immunity and inflammatory processes. In the present study, we identified the completed IL-34 gene in 25 various mammalian genomes and found that IL-34 existed in all types of vertebrates, including fish, amphibians, birds and mammals. These species have a similar 7 exon/6 intron gene organization. The phylogenetic tree indicated that the IL-34 gene from the primate lineage, rodent lineage and teleost lineage form a species-specific cluster. It was found mammalian that IL-34 was under positive selection pressure with the identified positively selected site, 196Val. Fifty-five functionally relevant single nucleotide polymorphisms (SNPs), including 32 SNPs causing missense mutations, 3 exonic splicing enhancer SNPs and 20 SNPs causing nonsense mutations were identified from 2,141 available SNPs in the human IL-34 gene. IL-34 was expressed in various types of cancer, including blood, brain, breast, colorectal, eye, head and neck, lung, ovarian and skin cancer. A total of 5 out of 40 tests (1 blood cancer, 1 brain cancer, 1 colorectal cancer and 2 lung cancer) revealed an association between IL-34 gene expression and cancer prognosis. It was found that the association between the expression of IL-34 and cancer prognosis varied in different types of cancer, even in the same types of cancer from different databases. This suggests that the function of IL-34 in these tumors may be multidimensional. The upstream transcription factor 1 (USF1), regulatory factor X-1 (RFX1), the Sp1 transcription factor 1 , POU class 3 homeobox 2 (POU3F2) and forkhead box L1 (FOXL1) regulatory transcription factor binding sites were identified in the IL-34 gene upstream (promoter) region, which may be involved in the effects of IL-34 in tumors.


Subject(s)
Cytokines/genetics , Genomics , Neoplasms/diagnosis , Neoplasms/genetics , Alternative Splicing , Amino Acid Sequence , Animals , Cytokines/chemistry , Cytokines/metabolism , Databases, Genetic , Exons , Gene Expression Profiling , Gene Order , Genomics/methods , Humans , Interleukins/chemistry , Interleukins/genetics , Interleukins/metabolism , Introns , Meta-Analysis as Topic , Molecular Sequence Data , Mutation , Neoplasms/metabolism , Phylogeny , Polymorphism, Single Nucleotide , Prognosis , Proteomics/methods , Sequence Alignment , Vertebrates
16.
BMC Cancer ; 14: 322, 2014 May 07.
Article in English | MEDLINE | ID: mdl-24884756

ABSTRACT

BACKGROUND: RNA binding proteins (RBPs) play a fundamental role in posttranscriptional control of gene expression. Different RBPs have oncogenic or tumor-suppressive functions on human cancers. RNPC1 belongs to the RNA recognition motif (RRM) family of RBPs, which could regulate expression of diverse targets by mRNA stability in human cancer cells. Several studies reported that RNPC1 played an important role in cancer, mostly acting as an oncogene or up-regulating in tumors. However, its role in human breast cancer remains unclear. METHODS: In the present study, we investigated the functional and mechanistic roles of RNPC1 in attenuating invasive signal including reverse epithelial-mesenchymal transition (EMT) to inhibit breast cancer cells aggressiveness in vitro. Moreover, RNPC1 suppress tumorigenicity in vivo. Further, we studied the expression of RNPC1 in breast cancer tissue and adjacent normal breast tissue by quantitative RT-PCR (qRT-PCR) and Western blot. RESULTS: We observed that RNPC1 expression was silenced in breast cancer cell lines compared to breast epithelial cells. More important, RNPC1 was frequently silenced in breast cancer tissue compared to adjacent normal breast tissue. Low RNPC1 mRNA expression was associated with higher clinical stages and mutp53, while low level of RNPC1 protein was associated with higher lymph node metastasis, mutp53 and lower progesterone receptor (PR). Functional assays showed ectopic expression of RNPC1 could inhibit breast tumor cell proliferation in vivo and in vitro through inducing cell cycle arrest, and further suppress tumor cell migration and invasion partly through repressing mutant p53 (mutp53) induced EMT. CONCLUSIONS: Overall, our findings indicated that RNPC1 had a potential function to play a tumor-suppressor role which may be a potential marker in the therapeutic and prognostic of breast cancer.


Subject(s)
Breast Neoplasms/metabolism , RNA-Binding Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Adult , Aged , Aged, 80 and over , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Cycle Checkpoints , Cell Movement , Cell Proliferation , Epithelial-Mesenchymal Transition , Female , Gene Expression Regulation, Neoplastic , Humans , Lymphatic Metastasis , MCF-7 Cells , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Mutation , Neoplasm Invasiveness , RNA Interference , RNA-Binding Proteins/genetics , Receptors, Progesterone/metabolism , Time Factors , Transfection , Tumor Burden , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics
17.
Mol Med Rep ; 10(3): 1461-8, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24938427

ABSTRACT

Secreted protein acidic and rich in cysteine (SPARC), also termed osteonectin or basement­membrane­40 (BM­40), is a matrix­associated protein that elicits changes in cell shape, inhibits cell­cycle progression and affects the synthesis of extracellular matrix (ECM). The final mature SPARC protein has 286 amino acids with three distinct domains, including an NH2­terminal acidic domain (NT), follistatin­like domain (FS) and C terminus domain (EC). The present study identified SPARC genes from 14 vertebrate genomes and revealed that SPARC existed in all types of vertebrates, including fish, amphibians, birds and mammals. In total, 21 single nucleotide polymorphisms (SNPs) causing missense mutations were identified, which may affect the formation of the truncated form of the SPARC protein. The human SPARC gene was found to be expressed in numerous tissues or organs, including in the bone marrow, whole blood, lymph node, thymus, brain, cerebellum, retina, heart, smooth muscle, skeletal muscle, spinal cord, intestine, colon, adipocyte, kidney, liver, pancreas, thyroid, salivary gland, skin, ovary, uterus, placenta, cervix and prostate. When searched in the PrognoScan database, the human SPARC gene was also found to be expressed in bladder, blood, breast, glioma, esophagus, colorectal, head and neck, ovarian, lung and skin cancer tissues. It was revealed that the association between the expression of SPARC and prognosis varied in different types of cancer, and even in the same cancer from different databases. It implied that the function of SPARC in these tumors may be multidimensional, functioning not just as a tumor suppressor or oncogene.


Subject(s)
Gene Expression Regulation, Neoplastic , Genomics/methods , Neoplasms/genetics , Osteonectin/genetics , Amphibians , Animals , Birds , Fishes , Gene Expression Profiling , Genome , Humans , Mammals , Meta-Analysis as Topic , Osteonectin/metabolism , Polymorphism, Single Nucleotide , Prognosis , Proteomics/methods
18.
Int J Oncol ; 45(2): 757-63, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24842044

ABSTRACT

Natural plant products occupy a very important position in the area of cancer chemotherapy. Many triterpenoid saponins have been proved as potential agents for chemoprevention and therapy of breast cancer. α-hederin, a monodesmosidic triterpenoid saponin distributed in Hedera or Nigella species, displays many biological activities. It is increasingly investigated for its promising anticancer potential since it has been shown to have cytotoxicity against several types of cancer cells. However, studies of α-hederin on breast cancer are limited, most of which focus on biological activity, while the mechanisms have not been widely reported yet. Previously, we purified and identified α-hederin from Clematis ganpiniana, a herb used in traditional Chinese medicine with antitumor action. In the present study, α-hederin showed strong inhibitory activity on the growth of breast cancer cells and induced apoptosis in these cells. α-hederin induced depolarization of mitochondrial membrane potential which released Apaf-1 and cytochrome c from the intermembrane space into the cytosol, where they promoted caspase-3 and caspase-9 activation. This is the first report on the growth inhibition and pro-apoptotic effects of α-hederin on breast cancer cells and the relative apoptosis pathways. It implied that triterpenoid saponin α-hederin could be a promising candidate for chemotherapy of breast cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/metabolism , Oleanolic Acid/analogs & derivatives , Phytotherapy/methods , Saponins/pharmacology , Apoptosis/drug effects , Blotting, Western , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Clematis , Flow Cytometry , Humans , Membrane Potential, Mitochondrial/drug effects , Oleanolic Acid/pharmacology , Plant Extracts/pharmacology
19.
PLoS One ; 9(6): e90848, 2014.
Article in English | MEDLINE | ID: mdl-24603880

ABSTRACT

There is growing interest in development of natural products as anti-cancer and chemopreventive agents. Many triterpenoids have been proved as potential agents for chemoprevention and therapy of breast cancer. Ginsenosides from ginseng, which mostly belong to dammarane-type triterpenoids, have gained great attention for their anti-breast cancer activity with diverse mechanisms. However, studies of other kinds of triterpenoid saponins on breast cancer are limited. Previously, we purified and identified a novel oleanane-type triterpene saponin named D Rhamnose ß-hederin (DRß-H) from Clematis ganpiniana, a Chinese traditional anti-tumor herb. In the present study, DRß-H showed strong inhibitory activity on the growth of various breast cancer cells and induced apoptosis in these cells. DRß-H inhibited PI3K/AKT and activated ERK signaling pathway. PI3K inhibitor LY294002 synergistically enhanced DRß-H-induced apoptosis whereas MEK inhibitor U0126 reduced the apoptosis rate. Moreover, DRß-H regulated the ratio of pro-apoptotic and anti-apoptotic Bcl-2 family proteins. Furthermore, DRß-H induced depolarization of mitochondrial membrane potential which released Apaf-1 and Cytochrome C from the inter membrane space into the cytosol, where they promoted caspase-9 and caspase-3 activation. This is the first report on the pro-apoptotic effects of DRß-H, a novel oleanane-type triterpenoid saponin, on breast cancer cells and its comprehensive apoptosis pathways. It implied that oleanane-type triterpenoid saponin DRß-H could be a promising candidate for chemotherapy of breast cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Oleanolic Acid/analogs & derivatives , Saponins/pharmacology , Apoptotic Protease-Activating Factor 1/metabolism , Breast Neoplasms , Caspase 3/metabolism , Caspase 9/metabolism , Chromones/pharmacology , Cytochromes c/metabolism , Drug Screening Assays, Antitumor , Drug Synergism , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , MAP Kinase Signaling System , MCF-7 Cells , Membrane Potential, Mitochondrial/drug effects , Morpholines/pharmacology , Oleanolic Acid/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism
20.
Pak J Pharm Sci ; 26(2): 233-7, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23455189

ABSTRACT

The extracts prepared from Liriodendron tulipifera Linn., L. chinense (Hemsl.) Sarg., and their hybrid L. chinense x L. tulipifera, were investigated for their cytotoxic abilities in vitro against five human cancer cell lines: MDA-MB-231 and MCF-7 breast cancer cells, SGC-7901 gastric cancer cells, HuH-7 hepatocarcinoma cells, and HCT-15 colon carcinoma cells, and then measured their phenols and alkaloids contents. Of these plant extracts, some of them, especially the lower polar extracts from barks, exhibited potent cytotoxic effects on five tested tumor cell lines.


Subject(s)
Alkaloids/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Liriodendron/chemistry , Neoplasms/pathology , Phenols/pharmacology , Plant Extracts/pharmacology , Alkaloids/analysis , Alkaloids/isolation & purification , Antineoplastic Agents, Phytogenic/analysis , Antineoplastic Agents, Phytogenic/isolation & purification , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , Inhibitory Concentration 50 , MCF-7 Cells , Phenols/analysis , Phenols/isolation & purification , Plant Bark , Plant Extracts/analysis , Plant Extracts/isolation & purification , Plant Leaves , Plants, Medicinal
SELECTION OF CITATIONS
SEARCH DETAIL
...