Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Yebo Fu is active.

Publication


Featured researches published by Yebo Fu.


Journal of Cancer | 2012

Cell-free Circulating miRNA Biomarkers in Cancer

Meng-Hsuan Mo; Liang Chen; Yebo Fu; Wendy Wang; Sidney W. Fu

Considerable attention and an enormous amount of resources have been dedicated to cancer biomarker discovery and validation. However, there are still a limited number of useful biomarkers available for clinical use. An ideal biomarker should be easily assayed with minimally invasive medical procedures but possess high sensitivity and specificity. Commonly used circulating biomarkers are proteins in serum, most of which require labor-intensive analysis hindered by low sensitivity in early tumor detection. Since the deregulation of microRNA (miRNA) is associated with cancer development and progression, profiling of circulating miRNAs has been used in a number of studies to identify novel minimally invasive miRNA biomarkers. In this review, we discuss the origin of the circulating cell-free miRNAs and their carriers in blood. We summarize the clinical use and function of potentially promising miRNA biomarkers in a variety of different cancers, along with their downstream target genes in tumor initiation and development. Additionally, we analyze some technical challenges in applying miRNA biomarkers to clinical practice.


PLOS ONE | 2013

Role of deregulated microRNAs in breast cancer progression Using FFPE tissue

Liang Chen; Youhuai Li; Yebo Fu; Jin Peng; Meng Hsuan Mo; Michael Stamatakos; Christine B. Teal; Rachel F. Brem; Alexander Stojadinovic; Michael Grinkemeyer; Timothy A. McCaffrey; Yan Gao Man; Sidney W. Fu

MicroRNAs (miRNAs) contribute to cancer initiation and progression by silencing the expression of their target genes, causing either mRNA molecule degradation or translational inhibition. Intraductal epithelial proliferations of the breast are histologically and clinically classified into normal, atypical ductal hyperplasia (ADH), ductal carcinoma in situ (DCIS) and invasive ductal carcinoma (IDC). To better understand the progression of ductal breast cancer development, we attempt to identify deregulated miRNAs in this process using Formalin-Fixed, Paraffin-Embedded (FFPE) tissues from breast cancer patients. Following tissue microdissection, we obtained 8 normal, 4 ADH, 6 DCIS and 7 IDC samples, which were subject to RNA isolation and miRNA expression profiling analysis. We found that miR-21, miR-200b/c, miR-141, and miR-183 were consistently up-regulated in ADH, DCIS and IDC compared to normal, while miR-557 was uniquely down-regulated in DCIS. Interestingly, the most significant miRNA deregulations occurred during the transition from normal to ADH. However, the data did not reveal a step-wise miRNA alteration among discrete steps along tumor progression, which is in accordance with previous reports of mRNA profiling of different stages of breast cancer. Furthermore, the expression of MSH2 and SMAD7, two important molecules involving TGF-β pathway, was restored following miR-21 knockdown in both MCF-7 and Hs578T breast cancer cells. In this study, we have not only identified a number of potential candidate miRNAs for breast cancer, but also found that deregulation of miRNA expression during breast tumorigenesis might be an early event since it occurred significantly during normal to ADH transition. Consequently, we have demonstrated the feasibility of miRNA expression profiling analysis using archived FFPE tissues, typically with rich clinical information, as a means of miRNA biomarker discovery.


Breast Cancer Research | 2014

miR-638 mediated regulation of BRCA1 affects DNA repair and sensitivity to UV and cisplatin in triple-negative breast cancer

Xiaohui Tan; Jin Peng; Yebo Fu; Shejuan An; M. Katayoon Rezaei; Sana Tabbara; Christine B. Teal; Yan-gao Man; Rachel F. Brem; Sidney W. Fu

IntroductionTriple-negative breast cancer (TNBC) represents 15 to 20% of all types of breast cancer; however, it accounts for a large number of metastatic cases and deaths, and there is still no effective treatment. The deregulation of microRNAs (miRNAs) in breast cancer has been widely reported. We previously identified that miR-638 was one of the most deregulated miRNAs in breast cancer progression. Bioinformatics analysis revealed that miR-638 directly targets BRCA1. The aim of this study was to investigate the role of miR-638 in breast cancer prognosis and treatment.MethodsFormalin-fixed, paraffin-embedded (FFPE) breast cancer samples were microdissected into normal epithelial and invasive ductal carcinoma (IDC) cells, and total RNA was isolated. Several breast cancer cell lines were used for the functional analysis. miR-638 target genes were identified by TARGETSCAN-VERT 6.2 and miRanda. The expression of miR-638 and its target genes was analyzed by real-time qRT-PCR and Western blotting. Dual-luciferase reporter assay was employed to confirm the specificity of miR-638 target genes. The biological function of miR-638 was analyzed by MTT chemosensitivity, matrigel invasion and host cell reactivation assays.ResultsThe expression of miR-638 was decreased in IDC tissue samples compared to their adjacent normal controls. The decreased miR-638 expression was more prevalent in non-TNBC compared with TNBC cases. miR-638 expression was significantly downregulated in breast cancer cell lines compared to the immortalized MCF-10A epithelial cells. BRCA1 was predicted as one of the direct targets of miR-638, which was subsequently confirmed by dual-luciferase reporter assay. Forced expression of miR-638 resulted in a significantly reduced proliferation rate as well as decreased invasive ability in TNBC cells. Furthermore, miR-638 overexpression increased sensitivity to DNA-damaging agents, ultraviolet (UV) and cisplatin, but not to 5-fluorouracil (5-FU) and epirubicin exposure in TNBC cells. Host cell reactivation assays showed that miR-638 reduced DNA repair capability in post UV/cisplatin-exposed TNBC cells. The reduced proliferation, invasive ability, and DNA repair capabilities are associated with downregulated BRCA1 expression.ConclusionsOur findings suggest that miR-638 plays an important role in TNBC progression via BRCA1 deregulation. Therefore, miR-638 might serve as a potential prognostic biomarker and therapeutic target for breast cancer.


Acta Biomaterialia | 2015

Engineering a biomimetic three-dimensional nanostructured bone model for breast cancer bone metastasis study.

Wei Zhu; Mian Wang; Yebo Fu; Nathan J. Castro; Sidney W. Fu; Lijie Grace Zhang

Traditional breast cancer (BrCa) bone metastasis models contain many limitations with regards to controllability, reproducibility and flexibility of design. In this study, a novel biomimetic bone microenvironment was created by integrating hydroxyapatite (HA) and native bioactive factors deposited by osteogenic induction of human bone marrow mesenchymal stem cells (MSCs) within a cytocompatible chitosan hydrogel. It was found that a 10% nanocrystalline HA (nHA) chitosan scaffold exhibited the highest BrCa adhesion and proliferation when compared to chitosan scaffolds with 20% nHA, 10% and 20% microcrystalline HA as well as amorphous HA. This 3-D tunable bone scaffold can provide a biologically relevant environment, increase cell-cell and cell-matrix interactions as found in native bone, and retain the behavior of BrCa cells with different metastasis potential (i.e. highly metastatic MDA-MB-231, less metastatic MCF-7 and transfected MDA-MB-231). The co-culture of MSCs and MDA-MB-231 in this bone model illustrated that MSCs have the capacity to upregulate the expression of the well-known metastasis-associated gene metadherin within BrCa cells. In summary, this study illustrates the ability of our 3-D bone model to create a biomimetic environment conducive to recapitulating the behavior of metastatic BrCa cells, making it a promising tool for in vitro BrCa cell bone metastasis study and for the discovery of potential therapeutics.


Oncotarget | 2016

miR-671-5p inhibits epithelial-to-mesenchymal transition by downregulating FOXM1 expression in breast cancer

Xiaohui Tan; Yebo Fu; Liang Chen; Woojin Lee; Yinglei Lai; M. Katayoon Rezaei; Sana Tabbara; Patricia S. Latham; Christine B. Teal; Yan-gao Man; Robert S. Siegel; Rachel F. Brem; Sidney W. Fu

MicroRNA (miRNA) dysfunction is associated with a variety of human diseases, including cancer. Our previous study showed that miR-671-5p was deregulated throughout breast cancer progression. Here, we report for the first time that miR-671-5p is a tumor-suppressor miRNA in breast tumorigenesis. We found that expression of miR-671-5p was decreased significantly in invasive ductal carcinoma (IDC) compared to normal in microdissected formalin-fixed, paraffin-embedded (FFPE) tissues. Forkhead Box M1 (FOXM1), an oncogenic transcription factor, was predicted as one of the direct targets of miR-671-5p, which was subsequently confirmed by luciferase assays. Forced expression of miR-671-5p in breast cancer cell lines downregulated FOXM1 expression, and attenuated the proliferation and invasion in breast cancer cell lines. Notably, overexpression of miR-671-5p resulted in a shift from epithelial-to-mesenchymal transition (EMT) to mesenchymal-to-epithelial transition (MET) phenotypes in MDA-MB-231 breast cancer cells and induced S-phase arrest. Moreover, miR-671-5p sensitized breast cancer cells to cisplatin, 5-fluorouracil (5-FU) and epirubicin exposure. Host cell reactivation (HCR) assays showed that miR-671-5p reduces DNA repair capability in post-drug exposed breast cancer cells. cDNA microarray data revealed that differentially expressed genes when miR-671-5p was transfected are associated with cell proliferation, invasion, cell cycle, and EMT. These data indicate that miR-671-5p functions as a tumor suppressor miRNA in breast cancer by directly targeting FOXM1. Hence, miR-671-5p may serve as a novel therapeutic target for breast cancer management.


Journal of Cancer | 2014

MicroRNA-21 Down-regulates Rb1 Expression by Targeting PDCD4 in Retinoblastoma

Fengmei Shen; Meng-Hsuan Mo; Liang Chen; Shejuan An; Xiaohui Tan; Yebo Fu; M. Katayoon Rezaei; Zuoren Wang; Lin Zhang; Sidney W. Fu

Retinoblastoma (RB) is a childrens ocular cancer caused by mutated retinoblastoma 1 (Rb1) gene on both alleles. Rb1 and other related genes could be regulated by microRNAs (miRNA) via complementarily pairing with their target sites. MicroRNA-21 (miR-21) possesses the oncogenic potential to target several tumor suppressor genes, including PDCD4, and regulates tumor progression and metastasis. However, the mechanism of how miR-21 regulates PDCD4 is poorly understood in RB. We investigated the expression of miRNAs in RB cell lines and identified that miR-21 is one of the most deregulated miRNAs in RB. Using qRT-PCR, we verified the expression level of several miRNAs identified by independent microarray assays, and analyzed miRNA expression patterns in three RB cell lines, including Weri-Rb1, Y79 and RB355. We found that miR-19b, -21, -26a, -195 and -222 were highly expressed in all three cell lines, suggesting their potential role in RB tumorigenesis. Using the TargetScan program, we identified a list of potential target genes of these miRNAs, of which PDCD4 is one the targets of miR-21. In this study, we focused on the regulatory mechanism of miR-21 on PDCD4 in RB. We demonstrated an inverse correlation between miR-21 and PDCD4 expression in Weri-Rb1 and Y79 cells. These data suggest that miR-21 down-regulates Rb1 by targeting PDCD4 tumor suppressor. Therefore, miR-21 could serve as a therapeutic target for retinoblastoma.


Cancer Research | 2015

Abstract 3062: miR-671-5p promotes epithelial-to-mesenchymal transition by downregulating FOXM1 expression in breast cancer

Xiaohui Tan; Yebo Fu; Liang Chen; Shejuan An; Woojin Lee; Yinglei Lai; Katayoon Rezaei; Sana Tabbara; Christine B. Teal; Yan-gao Man; Robert J. Siegel; Rachel F. Brem; Sidney W. Fu

microRNA (miRNA) dysfunction is associated with a variety of human diseases including cancer. Our previous study showed that miR-671-5p was deregulated during breast cancer progression. We aim to decipher the functional mechanism of miR-671-5p in breast cancer. We found that expression of miR-671-5p was decreased significantly in ductal carcinoma in situ (DCIS) and invasive ductal carcinoma (IDC) compared to normal and atypical ductal hyperplasia (ADH) in the microdissected formalin-fixed, paraffin-embedded (FFPE) tissues. Forkhead Box M1 (FOXM1), as an oncogenic transcription factor, was predicted as one of the direct targets of miR-671-5p, which was subsequently confirmed by luciferase assays. Forced expression of miR-671-5p in breast cancer cell lines downregulated FOXM1 expression, and attenuated the proliferation and invasion in breast cancer cell lines. Notably, overexpression of miR-671-5p resulted in a shift from epithelial-to-mesenchymal transition (EMT) to mesenchymal-to-epithelial transition (MET) phenotypes in MDA-MB-231 breast cancer cells and induced S-phase arrest. Moreover, miR-671-5p sensitized breast cancer cells to cisplatin, 5-fluorouracil (5-FU) and epirubicin exposure. Host cell reactivation (HCR) assays showed that miR-671-5p reduces DNA repair capability in post-drug exposed breast cancer cells. These data indicates that miR-671-5p functions as a tumor suppressor miRNA by directly targeting FOXM1 in breast cancer. Hence, miR-671-5p may serve as a novel therapeutic target for breast cancer management. Citation Format: Xiaohui Tan, Yebo Fu, Liang Chen, Shejuan An, Woojin Lee, Yinglei Lai, Katayoon Rezaei, Sana Tabbara, Christine B. Teal, Yan-gao Man, Robert Siegel, Rachel F. Brem, Sidney W. Fu. miR-671-5p promotes epithelial-to-mesenchymal transition by downregulating FOXM1 expression in breast cancer. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 3062. doi:10.1158/1538-7445.AM2015-3062


Cancer Research | 2013

Abstract 859: miR-638, a novel tumor suppressor for ER-negative breast cancer.

Jin Peng; Yebo Fu; Rachel F. Brem; Christine B. Teal; Sidney W. Fu

MicroRNAs (miRNAs) are endogenous non-coding RNAs of 19∼25 nt in length, which regulate many protein-coding genes. miRNA dysregulation is observed in a variety of cancers, including breast cancer. miRNAs have emerged as an important set of biomarkers. miRNA profiling studies have led to the identification of miRNAs that are aberrantly expressed in human breast cancer, with miR-10b, miR-125b and miR-145 being down-regulated and miR-21, and 155 being up-regulated. In our previous miRNA expression profiling studies using microdissected FFPE tissues, we identified 8 clusters of miRNAs, which discrete the normal and breast lesions such as ADH, DCIS and IDC. miR-638 is one of the differentially expressed miRNAs during breast cancer progression. We first sought to determine the expression of miR-638 in different breast cancer cell lines such as MCF-7, Hs578T, T47D and MDA-MB-231 using qRT-PCR, which shows a very low expression compared to normal tissues. This led us to hypothesize that miR-638 may function as a tumor suppressor during breast cancer development. To determine the role of miR-638 in both ER+ and ER- breast cancer cells, we transfected miR-638 mimic oligos and scrambled oligo mocks into MCF-7 (ER+), T47D (ER+), MDA-MB-231 (ER-) and Hs578T (ER-) cells. After 48 hours, we found that the proliferation rate was significantly down-regulated in ER- cells but not in ER+ cells by MTT assay. Our preliminary matrigel analysis shows that miR-638 overexpressing MDA-MB-231 cells had 3∼4 fold invasiveness decrease compared to the control. Further, our TargetScan analysis revealed that BRCA1 is one of the direct targets of miR-638 among the 30 conservative target genes. We found that overexpression of miR-638 resulted in an upregulation of BRCA1 mRNA expressions in ER- cell lines, MDA-MB-231 and Hs578T but not in ER+ cell lines, T47D and MCF-7. Taken together, these findings showed that miR-638 may function as a tumor suppressor in ER- breast cancer, possibly in part by upregulating BRCA1 tumor suppressor gene. Further functional analysis is underway to decipher the exact role of miR-638 in ER- breast cancer, which could become a therapeutic target. Citation Format: Jin Peng, Yebo Fu, Rachel F. Brem, Christine B. Teal, Sidney W. Fu. miR-638, a novel tumor suppressor for ER-negative breast cancer. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 859. doi:10.1158/1538-7445.AM2013-859


Cancer Research | 2010

Abstract 3910: DLX4 homeprotein enhances the invasiveness of Hs578T ER-negative breast cancer cells

Yebo Fu; Yi Lian; Kyung Soon Kim; Norman H. Lee; Sidney W. Fu

Tumor invasion and metastasis remain a major cause of mortality in breast cancer patients. It was reported that BP1, a homeobox isoform of DLX4, is overexpressed in 80% of breast cancer patients and in 100% of estrogen receptor negative (ER-) tumors. The prevalence of BP1 positive cells and the intensity of BP1 immunoreactivity increased with the extent of ductal proliferation and tumorigenesis. These findings imply that BP1 may play an important role in ER- breast cancer. We sought to determine the effects and mechanisms of BP1 on cell proliferation and metastasis using ER- Hs578T cells as a model. Cells were transfected with either pcDNA3.2 plasmid containing BP1 gene, or pcDNA3.2 vector, then selected and cloned. Overexpression of BP1 increased cell proliferation rate by 2-5 fold (p =2.0. Of those genes, 49 were up-regulated and 22 were down-regulated. Further analysis has identified several important molecular pathways using the differentially expressed gene list, including CREB signaling, BCL2L1, AKT signaling, apoptosis, androgen receptor pathways, etc. CREB1, as an oncogenic transcription factor, induces transcription of genes in response to hormonal stimulation of the cAMP pathway. CREB1 can be activated through phosphorylation by a number of kinases, including AKT, protein kinase A, and calcium/calmodulin-dependent kinases and regulates genes whose deregulated expression promotes oncogenesis, including cyclins, BCL2 family members, and EGR1. It was reported that BP1 can directly bind to and activate the expression of BCL1, which is an antiapoptotic gene and considered to be an oncogene. BCL2L1 is a BCL2 protein family member that acts as anti- or pro-apoptotic regulators involving in a wide variety of cellular activities. These data demonstrated that overexpression of BP1 significantly enhanced cell proliferation and metastatic potential in ER- Hs578T cells. Further analysis with more ER- cell lines and patient samples is warranted to establish BP1 as a therapeutic target. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 3910.


Journal of Cancer | 2010

BP1 Homeoprotein Enhances Metastatic Potential in ER-negative Breast Cancer

Yebo Fu; Yi Lian; Kyung Soon Kim; Lei Zhang; A. Katharine Hindle; Fred Brody; Robert S. Siegel; Timothy A. McCaffrey; Sidney W. Fu

Collaboration


Dive into the Yebo Fu's collaboration.

Top Co-Authors

Avatar

Sidney W. Fu

George Washington University

View shared research outputs
Top Co-Authors

Avatar

Liang Chen

George Washington University

View shared research outputs
Top Co-Authors

Avatar

Christine B. Teal

George Washington University

View shared research outputs
Top Co-Authors

Avatar

Rachel F. Brem

George Washington University

View shared research outputs
Top Co-Authors

Avatar

Xiaohui Tan

George Washington University

View shared research outputs
Top Co-Authors

Avatar

Yan-gao Man

Armed Forces Institute of Pathology

View shared research outputs
Top Co-Authors

Avatar

Jin Peng

George Washington University

View shared research outputs
Top Co-Authors

Avatar

M. Katayoon Rezaei

George Washington University

View shared research outputs
Top Co-Authors

Avatar

Meng-Hsuan Mo

George Washington University

View shared research outputs
Top Co-Authors

Avatar

Robert S. Siegel

Washington University in St. Louis

View shared research outputs
Researchain Logo
Decentralizing Knowledge