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










Publication year range
1.
Am J Transl Res ; 16(5): 1568-1586, 2024.
Article in English | MEDLINE | ID: mdl-38883394

ABSTRACT

Lung adenocarcinoma (LUAD) is one of the most prevalent and lethal cancers worldwide, signifying a critical need for improved prognostic tools. A growing number of studies have highlighted the role of microRNAs (miRNAs) and their regulatory functions in tumorigenesis and cancer progression. In this context, we performed an extensive analysis of bulk RNA- and miRNA-sequencing to identify LUAD-associated prognostic genes. A risk score system based on 11 miRNA-regulated and surface-protein genes was developed, which was later validated by internally and externally using the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO), respectively. Further single-cell RNA sequencing analysis revealed significant interactions between various cellular subpopulations within the tumor microenvironment, with the most pronounced differences observed between endothelial and epithelial cells. The mutational analysis highlighted TP53 as a key signaling pathway associated with the risk score. The study underscores that immune suppression, indicated by a positive association with regulatory T cells (Tregs) and an inverse correlation with M1-type macrophages, is prevalent in high-risk LUAD patients. These findings provide a promising prognostic tool for clinical outcomes of LUAD patients, facilitating future development of therapeutic strategies and enhancing our understanding of the regulatory function of miRNAs in LUAD.

2.
Front Biosci (Landmark Ed) ; 28(2): 26, 2023 02 08.
Article in English | MEDLINE | ID: mdl-36866547

ABSTRACT

BACKGROUND: The stemness characteristics of cancer cells, such as self-renewal and tumorigenicity, are considered to be responsible, in part, for tumor metastasis. Epithelial-to-mesenchymal transition (EMT) plays an important role in promoting both stemness and tumor metastasis. Although the traditional medicine juglone is thought to play an anticancer role by affecting cell cycle arrest, induction of apoptosis, and immune regulation, a potential function of juglone in regulating cancer cell stemness characteristics remains unknown. METHODS: In the present study, tumor sphere formation assay and limiting dilution cell transplantation assays were performed to assess the function of juglone in regulating maintenance of cancer cell stemness characteristics. EMT of cancer cells was assessed by western blot and transwell assay in vitro, and a liver metastasis model was also performed to demonstrate the effect of juglone on colorectal cancer cells in vivo. RESULTS: Data gathered indicates juglone inhibits stemness characteristics and EMT in cancer cells. Furthermore, we verified that metastasis was suppressed by juglone treatment. We also observed that these effects were, in part, achieved by inhibiting Peptidyl-prolyl cis-trans isomerase NIMA-interacting 1 (Pin1). CONCLUSIONS: These results indicate that juglone inhibits maintenance of stemness characteristics and metastasis in cancer cells.


Subject(s)
Epithelial-Mesenchymal Transition , Naphthoquinones , Neoplasms , Neoplastic Stem Cells , Apoptosis , Blotting, Western , Neoplasms/drug therapy , Neoplasm Metastasis/prevention & control , Naphthoquinones/pharmacology
3.
Stem Cell Res Ther ; 14(1): 25, 2023 02 13.
Article in English | MEDLINE | ID: mdl-36782269

ABSTRACT

BACKGROUND: Hair follicle stem cells (HFSCs) are considered as a promising cell type in the stem cell transplantation treatment of neurological diseases because of their rich sources, easy access, and the same ectoderm source as the nervous system. Hepatocyte growth factor (HGF) is a pleiotropic cytokine that shows neuroprotective function in ischemic stroke. Here we assessed the therapeutic effects of HFSCs on ischemic stroke injury and the synthetic effect of HGF along with HFSCs. METHODS: Rat HFSCs were intravenously transplanted into a middle cerebral artery ischemia/reperfusion (I/R) rat model. Neurological scoring and TTC staining were performed to assess the benefits of HFSC transplantation. Inflammatory cytokines, blood-brain barrier integrity and angiogenesis within penumbra were estimated by Western blot and immunohistochemistry. The differentiation of HFSCs was detected by immunofluorescence method 2 weeks after transplantation. RESULTS: HFSC transplantation could significantly inhibit the activation of microglia, improve the integrity of blood-brain barrier and reduce brain edema. Moreover, the number of surviving neurons and microvessels density in the penumbra were upregulated by HFSC transplantation, leading to better neurological score. The combination of HFSCs and HGF could significantly improve the therapeutic benefit. CONCLUSION: Our results indicate for the first time that HGF modified HFSCs can reduce I/R injury and promote the neurological recovery by inhibiting inflammatory response, protecting blood-brain barrier and promoting angiogenesis.


Subject(s)
Brain Ischemia , Ischemic Stroke , Reperfusion Injury , Rats , Animals , Hepatocyte Growth Factor/metabolism , Hair Follicle , Reperfusion Injury/therapy , Reperfusion Injury/metabolism , Stem Cells/metabolism , Brain Ischemia/therapy , Brain Ischemia/metabolism
4.
Cell Death Dis ; 13(1): 21, 2021 12 20.
Article in English | MEDLINE | ID: mdl-34930897

ABSTRACT

Premature ovarian insufficiency (POI) is a heterogeneous and multifactorial disorder. In recent years, there has been an increasing interest in research on the pathogenesis and treatment of POI, owing to the implementation of the second-child policy in China. Cytoplasmic polyadenylation element-binding protein 3 (CPEB3) is an RNA-binding protein that can bind to specific RNA sequences. CPEB3 can bind to and affect the expression, cellular location, and stability of target RNAs. Cpeb3 is highly expressed in the ovary; however, its functions remain unknown. In this study, Cpeb3-mutant mice were used to characterize the physiological functions of CPEB3. Cpeb3-mutant female mice manifested signs of gradual loss of ovarian follicles, ovarian follicle development arrest, increased follicle atresia, and subfertility with a phenotype analogous to POI in women. Further analysis showed that granulosa cell proliferation was inhibited and apoptosis was markedly increased in Cpeb3-mutant ovaries. In addition, the expression of Gdf9, a potential target of CPEB3, was decreased in Cpeb3-mutant ovaries and oocytes. Altogether, these results reveal that CPEB3 is essential for ovarian follicle development and female fertility as it regulates the expression of Gdf9 in oocytes, disruption of which leads to impaired ovarian follicle development and POI.


Subject(s)
Fertility/genetics , Granulosa Cells/metabolism , Mutation , Primary Ovarian Insufficiency/metabolism , RNA-Binding Proteins/metabolism , Signal Transduction/genetics , Animals , Apoptosis/genetics , CRISPR-Cas Systems , Cell Proliferation/genetics , Disease Models, Animal , Female , Growth Differentiation Factor 9/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oocytes/metabolism , Phenotype , Pregnancy , Primary Ovarian Insufficiency/genetics , RNA-Binding Proteins/genetics
5.
Cell Death Dis ; 11(9): 792, 2020 09 23.
Article in English | MEDLINE | ID: mdl-32968053

ABSTRACT

Cytoplasmic polyadenylation element-binding protein 3 (CPEB3) is a sequence-specific RNA-binding protein. We had reported that CPEB3 is involved in hepatocellular carcinoma (HCC) progression. However, the underlying mechanisms of CPEB3 in HCC remain unclear. In this study, we firstly performed RNA immunoprecipitation to uncover the transcriptome-wide CPEB3-bound mRNAs (CPEB3 binder) in HCC. Bioinformatic analysis indicates that CPEB3 binders are closely related to cancer progression, especially HCC metastasis. Further studies confirmed that metadherin (MTDH) is a direct target of CPEB3. CPEB3 can suppress the translation of MTDH mRNA in vivo and in vitro. Besides, luciferase assay demonstrated that CPEB3 interacted with 3'-untranslated region of MTDH mRNA and inhibited its translation. Subsequently, CPEB3 inhibited the epithelial-mesenchymal transition and metastasis of HCC cells through post-transcriptional regulation of MTDH. In addition, cpeb3 knockout mice are more susceptible to carcinogen-induced hepatocarcinogenesis and subsequent lung metastasis. Our results also indicated that CPEB3 was a good prognosis marker, which is downregulated in HCC tissue. In conclusion, our results demonstrated that CPEB3 played an important role in HCC progression and targeting CPEB3-mediated mRNA translation might be a favorable therapeutic approach.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Membrane Proteins/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Carcinogenesis/genetics , Carcinoma, Hepatocellular/metabolism , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Liver Neoplasms/metabolism , Membrane Proteins/metabolism
6.
Int Immunopharmacol ; 73: 118-127, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31085459

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) contribute to immune activity suppression and promote the tumor progression. Elimination of MDSCs is a promising cancer therapeutic strategy, and some chemotherapeutic agents have been reported to hamper tumor progression by suppressing MDSCs. Juglone has been showed to exert a direct cytotoxic effect on tumor cells. However, the effect of juglone on MDSCs and anti-tumor immune statue has remained unexplored. In our study, we observed that juglone suppressed tumor growth and metastasis markedly, and the tumor growth suppression in immunocompetent mice was more drastic than that in immunodeficient mice. Juglone reduced the accumulation of MDSCs and increased IFN-γ production by CD8+ T cells. Consistently, juglone affected myeloid cells differentiation and maturation, impairing the immunosuppressive functions of MDSCs. Moreover, juglone down-regulated the level of IL-1ß which was mediating accumulation of MDSCs. In addition, juglone inhibited 5FU-induced liver injury in a colorectal carcinoma-bearing mice model. Thus, our work suggests that the anti-tumor effect of juglone is mediated, at least in part, by eliminating accumulation of MDSCs.


Subject(s)
Antineoplastic Agents/pharmacology , Myeloid-Derived Suppressor Cells/drug effects , Naphthoquinones/pharmacology , Neoplasms/immunology , Animals , Antineoplastic Agents/therapeutic use , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Chemical and Drug Induced Liver Injury/drug therapy , Fluorouracil/adverse effects , Interferon-gamma/immunology , Interleukin-1beta/immunology , Liver/drug effects , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Naphthoquinones/therapeutic use , Neoplasms/drug therapy , Neoplasms/pathology
7.
J Exp Clin Cancer Res ; 37(1): 46, 2018 Mar 05.
Article in English | MEDLINE | ID: mdl-29506532

ABSTRACT

BACKGROUND: Cell adhesion molecules (CADMs) comprise of a protein family whose functions include maintenance of cell polarity and tumor suppression. Hypo-expression of CADM2 gene expression has been observed in several cancers including hepatocellular carcinoma (HCC). However, the role and mechanisms of CADM2 in HCC remain unclear. METHODS: The expression of CADM2 and miRNA-10b (miR-10b) in HCC tissues and cell lines were detected using real-time PCR and Western blotting. Immunofluorescence was used to detect Epithelial-mesenchymal transition (EMT) progression in HCC cell lines. Dual-luciferase reporter assay was used to determine miR-10b binding to CADM2 3'UTR. Wound healing assay and Transwell assay were performed to examine the migration and invasion of HCC cells. RESULTS: We report the effect of CADM2 as a tumor suppressor in HCC. Firstly, we confirmed that CADM2 expression was significantly down regulated in HCC tissues compared to normal tissues according to TCGA data analysis and fresh HCC sample detection. Secondly, overexpression of CADM2 could inhibit EMT process, migratory and invasion ability of HCC cells. Furthermore, the results indicated that CADM2 is a direct target of miR-10b in HCC cells and miR-10b/CADM2 modulates EMT process and migration ability via focal adhesion kinase (FAK) /AKT signaling pathway in HCC. CONCLUSIONS: Our study demonstrates that miR-10b-CADM2-FAK/AKT axis plays an important role in HCC metastasis, which might be a novel potential therapeutic option for HCC treatment.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Adhesion Molecules/genetics , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , 3' Untranslated Regions , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Cell Adhesion Molecules/metabolism , Cell Movement/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression , Genes, Reporter , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Models, Biological , Neoplasm Metastasis , Neoplasm Staging , Prognosis , RNA Interference
9.
Oncol Rep ; 36(5): 2715-2722, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27571925

ABSTRACT

Heme metabolism system is involved in microRNA (miRNA) biogenesis. The complicated interplay between heme oxygenase-1 (HO-1) and miRNA has been observed in various tissues and diseases, including human malignancy. In the present study, our data showed that stable HO-1 overexpression in hepatocellular carcinoma (HCC) cells downregulated several oncomiRs. The most stably downregulated are miR-30d and miR-107. Iron, one of HO-1 catalytic products, was an important mediator in this regulation. Cell function analysis demonstrated that HO-1 inhibited the proliferation and metastasis of HepG2 cells, whereas miR-30d/miR-107 improved the proliferative and migratory ability of HepG2 cells. The beneficial effect of HO-1 in HCC inhibition could be reversed by upregulating miR-30d and miR-107. Akt and ERK pathways may be involved in the regulation of HO-1/miR-30d/miR-107 in HCC. These data indicate that HO-1 significantly suppresses HCC progression by regulating the miR-30d/miR-107 level, suggesting miR-30d/miR-107 regulation as a new molecular mechanism of HO-1 anticancer effect.


Subject(s)
Carcinoma, Hepatocellular/genetics , Heme Oxygenase-1/biosynthesis , Liver Neoplasms/genetics , MicroRNAs/genetics , Carcinoma, Hepatocellular/pathology , Cell Proliferation/genetics , Disease Progression , Gene Expression Regulation, Neoplastic , Heme Oxygenase-1/genetics , Hep G2 Cells , Humans , Liver Neoplasms/pathology , MicroRNAs/biosynthesis , Signal Transduction
10.
Oncotarget ; 7(1): 266-78, 2016 Jan 05.
Article in English | MEDLINE | ID: mdl-26497556

ABSTRACT

MicroRNAs (miRNAs) are dysregulated in many types of malignancies, including human hepatocellular carcinoma (HCC). MiR-107 has been implicated in several types of cancer regulation; however, relatively little is known about miR-107 in human HCC. In the present study, we showed that the overexpression of miR-107 accelerates the tumor progression of HCC in vitro and in vivo through its new target gene, CPEB3. Furthermore, our results demonstrated that CPEB3 is a newly discovered tumor suppressor that acts via the EGFR pathway. Therefore, our study demonstrates that the newly discovered miR-107/CPEB3/EGFR axis plays an important role in HCC progression and might represent a new potential therapeutic target for HCC treatment.


Subject(s)
Carcinoma, Hepatocellular/genetics , ErbB Receptors/genetics , Liver Neoplasms/genetics , MicroRNAs/genetics , RNA-Binding Proteins/genetics , 3' Untranslated Regions/genetics , Animals , Blotting, Western , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Disease Progression , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , RNA Interference , RNA-Binding Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous , Tumor Burden/genetics
11.
PLoS One ; 10(3): e0120698, 2015.
Article in English | MEDLINE | ID: mdl-25799050

ABSTRACT

MicroRNAs (miRNAs) are deregulated in a number of cancers including colorectal cancer. MiR-30c belongs to miR-30 family, and is involved in a variety of malignant diseases. In this study, we detected the expression of miR-30c in colon cancer cell lines and clinical colon cancer specimens. MiR-30c was shown to be dramatically down-regulated both in cell lines and cancer tissues. Additionally, miR-30c could inhibit cancer cell growth, migration and invasion in vitro. Consistently, stable over-expression of miR-30c inhibited the growth and lung metastasis of colon cancer cell xenografts in vivo. Furthermore, bioinformatics algorithm and luciferase reporter assay indicated ADAM19 as a direct target of miR-30c. Of interest, further experiments demonstrated that inhibition of ADAM19 by miR-30c partially mediated the anti-tumor effect of miR-30c. Overall, our study provides the new insight that miR-30c inhibited colon cancer cells via targeting ADAM19. Thus, miR-30c might serve as a promising therapeutic strategy for colon cancer treatment.


Subject(s)
ADAM Proteins/genetics , Colorectal Neoplasms/genetics , MicroRNAs/genetics , RNA Interference , ADAM Proteins/chemistry , Aged , Aged, 80 and over , Animals , Base Sequence , Binding Sites , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Colorectal Neoplasms/pathology , Disease Models, Animal , Down-Regulation , Female , Gene Expression , Gene Expression Regulation, Neoplastic , Genes, Reporter , Humans , Male , Mice , MicroRNAs/chemistry , Middle Aged , Neoplasm Grading , Neoplasm Staging , Transfection , Tumor Burden , Xenograft Model Antitumor Assays
12.
Mol Cell Biol ; 35(6): 988-1000, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25582198

ABSTRACT

MicroRNAs (miRNAs) are dysregulated in many types of malignant diseases, including colorectal cancer. miRNA 30a (miR-30a) is a member of the miR-30 family and has been implicated in many types of cancers. In this study, we determined the expression of miR-30a in human colon cancer tissues and cell lines. miR-30a was found to be significantly downregulated in both the tissues and cell lines. Furthermore, overexpression of miR-30a inhibited, while silencing of miR-30a promoted, cell proliferation, migration, and invasion in vitro. Consistently, stable overexpression of miR-30a suppressed the growth of colon cancer cell xenografts in vivo. Moreover, bioinformatic algorithms and luciferase reporter assays revealed that insulin receptor substrate 2 (IRS2) is a direct target of miR-30a. Further functional studies suggested that repression of IRS2 by miR-30a partially mediated the tumor suppressor effect of miR-30a. In addition, miR-30a inhibited constitutive phosphorylation of Akt by targeting IRS2. Additionally, clinicopathological analysis indicated that miR-30a has an inverse correlation with the staging in patients with colon cancer. Taken together, our study provides the first evidence that miR-30a suppressed colon cancer cell growth through inhibition of IRS2. Thus, miR-30a might serve as a promising therapeutic strategy for colon cancer treatment.


Subject(s)
Carcinogenesis/genetics , Colorectal Neoplasms/genetics , Insulin Receptor Substrate Proteins/genetics , MicroRNAs/genetics , Animals , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Down-Regulation/genetics , Female , Genes, Tumor Suppressor , HCT116 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Proto-Oncogene Proteins c-akt/genetics
13.
Tumour Biol ; 36(3): 1701-10, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25371073

ABSTRACT

MicroRNA-93 (miR-93) is involved in several carcinoma progressions. It has been reported that miR-93 acts as a promoter or suppressor in different tumors. However, till now, the role of miR-93 in colon cancer is unclear. Herein, we have found that expression of miR-93 was lower in human colon cancer tissue and colorectal carcinoma cell lines compared with normal colon mucosa. Forced expression of miR-93 in colon cancer cells inhibits colon cancer invasion, migration, and proliferation. Furthermore, miR-93 may downregulate the Wnt/ß-catenin pathway, which was confirmed by measuring the expression level of the ß-catenin, axin, c-Myc, and cyclin-D1 in this pathway. Mothers against decapentaplegic homolog 7 (Smad7), as an essential molecular protein for nuclear accumulation of ß-catenin in the canonical Wnt signaling pathway, is predicted as a putative target gene of miR-93 by the silico method and demonstrated that it may be suppressed by targeting its 3'UTR. These findings showed that miR-93 suppresses colorectal cancer development via downregulating Wnt/ß-catenin, at least in part, by targeting Smad7. This study revealed that miR-93 is an important negative regulator in colon cancer and suggested that miR-93 may serve as a novel therapeutic agent that offers benefits for colon cancer treatment.


Subject(s)
Colonic Neoplasms/genetics , MicroRNAs/genetics , Wnt Proteins/genetics , 3' Untranslated Regions , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Cyclin D1/genetics , Down-Regulation , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , HCT116 Cells , HT29 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Signal Transduction , beta Catenin/genetics
14.
Nucleic Acids Res ; 41(9): e101, 2013 May.
Article in English | MEDLINE | ID: mdl-23482392

ABSTRACT

Various 'omics' technologies, including microarrays and gas chromatography mass spectrometry, can be used to identify hundreds of interesting genes, proteins and metabolites, such as differential genes, proteins and metabolites associated with diseases. Identifying metabolic pathways has become an invaluable aid to understanding the genes and metabolites associated with studying conditions. However, the classical methods used to identify pathways fail to accurately consider joint power of interesting gene/metabolite and the key regions impacted by them within metabolic pathways. In this study, we propose a powerful analytical method referred to as Subpathway-GM for the identification of metabolic subpathways. This provides a more accurate level of pathway analysis by integrating information from genes and metabolites, and their positions and cascade regions within the given pathway. We analyzed two colorectal cancer and one metastatic prostate cancer data sets and demonstrated that Subpathway-GM was able to identify disease-relevant subpathways whose corresponding entire pathways might be ignored using classical entire pathway identification methods. Further analysis indicated that the power of a joint genes/metabolites and subpathway strategy based on their topologies may play a key role in reliably recalling disease-relevant subpathways and finding novel subpathways.


Subject(s)
Metabolic Networks and Pathways/genetics , Metabolomics , Transcriptome , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Histamine/metabolism , Humans , Male , Neoplasm Metastasis , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology
15.
Cancer Lett ; 335(1): 168-74, 2013 Jul 10.
Article in English | MEDLINE | ID: mdl-23435373

ABSTRACT

Because it is a member of the miR-106b~25 cluster, microRNA-25 (miR-25) is known to be dysregulated in human cancers. However, the expression and role of miR-25 in colon cancer remain unclear. In this study, miR-25 was found to be down-regulated in human colon cancer tissues when compared to those in matched, non-neoplastic mucosa tissues. Functional studies revealed that restoration of miR-25 expression inhibited cell proliferation and migration. In contrast, miR-25 inhibition could promote the proliferation and migratory ability of cells. Stable over-expression of miR-25 also suppressed the growth of colon cancer-cell xenografts in vivo. Furthermore, bioinformatic predictions and experimental validation were used to identify Smad7 as a direct target of miR-25. Functional reverse experiments indicated that the antitumor effects of miR-25 were probably mediated by its repression of Smad7. These results suggest that miR-25 may function as a tumor suppressor by targeting Smad7 in colon cancer. Thus, miR-25 may serve as a potential therapeutic agent or target for cancer therapy.


Subject(s)
Colonic Neoplasms/metabolism , Genes, Tumor Suppressor , MicroRNAs/genetics , RNA Interference , Smad7 Protein/genetics , 3' Untranslated Regions , Aged , Animals , Base Sequence , Binding Sites , Cell Movement , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/metabolism , MicroRNAs/physiology , Middle Aged , Neoplasm Transplantation , Smad7 Protein/metabolism , Tumor Burden/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...