Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Front Immunol ; 14: 1275304, 2023.
Article in English | MEDLINE | ID: mdl-38022650

ABSTRACT

T cell engagers, a category of T cell-retargeting immunotherapy, are rapidly transforming clinical cancer care. However, the lack of tumor-specific targets poses a significant roadblock for broad adaptation of this therapeutic modality in many indications, often resulting in systemic on-target off-tumor toxicity. Though various tumor-derived intracellular mutations provide a massive pool of potential tumor-specific antigens, targeting them is extremely challenging, partly due to the low copy number of tumor associated antigen (TAA)-derived pMHC on tumor cell surface. Further, the interplay of binding geometry and format valency in relation to the capacity of a T cell engager to efficiently target low density cell-surface pMHC is not well understood. Using the Wilms' tumor 1 (WT1) oncoprotein as a proof-of-principle TAA, combined with an array of IgG-like T cell engager modalities that differ in their anti-TAA valency and binding geometry, we show that the ability to induce an immunological synapse formation, resulting in potent killing of WT1 positive cancer cell lines is primarily dependent on the distinct geometrical conformations between the Fab arms of anti-WT1-HLA-A*02:01 and anti-CD3. The augmented avidity conferred by the binding of two anti-WT1-HLA-A*02:01 Fab arms has only minimal influence on cell killing potency. These findings demonstrate the need for careful examination of key design parameters for the development of next-generation T cell engagers targeting low density TAA-pMHCs on tumor cells.


Subject(s)
Neoplasms , T-Lymphocytes , Humans , WT1 Proteins/genetics , Neoplasms/genetics , Neoplasms/therapy , Antigens, Neoplasm , Immunoproteins , HLA-A Antigens , Peptides
2.
Cancer Cell ; 40(5): 479-493.e6, 2022 05 09.
Article in English | MEDLINE | ID: mdl-35452604

ABSTRACT

A common theme across multiple successful immunotherapies for cancer is the recognition of tumor-specific mutations (neoantigens) by T cells. The rapid discovery of such antigen responses could lead to improved therapies through the adoptive transfer of T cells engineered to express neoantigen-reactive T cell receptors (TCRs). Here, through CITE-seq (cellular indexing of transcriptomes and epitopes by sequencing) and TCR-seq of non-small cell lung cancer (NSCLC) tumor-infiltrating lymphocytes (TILs), we develop a neoantigen-reactive T cell signature based on clonotype frequency and CD39 protein and CXCL13 mRNA expression. Screening of TCRs selected by the signature allows us to identify neoantigen-reactive TCRs with a success rate of 45% for CD8+ and 66% for CD4+ T cells. Because of the small number of samples analyzed (4 patients), generalizability remains to be tested. However, this approach can enable the quick identification of neoantigen-reactive TCRs and expedite the engineering of personalized neoantigen-reactive T cells for therapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Antigens, Neoplasm , CD8-Positive T-Lymphocytes , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lymphocytes, Tumor-Infiltrating , Receptors, Antigen, T-Cell , T-Lymphocytes
3.
Mol Cancer ; 17(1): 11, 2018 01 19.
Article in English | MEDLINE | ID: mdl-29351796

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is a severe health problem worldwide. Clarifying the mechanisms for the deregulation of oncogenes and tumour suppressors in CRC is vital for its diagnosis, treatment, prognosis and prevention. Hu antigen R (HuR), which is highly upregulated in CRC, functions as a pivotal oncogene to promote CRC progression. However, the underlying cause of its dysregulation is poorly understood. METHODS: In CRC tissue sample pairs, HuR protein levels were measured by Western blot and immunohistochemical (IHC) staining, respectively. HuR mRNA levels were also monitored by qRT-PCR. Combining meta-analysis and microRNA (miRNA) target prediction software, we predicted miRNAs that targeted HuR. Pull-down assay, Western blot and luciferase assay were utilized to demonstrate the direct binding of miR-22 on HuR's 3'-UTR. The biological effects of HuR and miR-22 were investigated both in vitro by CCK-8, EdU and Transwell assays and in vivo by a xenograft mice model. JASPAR and SABiosciences were used to predict transcriptional factors that could affect miR-22. Luciferase assay was used to explore the validity of putative Jun binding sites for miR-22 regulation. ChIP assay was performed to test the Jun's occupancy on the C17orf91 promoter. RESULTS: We observed a significant upregulation of HuR in CRC tissue pairs and confirmed the oncogenic function of HuR both in vitro and in vivo. We found that an important tumour-suppressive miRNA, miR-22, was significantly downregulated in CRC tissues and inversely correlated with HuR in both CRC tissues and CRC cell lines. We demonstrated that miR-22 directly bound to the 3'-UTR of HuR and led to inhibition of HuR protein, which repressed CRC proliferation and migration in vitro and decelerated CRC xenografted tumour growth in vivo. Furthermore, we found that the onco-transcription factor Jun could inhibit the transcription of miR-22. CONCLUSIONS: Our findings highlight the critical roles of the Jun/miR-22/HuR regulatory axis in CRC progression and may provide attractive potential targets for CRC prevention and treatment.


Subject(s)
Cell Transformation, Neoplastic/genetics , Colorectal Neoplasms/genetics , ELAV-Like Protein 1/genetics , Gene Expression Regulation, Neoplastic , Genes, jun , MicroRNAs/genetics , 3' Untranslated Regions , Animals , Cell Line, Tumor , Cell Proliferation , Databases, Genetic , Disease Models, Animal , Genes, Reporter , Heterografts , Humans , Mice , Models, Biological , Oncogenes , RNA Interference , Transcription, Genetic
4.
Sci Rep ; 7(1): 16979, 2017 12 05.
Article in English | MEDLINE | ID: mdl-29208934

ABSTRACT

Salmonella can hijack host atypical miRNA processing machinery to cleave its small non-coding RNA into a ~22-nt RNA fragment, Sal-1, which facilitates Salmonella survival in the infected host. The mechanism through which Sal-1 promotes Salmonella survival, however, remains unknown. In the present study, we reported that Sal-1 targets cellular inducible nitric oxide synthase (iNOS) in a miRNA manner, leading to attenuation of host cell iNOS/NO-mediated anti-microbial capacity. First, depletion of Sal-1 in Salmonella-infected epithelial cells significantly increased the iNOS level but not the levels of various inflammatory cytokines. Bioinformatics analysis and mutagenesis strategies were consistent with the identification of mRNA of iNOS as a target of Sal-1 in both human and mice. Second, western blot and immunohistochemical analysis confirmed that Sal-1 suppressed iNOS expression in vitro and in vivo, thus reducing the production of NO. Finally, Sal-1 facilitating Salmonella survival through suppressing iNOS induction was confirmed in mouse model by expressing mutated iNOS that is not targeted by Sal-1 in mice colon. In conclusion, our study provides new insight into the pathogenic mechanism of intracellular bacteria to modulate host innate immune response.


Subject(s)
Host-Pathogen Interactions/genetics , MicroRNAs/metabolism , Nitric Oxide Synthase Type II/genetics , RNA, Bacterial/metabolism , Salmonella enteritidis/genetics , Animals , Female , HT29 Cells , Humans , Mice, Inbred BALB C , MicroRNAs/genetics , Mutation , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , RNA, Bacterial/genetics , Salmonella Infections/genetics , Salmonella Infections/microbiology , Salmonella enteritidis/pathogenicity
5.
PLoS One ; 12(9): e0184113, 2017.
Article in English | MEDLINE | ID: mdl-28910318

ABSTRACT

Cavities are important in clinical diagnosis of pulmonary tuberculosis (TB) infected by Mycobacterium tuberculosis. Although microRNAs (miRNAs) play a vital role in the regulation of inflammation, the relation between plasma miRNA and pulmonary tuberculosis with cavity remains unknown. In this study, plasma samples were derived from 89 cavitary pulmonary tuberculosis (CP-TB) patients, 89 non-cavitary pulmonary tuberculosis (NCP-TB) patients and 95 healthy controls. Groups were matched for age and gender. In the screening phase, Illumina high-throughput sequencing technology was employed to analyze miRNA profiles in plasma samples pooled from CP-TB patients, NCP-TB patients and healthy controls. During the training and verification phases, quantitative RT-PCR (qRT-PCR) was conducted to verify the differential expression of selected miRNAs among groups. Illumina high-throughput sequencing identified 29 differentially expressed plasma miRNAs in TB patients when compared to healthy controls. Furthermore, qRT-PCR analysis validated miR-769-5p, miR-320a and miR-22-3p as miRNAs that were differently present between TB patients and healthy controls. ROC curve analysis revealed that the potential of these 3 miRNAs to distinguish TB patients from healthy controls was high, with the area under the ROC curve (AUC) ranged from 0.692 to 0.970. Moreover, miR-320a levels were decreased in drug-resistant TB patients than pan-susceptible TB patients (AUC = 0.882). In conclusion, we identified miR-769-5p, miR-320a and miR-22-3p as potential blood-based biomarkers for TB. In addition, miR-320a may represent a biomarker for drug-resistant TB.


Subject(s)
MicroRNAs/blood , Tuberculosis, Multidrug-Resistant/blood , Tuberculosis, Pulmonary/blood , Adolescent , Adult , Biomarkers/blood , Female , Humans , Male , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction
6.
Sci Rep ; 7(1): 2392, 2017 05 24.
Article in English | MEDLINE | ID: mdl-28539638

ABSTRACT

Salmonella have developed a sophisticated machinery to evade immune clearance and promote survival in the infected cells. Previous studies were mostly focused on either bacteria itself or host cells, the interaction mechanism of host-pathogen awaits further exploration. In the present study, we show that Salmonella can exploit mammalian cell non-classical microRNA processing machinery to further process bacterial small non-coding RNAs into microRNA-like fragments. Sal-1, one such fragment with the highest copy number in the infected cells, is derived from Salmonella 5'-leader of the ribosomal RNA transcript and has a 'stem' structure-containing precursor. Processing of Sal-1 precursors to mature Sal-1 is dependent on host cell Argonaute 2 (AGO2) but not Dicer. Functionally, depleting cellular Sal-1 strongly renders the Salmonella bacteria less resistant to the host defenses both in vitro and in vivo. In conclusion, we demonstrate a novel strategy for Salmonella evading the host immune clearance, in which Salmonella produce microRNA-like functional RNA fragments to establish a microenvironment facilitating bacterial survival.


Subject(s)
Argonaute Proteins/genetics , MicroRNAs/genetics , RNA, Bacterial/genetics , Salmonella Infections/genetics , Salmonella enteritidis/genetics , Salmonella enteritidis/pathogenicity , Animals , Argonaute Proteins/metabolism , Base Sequence , Cytoplasm , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Gene Expression Regulation , HT29 Cells , HeLa Cells , Host-Pathogen Interactions , Humans , Mice , MicroRNAs/metabolism , Microbial Viability , RAW 264.7 Cells , RNA, Bacterial/metabolism , Ribonuclease III/genetics , Ribonuclease III/metabolism , Salmonella Infections/metabolism , Salmonella Infections/microbiology , Salmonella Infections/pathology , Salmonella enteritidis/growth & development , Salmonella enteritidis/metabolism , Signal Transduction , Virulence
7.
Sci Rep ; 6: 37421, 2016 11 18.
Article in English | MEDLINE | ID: mdl-27857177

ABSTRACT

microRNAs (miRNAs) have emerged as major regulators of the initiation and progression of human cancers, including breast cancer. The aim of this study is to determine the expression pattern of miR-96 in breast cancer and to investigate its biological role during tumorigenesis. We showed that miR-96 was significantly upregulated in breast cancer. We then investigated its function and found that miR-96 significantly promoted cell proliferation, migration and invasion in vitro and enhanced tumor growth in vivo. Furthermore, we explored the molecular mechanisms by which miR-96 contributes to breast cancer progression and identified PTPN9 (protein tyrosine phosphatase, non-receptor type 9) as a direct target gene of miR-96. Finally, we showed that PTPN9 had opposite effects to those of miR-96 on breast cancer cells, suggesting that miR-96 may promote breast tumorigenesis by silencing PTPN9. Taken together, this study highlights an important role for miR-96 in the regulation of PTPN9 in breast cancer cells and may provide insight into the molecular mechanisms of breast carcinogenesis.


Subject(s)
Breast Neoplasms/genetics , Carcinogenesis/genetics , MicroRNAs/genetics , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Breast Neoplasms/pathology , Cell Movement/genetics , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Neoplasm Invasiveness/genetics , Signal Transduction
8.
Oncotarget ; 7(39): 64100-64108, 2016 Sep 27.
Article in English | MEDLINE | ID: mdl-27602768

ABSTRACT

Protein tyrosine phosphatase receptor type G (PTPRG) is an important tumor suppressor gene in multiple human cancers. In this study, we found that PTPRG protein levels were downregulated in breast cancer tissues while the mRNA levels varied irregularly, implying a post-transcriptional mechanism was involved. Because microRNAs are powerful post-transcriptional regulators of gene expression, we used bioinformatics analysis to search for microRNAs that potentially targets PTPRG in the setting of breast cancer. We identified two specific binding sites for miR-19b in the 3'-untranslated region of PTPRG. We further identified an inverse correlation between miR-19b and PTPRG protein levels, but not mRNA levels, in human breast cancer tissues. By overexpressing or knocking down miR-19b in MCF-7 cells and MDA-231 cells, we experimentally confirmed that miR-19b directly suppresses PTPRG expression. Furthermore, we determined that the inhibition of PTPRG by miR-19b leads to increased proliferation, stimulated cell migration and reduced apoptosis. Taken together, our findings provide the first evidence that miR-19b inhibits PTPRG expression to promote tumorigenesis in human breast cancer.


Subject(s)
Breast Neoplasms/genetics , MicroRNAs/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 5/metabolism , 3' Untranslated Regions , Apoptosis , Binding Sites , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation/genetics , Cell Survival , Cell Transformation, Neoplastic/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 5/genetics
9.
Protein Cell ; 7(10): 722-734, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27647131

ABSTRACT

Programmed cell death 4 (PDCD4) is a RNA-binding protein that acts as a tumor suppressor in many cancer types, including colorectal cancer (CRC). During CRC carcinogenesis, PDCD4 protein levels remarkably decrease, but the underlying molecular mechanism for decreased PDCD4 expression is not fully understood. In this study, we performed bioinformatics analysis to identify miRNAs that potentially target PDCD4. We demonstrated miR-181b as a direct regulator of PDCD4. We further showed that activation of IL6/STAT3 signaling pathway increased miR-181b expression and consequently resulted in downregulation of PDCD4 in CRC cells. In addition, we investigated the biological effects of PDCD4 inhibition by miR-181b both in vitro and in vivo and found that miR-181b could promote cell proliferation and migration and suppress apoptosis in CRC cells and accelerate tumor growth in xenograft mice, potentially through targeting PDCD4. Taken together, this study highlights an oncomiR role for miR-181b in regulating PDCD4 in CRC and suggests that miR-181b may be a novel molecular therapeutic target for CRC.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Cell Proliferation , Colorectal Neoplasms/metabolism , MicroRNAs/metabolism , Neoplasm Proteins/metabolism , RNA, Neoplasm/metabolism , RNA-Binding Proteins/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Caco-2 Cells , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Heterografts , Humans , Male , Mice , Mice, Nude , Mice, SCID , MicroRNAs/genetics , Neoplasm Proteins/genetics , Neoplasm Transplantation , RNA, Neoplasm/genetics , RNA-Binding Proteins/genetics
10.
J Gen Virol ; 97(2): 299-305, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26644088

ABSTRACT

Since March 2013, more than 500 laboratory-confirmed human H7N9 influenza A virus infection cases have been recorded, with a case fatality rate of more than 30%. Clinical research has shown that cytokine and chemokine dysregulation contributes to the pathogenicity of the H7N9 virus. Here, we investigated cytokine profiles in primary human macrophages infected with the novel H7N9 virus, using cytokine antibody arrays. The levels of several pro-inflammatory cytokines, particularly TNF-α, were increased in H7N9-infected macrophages. Induction of the transcriptional and translational levels of the pro-inflammatory cytokines by H7N9 virus seemed to be intermediate between those induced by highly pathogenic avian H5N1 and pandemic human H1N1 viruses, which were detected by ELISA and real-time quantitative PCR, respectively. Additionally, compared with H5N1, the upregulation of pro-inflammatory cytokines caused by H7N9 infection occurred rapidly but mildly. Our results identified the overall profiles of cytokine and chemokine induction by the H7N9 influenza virus in an in vitro cell-culture model, and could provide potential therapeutic targets for the control of severe human H7N9 disease.


Subject(s)
Cytokines/analysis , Influenza A Virus, H7N9 Subtype/immunology , Macrophages/immunology , Macrophages/virology , Animals , Birds , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Gene Expression Profiling , Humans , Influenza A Virus, H1N1 Subtype/growth & development , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/growth & development , Influenza A Virus, H5N1 Subtype/immunology , Influenza A Virus, H7N9 Subtype/growth & development , Influenza A Virus, H7N9 Subtype/isolation & purification , Influenza in Birds/virology , Protein Array Analysis , Real-Time Polymerase Chain Reaction
11.
RNA Biol ; 12(3): 276-89, 2015.
Article in English | MEDLINE | ID: mdl-25826661

ABSTRACT

MicroRNA-200b and microRNA-200c (miR-200b/c) are 2 of the most frequently upregulated oncomiRs in colorectal cancer cells. The role of miR-200b/c during colorectal tumorigenesis, however, remains unclear. In the present study, we report that miR-200b/c can promote colorectal cancer cell proliferation via targeting the reversion-inducing cysteine-rich protein with Kazal motifs (RECK). Firstly, bioinformatics analysis predicted RECK as a conserved target of miR-200b/c. By overexpressing or knocking down miR-200b/c in colorectal cancer cells, we experimentally validated that miR-200b/c are direct regulators of RECK. Secondly, an inverse correlation between the levels of miR-200b/c and RECK protein was found in human colorectal cancer tissues and cell lines. Thirdly, we demonstrated that repression of RECK by miR-200b/c consequently triggered SKP2 (S-phase kinase-associated protein 2) elevation and p27(Kip1) (also known as cyclin-dependent kinase inhibitor 1B) degradation in colorectal cancer cells, which eventually promotes cancer cell proliferation. Finally, promoting tumor cell growth by miR-200b/c-targeting RECK was also observed in the xenograft mouse model. Taken together, our results demonstrate that miR-200b/c play a critical role in promoting colorectal tumorigenesis through inhibiting RECK expression and subsequently triggering SKP2 elevation and p27(Kip1) degradation.


Subject(s)
Carcinogenesis/genetics , Colorectal Neoplasms/genetics , GPI-Linked Proteins/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Animals , Caco-2 Cells , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Proliferation , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , GPI-Linked Proteins/metabolism , HT29 Cells , Humans , Mice , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Neoplasm Transplantation , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , S-Phase Kinase-Associated Proteins/genetics , S-Phase Kinase-Associated Proteins/metabolism , Signal Transduction , Transfection
12.
J Nutr Biochem ; 26(5): 505-12, 2015 May.
Article in English | MEDLINE | ID: mdl-25704478

ABSTRACT

The detection of exogenous plant microRNAs in human/animal plasma/sera lies at the foundation of exploring their cross-kingdom regulatory functions. It is necessary to establish a standard operation procedure to promote study in this nascent field. In this study, 18 plant miRNAs were assessed in watermelon juice and mixed fruits by quantitative reverse transcriptase polymerase chain reaction (qRT-PCR). CT values, no-template controls and standard curves for each miRNA were used to evaluate the specificity and sensitivity of qRT-PCR and to obtain concentrations. Sixteen miRNAs were selected and measured in human plasma from volunteers after drinking juice. The CT values of 6 plant miRNAs in human plasma fell outside the linear ranges of their standard curves. The remaining 10 miRNAs were present at high basal levels, and 6 of them showed a dynamic physiological pattern in plasma (absorption rates of 0.04% to 1.31%). Northern blotting was used to confirm the qRT-PCR results. Critical issues such as RNA extraction and internal controls were also addressed.


Subject(s)
MicroRNAs/blood , Plants/genetics , RNA, Plant/blood , Blotting, Northern , Humans , MicroRNAs/administration & dosage , Reverse Transcriptase Polymerase Chain Reaction
13.
PLoS One ; 9(12): e114420, 2014.
Article in English | MEDLINE | ID: mdl-25474488

ABSTRACT

Insulin-like growth factor 1 receptor (IGF1R) is a transmembrane receptor that is activated by insulin-like growth factor 1 (IGF-1) and by a related hormone called IGF-2. It belongs to the large class of tyrosine kinase receptors and plays an important role in colorectal cancer etiology and progression. In this study, we used bioinformatic analyses to search for miRNAs that potentially target IGF1R. We identified specific target sites for miR-143 and miR-145 (miR-143/145) in the 3'-untranslated region (3'-UTR) of the IGF1R gene. These miRNAs are members of a cluster of miRNAs that have been reported to exhibit tumor suppressor activity. Consistent with the bioinformatic analyses, we identified an inverse correlation between miR-143/145 levels and IGF1R protein levels in colorectal cancer tissues. By overexpressing miR-143/145 in Caco2, HT29 and SW480 colorectal cancer cells, we experimentally validated that miR-143/145 directly recognizes the 3'-UTR of the IGF1R transcript and regulates IGF1R expression. Furthermore, the biological consequences of the targeting of IGF1R by miR-143/145 were examined by cell proliferation assays in vitro. We demonstrated that the repression of IGF1R by miR-143/145 suppressed the proliferation of Caco2 cells. Taken together, our findings provide evidence for a role of the miR-143/145 cluster as a tumor suppressor in colorectal cancer through the inhibition of IGF1R translation.


Subject(s)
Adenocarcinoma/genetics , Colorectal Neoplasms/genetics , MicroRNAs/physiology , Receptors, Somatomedin/metabolism , 3' Untranslated Regions , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Base Sequence , Binding Sites , Caco-2 Cells , Cell Proliferation , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , HT29 Cells , Humans , Multigene Family , Protein Biosynthesis , RNA Interference , Receptor, IGF Type 1 , Receptors, Somatomedin/genetics
14.
Mol Cancer ; 13: 220, 2014 Sep 24.
Article in English | MEDLINE | ID: mdl-25248370

ABSTRACT

INTRODUCTION: ERBB3, one of the four members of the ErbB family of receptor tyrosine kinases, plays an important role in breast cancer etiology and progression. In the present study, we aimed to identify novel miRNAs that can potentially target ERBB3 and their biological functions. METHOD: The expression levels of miR-143/145 and target mRNA were examined by relative quantification RT-PCR, and the expression levels of target protein were detected by Western blot. We used bioinformatic analyses to search for miRNAs that can potentially target ERBB3. Luciferase reporter plasmids were constructed to confirm direct targeting. Furthermore, the biological consequences of the targeting of ERBB3 by miR-143/145 were examined by cell proliferation and invasion assays in vitro and by the mouse xenograft tumor model in vivo. RESULTS: We identified an inverse correlation between miR-143/145 levels and ERBB3 protein levels, but not between miR-143/145 levels and ERBB3 mRNA levels, in breast cancer tissue samples. We identified specific targeting sites for miR-143 and miR-145 (miR-143/145) in the 3'-untranslated region (3'-UTR) of the ERBB3 gene and regulate ERBB3 expression. We demonstrated that the repression of ERBB3 by miR-143/145 suppressed the proliferation and invasion of breast cancer cells, and that miR-143/145 showed an anti-tumor effect by negatively regulating ERBB3 in the xenograft mouse model. Interestingly, miR-143 and miR-145 showed a cooperative repression of ERBB3 expression and cell proliferation and invasion in breast cancer cells, such that the effects of the two miRNAs were greater than with either miR-143 or miR-145 alone. CONCLUSION: Taken together, our findings provide the first clues regarding the role of the miR-143/145 cluster as a tumor suppressor in breast cancer through the inhibition of ERBB3 translation. These results also support the idea that different miRNAs in a cluster can synergistically repress a given target mRNA.


Subject(s)
Breast Neoplasms/pathology , MicroRNAs/genetics , Receptor, ErbB-3/metabolism , 3' Untranslated Regions , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation , Female , Humans , MCF-7 Cells , Male , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Transplantation , Receptor, ErbB-3/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...