Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Cell Mol Biol (Noisy-le-grand) ; 70(2): 67-72, 2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38430042

ABSTRACT

To investigate the expression pattern and prognostic role of m6A RNA methylation regulators in non-small cell lung cancer (NSCLC), we downloaded data from 422 patients from The Cancer Genome Atlas (TCGA) database. The relationship between the expression levels of m6A RNA methylation regulators and clinicopathological variables of NSCLC was analysed using R language. By analysing glioma data in TCGA, we found that a prognostic risk score model could be constructed based on 18 genes with m6A methylation modification. m6A gene alterations were significantly associated with tumour grade and tumour stage. Least Absolute Shrinkage and Selection Operator (LASSO) Cox regression models were used to identify 2 m6A RNA methylation modifiers: IFG2BP2, and METTL14 to construct risk profiles. Based on the risk profile, patients were divided into high-risk and low-risk groups. The overall survival rate of the low-risk group was significantly higher than that of the high-risk group. The results suggest that the prognostic risk score model constructed by m6A methylation regulators can predict the prognosis of glioma patients. IFG2BP2 and METTL14 may be the key m6A methylation regulators involved in the development of NSCLC and can be used as the molecular markers for the prognosis of NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Glioma , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/genetics , RNA Methylation , Prognosis , Lung Neoplasms/diagnosis , Lung Neoplasms/genetics , Glioma/diagnosis , Glioma/genetics , RNA
2.
Oncol Rep ; 45(6)2021 Jun.
Article in English | MEDLINE | ID: mdl-33907845

ABSTRACT

After the publication of the article, and also the publication of a Corrigendum (see doi: 10.3892/or.2020.7744), there are further errors in the published paper that the authors wish to correct in a subsequent corrigendum. In the printed version of Fig. 5, the "NC" images were mistakenly presented in the data panels showing the results of the TCA8113 and TSCCA invasion assay experiments. Furthermore, in Fig. 4A and 6A, the ß­actin control bands were erroneously selected for these figures. The corrected versions of Figs. 4, 5 and 6 are shown opposite and on the next page, incorporating the correct data for Figs. 4A, 5 and 6A. These further corrections do not affect the results and conclusions of this work. The authors all agree to this Corrigendum, and are grateful to the Editor of Oncology Reports for allowing them to have the opportunity to correct these additional errors. Lastly, the authors apologize to the readership for any inconvenience these errors may have caused. [the original article was published in Oncology Reports 39: 1853­1859, 2018; DOI: 10.3892/or.2018.6231].

3.
Oncol Rep ; 44(5): 2319, 2020 11.
Article in English | MEDLINE | ID: mdl-33106884

ABSTRACT

During the preparation of the above article, the authors inadvertently selected an incorrect pair of data panels for Fig. 5B. Essentially, in the published version of Fig. 5B, the images were presented incorrectly for the NC migration and KD#3 migration and invasion data panels associated with the TSCCA cell Transwell assay experiments. Furthermore, in Fig. 7A, the GAPDH control bands were erroneously selected for the figure. Figs. 5 and 7 as they should have appeared in the Journal are shown opposite, incorporating the correct data for Figs. 5B and 7A. These errors did not affect either the results or the conclusions of this work. The authors all agree to this Corrigendum, and thank the Editor of Oncology Reports for allowing them to have the opportunity to present their corrected data. Furthermore, the authors apologize to the readership for any inconvenience these errors may have caused. [the original article was published in Oncology Reports 39: 1853­1859, 2018; DOI: 10.3892/or.2018.6231].

4.
Mol Immunol ; 124: 100-108, 2020 08.
Article in English | MEDLINE | ID: mdl-32554100

ABSTRACT

Programmed cell death protein 1 (PD1) is a cell-surface receptor that plays a vital regulatory role in suppressing inflammatory T cell activity; therefore, it is an ideal target for T cell-redirecting therapies. Here, we describe a cynomolgus macaque model for studying the transfer of PD1-modified T cells. We developed the first T cell engager targeting the disruption of PD1 by electroporation of plasmids encoding sgRNA and Cas9. There were no significant differences between mock T cells and PD1-knockout (PD1-KO) T cells in terms of cell viability, T cell signature marker expression, cell apoptosis, or cell cycling during prolonged in vitro culture. However, in a mixed lymphocyte reaction, PD1-KO T cells exhibited increased proliferation for both CD4+ and CD8+T cells and enhanced IFNγ release. We adoptively transferred autologous PD1-KO T cells into three cynomolgus monkeys. The PD1-KO T cells did not cause overt toxicity as measured by evaluating body weight, hematological parameters, and blood chemistry parameters. Histopathological analyses of tissues showed no lesions related to the infused PD1-KO T cells. Our findings demonstrate the utility of cynomolgus monkeys in expanding PD1-KO T cells and evaluating the safety of this immunotherapy and provide a new strategy for T cell-based adoptive cell therapies.


Subject(s)
Programmed Cell Death 1 Receptor/deficiency , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes/immunology , Adoptive Transfer , Animals , Gene Knockout Techniques/methods , Macaca fascicularis , T-Lymphocytes/transplantation
5.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-822989

ABSTRACT

@#[Abstract] Objective: To investigate the effects of CRISPR/Cas9 gene editing mediated PD-1 knockdown on the proliferation, phenotype, IFN-γ and IL-2 secretion of T cells in Cynomolgus monkeys. Methods: gRNA targeting PD-1 gene of Cynomolgus monkey was designed, and the corresponding plasmid was constructed and extracted. peripheral blood mononuclear cells (PBMCs) of Cynomolgus monkeys were isolated, and plasmid DNAs were added for transfection by using Lonza 4D electrorotometer. FACS analysis and fluorescence microscopy were used to detect transfection efficiency at 48 h after transfection. Genomic DNA of T cells was extracted for PCR amplification and T7E1 digestion identification. The proliferation of T cells was induced under the stimulation of human CD3 antibody and IL-2, and the cell growth curve was drawn. PI staining flow cytometry was used to detect cell cycle and the expression levels of CD4 and CD8, and ELISA was used to detect the secretion of IFN- γ and IL-2. Results: At 48 h after transfection, the cells with green fluorescent protein expression in experimental group were observed under fluorescence microscopy with a transfection efficiency of (21.6±3.2)%. T7E1 enzyme digestion results showed that the PCR product of genomic DNA of cells in experimental group showed 3 bands after digestion, including the target cleavage bands(243,197 bp). Compared with non-transfected cells, the cells in experimental group exhibited slow proliferation, delayed colony formation, with small volume and weak refraction; the number of T cells at G0/G1 phase of the experimental group was significantly increased (P<0.05), while the number of cells at G2/M phase was significantly reduced (P<0.05); and the secretion levels of IFN-γ and IL-2 in the cells of the experimental group increased significantly (both P<0.05). However, the difference in the expression levels of CD4 and CD8 was not statistically significant between the two groups (both P>0.05). Conclusion: PD-1 gene knockout can arrest T cells in Cynomolgus monkey at G0/G1 phase, thereby inhibiting its proliferation and increasing the secretion of IFN-γ and IL-2 in the meanwhile.

6.
Hum Immunol ; 80(10): 863-870, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31262519

ABSTRACT

Regulatory B (Breg) cells are a special subset of immunoregulatory cells with unique phenotypes and functions. In this study, human CD19+CD25high Breg cells were purified from human peripheral blood. Based on the coculture system of Breg cells and CD4+ T cells in vitro, Breg cells were found to promote the increase in regulatory T (Treg) cells while decreasing the number of Th17 cells. Breg cells regulate Treg cells through two processes: cell-cell contact and cytokines. TGF-ßsRII, a blocker of transforming growth factor-ß (TGF-ß), can attenuate the effects of Treg elevation, suggesting that TGF-ß is the main cytokine, while Breg cells rather than interleukin-10 (IL-10) regulate the differentiation of Treg cells. However, Th17 cells were mainly regulated by cytokines, without an obvious regulatory effect on cell-cell contacts. Breg cells may regulate Th17 cells by a pathway independent of TGF-ß and IL-6. The coculture of Breg cells and CD4+ T cells led to changes in the cytokine spectrum, which included significant increases in IL-4, IL-6 and IL-10 but not obvious changes in IL-2, IFN-γ and TNF. The inhibitory effect of Breg cells was weakened by blocking cell-cell contacts in cultures separated with the Transwell chamber because IL-10 decreased while IL-6 increased when compared with cocultured Breg and CD4+ T cells. When the IL-10 inhibitor IL-10sRα was added, IL-6 and TNF levels significantly increased, while treatment with the TGF-ß inhibitor TGF-ßsRII did not result in similar changes, suggesting that IL-10 is an important molecule to inhibit the proinflammatory factors IL-6 and TNF in this culture system.


Subject(s)
Antigens, CD19/metabolism , B-Lymphocytes, Regulatory/immunology , Forkhead Transcription Factors/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , T-Lymphocytes, Regulatory/immunology , Th1-Th2 Balance , Th17 Cells/immunology , Adult , Cell Differentiation/immunology , Cell Proliferation , Coculture Techniques/methods , Cytokines/metabolism , Female , Healthy Volunteers , Humans , Male
7.
In Vitro Cell Dev Biol Anim ; 54(8): 549-554, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30112696

ABSTRACT

T cells have been successfully applied to cancer immunotherapy. However, a challenge is an expansion of T cells from cynomolgus monkey, which is a valuable non-human primate model for T cell therapy transferring to the clinic. Here, we compared several strategies to expand cynomolgus monkey T cell and developed an appropriate method. Our study demonstrated that 100 ng/ml CD3 mAb + 1% PHA+ 1000 U/ml IL2 therapy significantly expanded peripheral blood CD3+ T cells without compromising T cell phenotype in vitro. The results of this study could be used for T cell remodeling, which has significant therapeutic potential in Chimeric Antigen Receptor-T (CAR-T) cell immunotherapy.


Subject(s)
Cell Culture Techniques/methods , T-Lymphocytes/cytology , Animals , Cell Proliferation , Humans , K562 Cells , Macaca fascicularis , Male , Phenotype
8.
Oncol Lett ; 15(3): 2771-2780, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29435003

ABSTRACT

The majority of tumors possess the features of hypoxia. It is generally accepted that hypoxia is a negative prognostic factor for cancer. Low levels of oxygen are able to modify basic cell metabolism status. Elucidating the basic response, including cell proliferation and migration, to hypoxia by cancer cells is important for understanding the role of hypoxia in the development of cancer. In the present study, CoCl2 stimulation was used to simulate hypoxia. A microRNA (miRNA/miR) array was used to systematically detect the changes in miRNA expression profiles. Following treatment with CoCl2 for 12 h, 15 miRNAs were markedly upregulated and 10 miRNAs were markedly decreased compared with the control. After 24 h CoCl2 incubation, 15 miRNAs were increased and 3 miRNAs were decreased compared with the control. Among them, 7 miRNAs were upregulated and 2 miRNAs were downregulated at 12 and 24 h following CoCl2 stimulation. The potential roles of these miRNA were reviewed and it was identified that the majority of them are associated with cell proliferation and migration. Additional experiments demonstrated that CoCl2 incubation inhibited the proliferation of MCF-7 cells but promoted cell migration. miR-491 may be a key miRNA for hypoxia-inhibited cell proliferation, as it was identified that hypoxia induced the downregulation of B-cell lymphoma-extra large in a miR-491-dependent manner. As the target of miR-302a, CXCR4 may be a key protein for hypoxia-promoted cell migration. In the present study, it was identified that in the early stage of hypoxia, cell proliferation was inhibited but cell migration was promoted. These results support the hypothesis that hypoxia may be a driving force for tumor cell escape from the primary tumor site to other organs, or other sites of the same organ.

9.
Oncol Rep ; 39(4): 1853-1859, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29393475

ABSTRACT

The primary reasons for the treatment failure of patients with oral tongue squamous cell carcinoma (OTSCC) are metastasis and tumor recurrence. Identifying the exact mechanisms underlying metastasis is a key point in improving patient prognosis. It has been reported that a hypoxic microenvironment plays an important role during the metastasis of malignancies. We found that the expression of fibronectin type III domain containing 3B (FNDC3B) is positively correlated with lymph node metastasis and advanced cTNM stage of OTSCC by IHC assay and correlation analysis. Furthermore, we found that knockdown of FNDC3B could suppress the migratory and invasive abilities of OTSCC cells. In addition, treating OTSCC cells with CoCl2 (a hypoxia mimetic agent) upregulated the mRNA and protein expression of FNDC3B via HIF-1α. Moreover, the resultant increase in FNDC3B expression significantly induced epithelial-mesenchymal transition (EMT) in OTSCC cells. The present study elucidated the important role played by FNDC3B in OTSCC metastasis and indicates FNDC3B as a potential target for the treatment of OTSCC metastasis. However, many questions remain to be explored.


Subject(s)
Carcinoma, Squamous Cell/genetics , Fibronectins/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Tongue Neoplasms/genetics , Adult , Aged , Carcinoma, Squamous Cell/pathology , Cell Hypoxia/genetics , Cell Line, Tumor , Cell Movement/genetics , Cobalt/pharmacology , Epithelial-Mesenchymal Transition/genetics , Female , Gene Knockdown Techniques , Humans , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Staging , Prognosis , Tongue Neoplasms/pathology , Tumor Microenvironment/genetics
10.
Oncol Res ; 26(8): 1245-1255, 2018 Sep 14.
Article in English | MEDLINE | ID: mdl-28550688

ABSTRACT

Long noncoding RNAs (lncRNAs), a new class of functional regulators involved in human tumorigenesis, have been attracting the increasing attention of researchers. The lncRNA colorectal neoplasia differentially expressed (CRNDE) gene, transcribed from chromosome 16 on the strand opposite the adjacent IRX5 gene, was originally found to be increased in CRC and was reported to be abnormally expressed in many cancers. However, its potential role and the molecular mechanism underlying the radioresistant phenotype formation of lung adenocarcinoma (LAD) remain unclear. In our present study, we identified that CRNDE was significantly upregulated in LAD tissue and radioresistant LAD cell lines. A high level of CRNDE expression was significantly correlated with poor differentiation, TNM stage, lymph node metastasis, radiotherapy response, and a significantly shorter overall survival. Gain- and loss-of-function tests revealed that CRNDE could influence the radiosensitivity of LAD cells by affecting the G1/S transition and causing apoptosis of LAD cells in vitro. Additionally, the mechanistic investigations showed that CRNDE could interact with PRC2 and recruit its core component EZH2 to p21 (CDKN1A) promoter regions and repress its transcription. Furthermore, rescue experiments were performed to confirm that CRNDE oncogenic function was partly through regulating p21. In conclusion, our data suggest that CRNDE may function as an oncogene by modulating p21, finally contributing to the radioresistant phenotype formation of LAD cells.


Subject(s)
Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/radiotherapy , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , RNA, Long Noncoding/metabolism , A549 Cells , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Cell Proliferation/radiation effects , Cyclin-Dependent Kinase Inhibitor p21/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Humans , Promoter Regions, Genetic , RNA, Long Noncoding/biosynthesis , RNA, Long Noncoding/genetics , Radiation Tolerance , Signal Transduction , Up-Regulation
11.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-821288

ABSTRACT

@#[Abstract] Objective: To establish a method for in vitro isolation and culture of T lymphocytes from peripheral blood of cynomolgus monkeys that induced by human CD3 antibody based on the foundation of protein homology of CD3 from human, cynomolgus monkey and porcine. Methods: The amino acid sequences of human, cynomolgus monkeys and porcine CD3 proteins were obtained from NCBI, and the sequence, homology and phylogenetic tree were analyzed by DNAMAN software. Western blotting was used to detect the expression of CD3 protein on T cell membranes from the three species. PBMCs of healthy cynomolgus were isolated and divided into three groups: group A was stimulated with anti-human CD3Ab alone, group B was stimulated with IL-2 alone, and group C was costimulated with human CD3Ab and IL-2. Cell morphology and growth status were observed under inverted microscope and the cell growth curve was plotted. Cell viability was detected by trypan blue staining and the expressions of CD3, CD4 and CD8 on T cell surface were detected by flow cytometry. Results: The homology of the amino acid sequence of human CD3 protein to cynomolgus monkey and porcine were 86.9% and 65.6% respectively. The expression levels of CD3 protein on cynomolgus and porcine T cell membrane were 79% and 17% contrast to human, respectively. Cells of group A did not proliferate. Proliferation, viability and CD3 expression [(93.8±3.6)% vs (70.3±4.7)%, P<0.01] in T cells of group C were significantly higher than those in group B. Growth curve of T cells in group C showed an S-shape, which is consistent with Logistic growth curve. T cells in group C exhibited high purity and expressed high level CD3; moreover, the CD8+T cell took a high proportion. Conclusion: The membrane of T lymphocytes from peripheral blood of cynomolgus can express CD3 protein that highly homological to human. Co-stimulation of human CD3Ab, IL-2 and 1% PHAcan induce the proliferation and differentiation of T lymphocytes of cynomolgus, and obtain T lymphocytes with good growth status, high proliferation ability and high purity.

12.
Cell Physiol Biochem ; 42(1): 211-221, 2017.
Article in English | MEDLINE | ID: mdl-28535539

ABSTRACT

BACKGROUND/AIMS: This study aimed to investigate the potential roles of miR-424 expression in non-small cell lung cancer (NSCLC) metastasis and growth and its underlying mechanism. METHODS: The expression of miR-424 in two NSCLC cell lines (A549 and H1975) was altered by transfection with miR-424 mimic and inhibitor. Effects of miR-424 overexpression and suppression on cells migration, invasion and colony formation were analyzed. Target genes for miR-424 were predicted using bioinformatics method and then verified using luciferase assay. Effects of miR-424 expression on cell migration, invasion and proliferation were reanalyzed on the condition of TNFAIP1 was silenced. Moreover, TNFAIP1 silencing and miR-424 modified A549 cells were subcutaneous injected into node BALB/c mice to confirm the regulation of miR-424 on TNFAIP1 in regulating tumor growth. RESULTS: Compared with the control, miR-424 overexpression significantly increased the migrated and invaded cells, as well as the proliferated colonies. TNFAIP1 was a predicted target gene for miR-424, and was negatively regulated by miR-424. TNFAIP1 silence significantly increased the migrated and invaded cells compared to that in control, while these increases were abolished by miR-424 suppression. Animal experiment further evidenced miR-424 affected tumor growth by regulating TNFAIP1. CONCLUSIONS: These data demonstrate that miR-424 may be a contributor for NSCLC progression and metastasis through involving in cell migration, invasion and proliferation via inhibiting TNFAIP1. This study may provide theoretical basis for miR-424 in NSCLC target therapeutic treatment.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , MicroRNAs/metabolism , Proteins/metabolism , 3' Untranslated Regions , A549 Cells , Adaptor Proteins, Signal Transducing , Animals , Antagomirs/metabolism , Base Sequence , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Progression , Humans , Lung Neoplasms/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Proteins/antagonists & inhibitors , Proteins/genetics , RNA Interference , RNA, Small Interfering/metabolism , Sequence Alignment , Transplantation, Heterologous
13.
Onco Targets Ther ; 9: 5737-5750, 2016.
Article in English | MEDLINE | ID: mdl-27729799

ABSTRACT

BACKGROUND: Semaphorin 4D (Sema4D) is highly expressed in certain types of tumors and functions in the regulation of tumor angiogenesis and growth. However, it is still not clear regarding the roles of Sema4D in breast cancer. This study was designed to explore the effects of Sema4D on proliferation, cell cycle progression, apoptosis, invasion, migration, tumor growth, and angiogenesis in breast cancer. MATERIALS AND METHODS: The expression level of Sema4D was investigated in MCF10A, 184A1, HCC1937, MDA-MB-468, MDA-MB-231, Hs578T, BT474, MCF-7, and T47D breast cancer cell lines by Western blotting analysis. Sema4D downregulation or overexpression was established by infection with lentiviruses-encoding Sema4D short hairpin RNA (shRNA) or Sema4D. To evaluate the effects of Sema4D on cell proliferation, cell cycle progression, apoptosis, invasion, and migration of MDA-MB-231 and MDA-MB-468 cells, methods including MTT assay, flow cytometry, wound healing assay, and transwell experiments were applied. BALB/c nude mice were injected with MDA-MB-231 cells, which were respectively infected with lentiviruses-encoding Sema4D, Sema4D shRNA, and GFP, followed by tumor angiogenesis assay. RESULTS: Sema4D was expressed at higher levels in breast cancer cell lines compared with the normal human breast epithelial cell lines, especially in MDA-MB-231 and MDA-MB-468 cells. Cell proliferation ability was remarkably inhibited in Sema4D downregulated condition, whereas the proportions of cells in the G0/G1 phase and apoptosis increased in MDA-MB-231 and MDA-MB-468 cells. In addition, the invasion and migration abilities of these cells were obviously reduced. Xenograft growth as well as angiogenesis was inhibited when infected with lentiviruses-encoding Sema4D shRNA in vivo. CONCLUSION: Downregulation of Sema4D had notable influence on cell proliferation ability, invasion, migration, and apoptosis of both MDA-MB-231 and MDA-MB-468 cells. Furthermore, infection with lentiviruses-encoding Sema4D shRNA obviously inhibited tumor growth and angiogenesis in BALB/c nude mice. Our results showed that Sema4D may represent a novel therapeutic target for human breast cancer.

14.
Arch Gynecol Obstet ; 294(5): 1047-1053, 2016 11.
Article in English | MEDLINE | ID: mdl-27365106

ABSTRACT

PURPOSE: We aimed to screen possible biomarkers associated with the molecular mechanism of breast cancer using microRNA (miRNA) microarray. METHODS: The miRNAs expression profile GSE45666 was downloaded from Gene Expression Omnibus database, which included 101 genechips from breast tumor samples and 15 from adjacent breast normal tissue samples. Limma package in R language was used to screen and identify differentially expressed miRNAs (DE-miRNAs) which were classified as up-regulated and down-regulated groups. Then, target genes regulated by the two groups of DE-miRNA were predicted, followed by the functional and pathway enrichment analysis using the DAVID system. RESULTS: Totally, 130 DE-miRNAs were screened out, including 59 up-regulated DE-miRNAs and 71 down-regulated DE-miRNAs. The functional enrichment indicated that target genes of up- and down-regulated DE-miRNA may be most highly associated with positive regulation of gene expression and regulation of cellular metabolic process, respectively. Target genes regulated by the up- and down-regulated DE-miRNAs were mainly enriched in 13 and 14 pathways, respectively, and both were most significant in subcategories in cancer. In addition, we identified three important miRNAs (miR-142-3p, miR-483-5p and miR-483-3p) pivotal for the initiation and progression of this malignant tumor. CONCLUSIONS: MiR-142-3p, miR-483-5p and miR-483-3p are potential key factors for further understanding the molecular mechanism of breast cancer by affecting the normal physiological function of cell.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/genetics , MicroRNAs/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Tissue Array Analysis
15.
Int J Oncol ; 47(4): 1371-8, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26693549

ABSTRACT

Gene therapy is one of the most important aspects of molecular targeted therapeutic approaches for tumors. A small molecule targeting carrier plays an important role in this process. PI, a new peptide found in our phage library, has been specifically suggested, combined with the human triple-negative breast cancer cell line MDA-MB­231, and may be developed as a targeting/individualization therapy strategy to be applied in breast cancer research. In this study, we further investigated whether this peptide could carry exogenous protein to the target cells by forming a fusion peptide. PI-GST and PI-TK were cloned into plasmids and used for expression studies, analyses of PI-mediated protein delivery efficiency, and to investigations into the effect of PI on thymidine kinase/ganciclovir-mediated cytotoxicity. Biodistribution profiles were also investigated in vivo. The results showed the PI fusion protein was expressed correctly in vitro, and could carry GST into the target cells. Under certain conditions, PI-TK sensitizes cells to ganciclovir more efficiently than TK. In vivo there was a trend for increased inhibition of tumor growth with PI-TK when ganciclovir was present. Therefore, our results suggest the potential of PI as a new specific target carrier in breast cancer therapy.


Subject(s)
Antineoplastic Agents/administration & dosage , Breast Neoplasms , Cell-Penetrating Peptides/administration & dosage , Genetic Therapy/methods , Recombinant Fusion Proteins/administration & dosage , Animals , Antineoplastic Agents/pharmacokinetics , Blotting, Western , Cell Line, Tumor , Cell Proliferation , Cell-Penetrating Peptides/pharmacokinetics , Fluorescent Antibody Technique , Ganciclovir/pharmacology , Genes, Transgenic, Suicide , Heterografts , Humans , Mice , Mice, Inbred BALB C , Organic Anion Transporters , Recombinant Fusion Proteins/pharmacokinetics , Thymidine Kinase/administration & dosage , Tomography, Emission-Computed, Single-Photon
SELECTION OF CITATIONS
SEARCH DETAIL
...