241 results on '"Yin-yuan Mo"'
Search Results
2. COVID-19 vaccination is associated with enhanced efficacy of anti-PD-(L)1 immunotherapy in advanced NSCLC patients: a real-world study
- Author
-
Yunfei Qian, Zhuxian Zhu, Yin-Yuan Mo, and Ziqiang Zhang
- Subjects
COVID-19 vaccination ,Immune checkpoint inhibitor (ICI) ,Non-small cell lung cancer (NSCLC) ,Progression-free survival (PFS) ,Overall survival (OS) ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 ,Infectious and parasitic diseases ,RC109-216 - Abstract
Abstract Background Coronavirus disease 2019 (COVID-19) vaccine has played a major role in ending the pandemic. However, little is known about the influence of COVID-19 vaccine on the efficacy of immunotherapy in patients with non-small cell lung cancer (NSCLC). Objectives The goal of this study is to explore whether COVID-19 vaccine impacts the efficacy of immune checkpoint inhibitors (ICIs) in NSCLC patients. Methods We retrospectively analyzed the survival data of ICI-treated 104 patients with stage III–IV NSCLC, who either received COVID-19 vaccination (n = 25) or no vaccination (n = 79). The potential risk factors, in particular roles of COVID-19 vaccination in the efficacy of ICIs in these patients, were evaluated. Results Our results showed significantly improved ORR (28.0% vs. 11.39%, p = 0.05) and DCR (88.0% vs. 54.43%, p = 0.005) in the COVID-19 vaccinated group compared with the non-vaccinated group. Regarding the long-term survival benefits, COVID-19 vaccine showed profound influence both on the PFS (HR = 0.16, p = 0.021) and OS (HR = 0.168, p = 0.019) in patients with NSCLC under ICIs treatment. The PFS (p
- Published
- 2023
- Full Text
- View/download PDF
3. Microbiome and spatially resolved metabolomics analysis reveal the anticancer role of gut Akkermansia muciniphila by crosstalk with intratumoral microbiota and reprogramming tumoral metabolism in mice
- Author
-
Zhuxian Zhu, Jixu Cai, Weiwei Hou, Ke Xu, Xuxiao Wu, Yuanlin Song, Chunxue Bai, Yin-Yuan Mo, and Ziqiang Zhang
- Subjects
Gut microbiota ,akkermansia muciniphila (akk) ,intratumoral microbiome ,spatially resolved metabolomics ,metabolism reprogramming ,crosstalk ,Diseases of the digestive system. Gastroenterology ,RC799-869 - Abstract
ABSTRACTAlthough gut microbiota has been linked to cancer, little is known about the crosstalk between gut- and intratumoral-microbiomes. The goal of this study was to determine whether gut Akkermansia muciniphila (Akk) is involved in the regulation of intratumoral microbiome and metabolic contexture, leading to an anticancer effect on lung cancer. We evaluated the effects of gut endogenous or gavaged exogenous Akk on the tumorigenesis using the Lewis lung cancer mouse model. Feces, blood, and tumor tissue samples were collected for 16S rDNA sequencing. We then conducted spatially resolved metabolomics profiling to discover cancer metabolites in situ directly and to characterize the overall Akk-regulated metabolic features, followed by the correlation analysis of intratumoral bacteria with metabolic network. Our results showed that both endogenous and exogenous gavaged Akk significantly inhibited tumorigenesis. Moreover, we detected increased Akk abundance in blood circulation or tumor tissue by 16S rDNA sequencing in the Akk gavaged mice, compared with the control mice. Of great interest, gavaged Akk may migrate into tumor tissue and influence the composition of intratumoral microbiome. Spatially resolved metabolomics analysis revealed that the gut-derived Akk was able to regulate tumor metabolic pathways, from metabolites to enzymes. Finally, our study identified a significant correlation between the gut Akk-regulated intratumoral bacteria and metabolic network. Together, gut-derived Akk may migrate into blood circulation, and subsequently colonize into lung cancer tissue, which contributes to the suppression of tumorigenesis by influencing tumoral symbiotic microbiome and reprogramming tumoral metabolism, although more studies are needed.
- Published
- 2023
- Full Text
- View/download PDF
4. Circulating eosinophils associated with responsiveness to COVID-19 vaccine and the disease severity in patients with SARS-CoV-2 omicron variant infection
- Author
-
Zhuxian Zhu, Jixu Cai, Qiang Tang, Yin-yuan Mo, Tiantian Deng, Xiaoyu Zhang, Ke Xu, Beishou Wu, Haicheng Tang, and Ziqiang Zhang
- Subjects
COVID-19 vaccine ,SARS-CoV-2 ,Omicron variant ,Eosinophil (EOS) ,T cell immunity ,Disease severity ,Diseases of the respiratory system ,RC705-779 - Abstract
Abstract Objective This study aimed to investigate the longitudinal circulating eosinophil (EOS) data impacted by the COVID-19 vaccine, the predictive role of circulating EOS in the disease severity, and its association with T cell immunity in patients with SARS-CoV-2 Omicron BA.2 variant infection in Shanghai, China. Methods We collected a cohort of 1,157 patients infected with SARS-CoV-2 Omicron/BA.2 variant in Shanghai, China. These patients were diagnosed or admitted between Feb 20, 2022, and May 10, 2022, and were classified as asymptomatic (n = 705), mild (n = 286) and severe (n = 166) groups. We compiled and analyzed data of patients’ clinical demographic characteristics, laboratory findings, and clinical outcomes. Results COVID-19 vaccine reduced the incidence of severe cases. Severe patients were shown to have declined peripheral blood EOS. Both the 2 doses and 3 doses of inactivated COVID-19 vaccines promoted the circulating EOS levels. In particular, the 3rd booster shot of inactivated COVID-19 vaccine was shown to have a sustained promoting effect on circulating EOS. Univariate analysis showed that there was a significant difference in age, underlying comorbidities, EOS, lymphocytes, CRP, CD4, and CD8 T cell counts between the mild and the severe patients. Multivariate logistic regression analysis and ROC curve analysis indicate that circulating EOS (AUC = 0.828, p = 0.025), the combination of EOS and CD4 T cell (AUC = 0.920, p = 0.017) can predict the risk of disease severity in patients with SARS-CoV-2 Omicron BA.2 variant infection. Conclusions COVID-19 vaccine promotes circulating EOS and reduces the risk of severe illness, and particularly the 3rd booster dose of COVID-19 vaccine sustainedly promotes EOS. Circulating EOS, along with T cell immunity, may have a predictive value for the disease severity in SARS-CoV-2 Omicron infected patients.
- Published
- 2023
- Full Text
- View/download PDF
5. N6-methyladenosine modified LINC00901 promotes pancreatic cancer progression through IGF2BP2/MYC axis
- Author
-
Wan-Xin Peng, Fei Liu, Jia-Hong Jiang, Hang Yuan, Ziqiang Zhang, Liu Yang, and Yin-Yuan Mo
- Subjects
IGF2BP2 ,LINC00901 ,MYC ,N6-methyladenosine modification (m6A) ,PDAC ,YTHDF1 ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Accumulating evidence indicates that RNA methylation at N6-methyladenosine (m6A) plays an important regulatory role in gene expression and aberrant mRNA m6A modification is often associated with a variety of cancers. However, little is known whether and how m6A-modification impacts long non-coding RNA (lncRNA) and lncRNA-mediated tumorigenesis, particularly in pancreatic ductal adenocarcinoma (PDAC). In the present study, we report that a previously uncharacterized lncRNA, LINC00901, promotes pancreatic cancer cell growth and invasion and moreover, LINC00901 is subject to m6A modification which regulates its expression. In this regard, YTHDF1 serves as a reader for the m6A modified LINC00901 and downregulates the LINC00901 level. Notably, two conserved m6A sites in LINC00901 are critical to the recognition of LINC00901 by YTHDF1. Finally, RNA sequencing (RNA-seq) and gene function analysis revealed that LINC00901 positively regulates MYC through upregulation of IGF2BP2, a known RNA binding protein that can enhance MYC mRNA stability. Together, our results suggest that there is a LINC00901-IGF2BP2-MYC axis through which LINC00901 promotes PDAC progression in an m6A dependent manner.
- Published
- 2023
- Full Text
- View/download PDF
6. Exosomal miR-1304-3p promotes breast cancer progression in African Americans by activating cancer-associated adipocytes
- Author
-
Dan Zhao, Kerui Wu, Sambad Sharma, Fei Xing, Shih-Ying Wu, Abhishek Tyagi, Ravindra Deshpande, Ravi Singh, Martin Wabitsch, Yin-Yuan Mo, and Kounosuke Watabe
- Subjects
Science - Abstract
The molecular mechanisms explaining racial disparity in breast cancer mortality are not completely elucidated. Here, the authors show that an African-associated SNP in American breast cancer patients, leads to higher levels of microRNA miR-1304-3p which promotes cancer by increasing lipids availability.
- Published
- 2022
- Full Text
- View/download PDF
7. LncRNA IPW inhibits growth of ductal carcinoma in situ by downregulating ID2 through miR-29c
- Author
-
Ravindra Pramod Deshpande, Sambad Sharma, Yin Liu, Puspa Raj Pandey, Xinhong Pei, Kerui Wu, Shih-Ying Wu, Abhishek Tyagi, Dan Zhao, Yin-Yuan Mo, and Kounosuke Watabe
- Subjects
LncRNA IPW ,DCIS ,ID2 ,miR-29c ,Toyocamycin ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Ductal carcinoma in situ (DCIS) of breast is the noninvasive lesion that has propensity to progress to the malignant form. At present, it is still unknown which lesions can potentially progress to invasive forms. In this study, we aimed to identify key lncRNAs involved in DCIS growth. Methods We employ disease-related lncProfiler array to identify IPW in specimens of DCIS and matching control samples and validate the observations in three DCIS-non-tumorigenic cell lines. Further, we examine the mechanism of IPW action and the downstream signaling in in vitro and in vivo assays. Importantly, we screened a library containing 390 natural compounds to identify candidate compound selectively inhibiting IPW low DCIS cells. Results We identified lncRNA IPW as a novel tumor suppressor critical for inhibiting DCIS growth. Ectopic expression of IPW in DCIS cells strongly inhibited cell proliferation, colony formation and cell cycle progression while silencing IPW in primary breast cells promoted their growth. Additionally, orthotropic implantation of cells with ectopic expression of IPW exhibited decreased tumor growth in vivo. Mechanistically, IPW epigenetically enhanced miR-29c expression by promoting H3K4me3 enrichment in its promoter region. Furthermore, we identified that miR-29c negatively regulated a stemness promoting gene, ID2, and diminished self-renewal ability of DCIS cells. Importantly, we screened a library containing 390 natural compounds and identified toyocamycin as a compound that selectively inhibited the growth of DCIS with low expression of IPW, while it did not affect DCIS with high IPW expression. Toyocamycin also suppressed genes associated with self-renewal ability and inhibited DCIS growth in vivo. Conclusion Our findings revealed a critical role of the IPW-miR-29c-ID2 axis in DCIS formation and suggested potential clinical use of toyocamycin for the treatment of DCIS.
- Published
- 2022
- Full Text
- View/download PDF
8. Stabilization of UCA1 by N6-methyladenosine RNA methylation modification promotes colorectal cancer progression
- Author
-
Rong-Zhang He, Jing Jiang, Xinglin Hu, Ming Lei, Jia Li, Weihao Luo, Lili Duan, Zheng Hu, Yin-Yuan Mo, Di-Xian Luo, and Wan-Xin Peng
- Subjects
CRC ,UCA1 ,m6A modification ,IGF2BP2 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 ,Cytology ,QH573-671 - Abstract
Abstract Background UCA1 is frequently upregulated in a variety of cancers, including CRC, and it can play an oncogenic role by various mechanisms. However, how UCA1 is regulated in cancer is largely unknown. In this study, we aimed to determine whether RNA methylation at N6-methyladenosine (m6A) can impact UCA1 expression in colorectal cancer (CRC). Methods qRT-PCR was performed to detect the level of UCA1 and IGF2BP2 in CRC samples. CRISPR/Cas9 was employed to knockout (KO) UCA1, METTL3 and WTAP in DLD-1 and HCT-116 cells, while rescue experiments were carried out to re-express METTL3 and WTAP in KO cells. Immunoprecipitation using m6A antibody was performed to determine the m6A modification of UCA1. In vivo pulldown assays using S1m tagging combined with site-direct mutagenesis was carried out to confirm the recognition of m6A-modified UCA1 by IGF2BP2. Cell viability was measured by MTT and colony formation assays. The expression of UCA1 and IGF2BP2 in TCGA CRC database was obtained from GEPIA ( http://gepia.cancer-pku.cn ). Results Our results revealed that IGF2BP2 serves as a reader for m6A modified UCA1 and that adenosine at 1038 of UCA1 is critical to the recognition by IGF2BP2. Importantly, we showed that m6A writers, METTL3 and WTAP positively regulate UCA1 expression. Mechanically, IGF2BP2 increases the stability of m6A-modified UCA1. Clinically, IGF2BP2 is upregulated in CRC tissues compared with normal tissues. Conclusion These results suggest that m6A modification is an important factor contributing to upregulation of UCA1 in CRC tissues.
- Published
- 2021
- Full Text
- View/download PDF
9. Lnc-DC promotes estrogen independent growth and tamoxifen resistance in breast cancer
- Author
-
Wan-Xin Peng, Pratirodh Koirala, Huaixiang Zhou, Jiahong Jiang, Ziqiang Zhang, Liu Yang, and Yin-Yuan Mo
- Subjects
Cytology ,QH573-671 - Abstract
Abstract Selective estrogen receptor modulators (SERMs) such as tamoxifen have proven to be effective in the treatment of estrogen receptor (ER) positive breast cancer. However, a major obstacle for such endocrine therapy is estrogen independent growth, leading to resistance, and the underlying mechanism is not fully understood. The purpose of this study was to determine whether long non-coding RNAs (lncRNAs) are involved in regulation of estrogen independent growth and tamoxifen resistance in ER positive breast cancer. Using a CRISPR/Cas9-based SAM (synergistic activation mediator) library against a focus group of lncRNAs, we identify Lnc-DC as a candidate lncRNA. Further analysis suggests that Lnc-DC is able to reduce tamoxifen-induced apoptosis by upregulation of anti-apoptotic genes such as Bcl2 and Bcl-xL. Furthermore, Lnc-DC activates STAT3 by phosphorylation (pSTAT3Y705), and the activated STAT3 subsequently induces expression of cytokines which in turn activate STAT3, forming an autocrine loop. Clinically, upregulation of Lnc-DC is associated with poor prognosis. In particular, analysis of a tamoxifen-treated patient cohort indicates that Lnc-DC expression can predict the response to tamoxifen. Together, this study demonstrates a previously uncharacterized function of Lnc-DC/STAT3/cytokine axis in estrogen independent growth and tamoxifen resistance, and Lnc-DC may serve as a potential predictor for tamoxifen response.
- Published
- 2021
- Full Text
- View/download PDF
10. Exosomal miR-19a and IBSP cooperate to induce osteolytic bone metastasis of estrogen receptor-positive breast cancer
- Author
-
Kerui Wu, Jiamei Feng, Feng Lyu, Fei Xing, Sambad Sharma, Yin Liu, Shih-Ying Wu, Dan Zhao, Abhishek Tyagi, Ravindra Pramod Deshpande, Xinhong Pei, Marco Gabril Ruiz, Hiroyuki Takahashi, Shunsuke Tsuzuki, Takahiro Kimura, Yin-yuan Mo, Yusuke Shiozawa, Ravi Singh, and Kounosuke Watabe
- Subjects
Science - Abstract
Bone metastasis is a major complication of breast cancer (BC) and ER+ tumors have a higher incidence of bone metastasis than ER− tumors. Here, the authors report that miR‐19a in exosomes and the bone matrix protein, IBSP, are upregulated and secreted by bone tropic ER+ BC cells, where they cooperatively induce osteoclastogenesis and promote bone colonization.
- Published
- 2021
- Full Text
- View/download PDF
11. Connecting N6‐methyladenosine modification to ferroptosis resistance in hepatoblastoma
- Author
-
Wan‐Xin Peng and Yin‐Yuan Mo
- Subjects
Medicine (General) ,R5-920 - Published
- 2022
- Full Text
- View/download PDF
12. Publisher Correction: Exosomal miR-1304-3p promotes breast cancer progression in African Americans by activating cancer-associated adipocytes
- Author
-
Dan Zhao, Kerui Wu, Sambad Sharma, Fei Xing, Shih-Ying Wu, Abhishek Tyagi, Ravindra Deshpande, Ravi Singh, Martin Wabitsch, Yin-Yuan Mo, and Kounosuke Watabe
- Subjects
Science - Published
- 2023
- Full Text
- View/download PDF
13. Emerging roles of lncRNAs in the post-transcriptional regulation in cancer
- Author
-
Rong-Zhang He, Di-Xian Luo, and Yin-Yuan Mo
- Subjects
Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Accumulating evidence indicates that long non-coding RNAs (lncRNAs) can play a pivotal role in regulation of diverse cellular processes. In particular, lncRNAs can serve as master gene regulators at transcriptional and posttranscriptional levels, leading to tumorigenesis. In this review, we discuss latest developments in lncRNA-meditated gene expression at the post-transcriptional level, including gene splicing, mRNA stability, protein stability and nuclear trafficking. Keywords: Alternative splicing, LncRNA, Posttranscriptional regulation, Protein stability, RNA binding proteins, RNA stability
- Published
- 2019
- Full Text
- View/download PDF
14. TADKB: Family classification and a knowledge base of topologically associating domains
- Author
-
Tong Liu, Jacob Porter, Chenguang Zhao, Hao Zhu, Nan Wang, Zheng Sun, Yin-Yuan Mo, and Zheng Wang
- Subjects
Topologically associating domains ,TADs ,Family classification ,Single-cell 3D genome structures ,Long non-coding RNAs ,lncRNAs ,Biotechnology ,TP248.13-248.65 ,Genetics ,QH426-470 - Abstract
Abstract Background Topologically associating domains (TADs) are considered the structural and functional units of the genome. However, there is a lack of an integrated resource for TADs in the literature where researchers can obtain family classifications and detailed information about TADs. Results We built an online knowledge base TADKB integrating knowledge for TADs in eleven cell types of human and mouse. For each TAD, TADKB provides the predicted three-dimensional (3D) structures of chromosomes and TADs, and detailed annotations about the protein-coding genes and long non-coding RNAs (lncRNAs) existent in each TAD. Besides the 3D chromosomal structures inferred by population Hi-C, the single-cell haplotype-resolved chromosomal 3D structures of 17 GM12878 cells are also integrated in TADKB. A user can submit query gene/lncRNA ID/sequence to search for the TAD(s) that contain(s) the query gene or lncRNA. We also classified TADs into families. To achieve that, we used the TM-scores between reconstructed 3D structures of TADs as structural similarities and the Pearson’s correlation coefficients between the fold enrichment of chromatin states as functional similarities. All of the TADs in one cell type were clustered based on structural and functional similarities respectively using the spectral clustering algorithm with various predefined numbers of clusters. We have compared the overlapping TADs from structural and functional clusters and found that most of the TADs in the functional clusters with depleted chromatin states are clustered into one or two structural clusters. This novel finding indicates a connection between the 3D structures of TADs and their DNA functions in terms of chromatin states. Conclusion TADKB is available at http://dna.cs.miami.edu/TADKB/.
- Published
- 2019
- Full Text
- View/download PDF
15. Gut microbiota regulate tumor metastasis via circRNA/miRNA networks
- Author
-
Zhuxian Zhu, Jianguo Huang, Xu Li, Jun Xing, Qiang Chen, Ruilin Liu, Feng Hua, Zhongmin Qiu, Yuanlin Song, Chunxue Bai, Yin-Yuan Mo, and Ziqiang Zhang
- Subjects
gut microbiota ,cancer ,metastasis ,circular rna (circrna) ,microrna ,interleukin-11(il-11) ,cancer stem cell ,Diseases of the digestive system. Gastroenterology ,RC799-869 - Abstract
Background Increasing evidence indicates that gut microbiota plays an important role in cancer progression. However, the underlying mechanism remains largely unknown. Here, we report that broad-spectrum antibiotics (ABX) treatment leads to enhanced metastasis by the alteration of gut microbiome composition. Methods Cancer LLC and B16-F10 cell metastasis mouse models, and microarray/RNA sequencing analysis were used to reveal the regulatory functions of microbiota-mediated circular RNA (circRNA)/microRNA (miRNA) networks that may contribute to cancer metastasis. Results The specific pathogen-free (SPF) mice with ABX treatment demonstrated enhanced lung metastasis. Fecal microbiota transplantation (FMT) from SPF mice or Bifidobacterium into germ-free mice significantly suppressed lung metastasis. Mechanistically, gut microbiota impacts circRNA expression to regulate levels of corresponding miRNAs. Specifically, such modulations of gut microbiota inhibit mmu_circ_0000730 expression in an IL-11-dependent manner. Bioinformatics analysis combined with luciferase reporter assays revealed reciprocal repression between mmu_circ_0000730 and mmu-miR-466i-3p. We further showed that both mmu-miR-466i-3p and mmu-miR-466 f-3p suppresses a number of genes involved in epithelial-mesenchymal transition (EMT) and stemness of cancer stem cells such as SOX9. Conclusions These results provide evidence of a previously unrecognized regulatory role of non-coding RNAs in microbiota-mediated cancer metastasis, and thus, the microbiome may serve as a therapeutic target.
- Published
- 2020
- Full Text
- View/download PDF
16. Comprehensive Network Analysis Reveals Alternative Splicing-Related lncRNAs in Hepatocellular Carcinoma
- Author
-
Junqing Wang, Xiuquan Wang, Akshay Bhat, Yixin Chen, Keli Xu, Yin-yuan Mo, Song Stephen Yi, and Yunyun Zhou
- Subjects
long non-coding RNAs (lncRNA) ,alternative splicing ,multi-graphic random walk ,gene-regulatory network analysis ,random walk ,hepatocellular carcinoma ,Genetics ,QH426-470 - Abstract
It is increasingly appreciated that long non-coding RNAs (lncRNAs) associated with alternative splicing (AS) could be involved in aggressive hepatocellular carcinoma. Although many recent studies show the alteration of RNA alternative splicing by deregulated lncRNAs in cancer, the extent to which and how lncRNAs impact alternative splicing at the genome scale remains largely elusive. We analyzed RNA-seq data obtained from 369 hepatocellular carcinomas (HCCs) and 160 normal liver tissues, quantified 198,619 isoform transcripts, and identified a total of 1,375 significant AS events in liver cancer. In order to predict novel AS-associated lncRNAs, we performed an integration of co-expression, protein-protein interaction (PPI) and epigenetic interaction networks that links lncRNA modulators (such as splicing factors, transcript factors, and miRNAs) along with their targeted AS genes in HCC. We developed a random walk-based multi-graphic (RWMG) model algorithm that prioritizes functional lncRNAs with their associated AS targets to computationally model the heterogeneous networks in HCC. RWMG shows a good performance evaluated by the ROC curve based on cross-validation and bootstrapping strategies. As a conclusion, our robust network-based framework has derived 31 AS-related lncRNAs that not only validates known cancer-associated cases MALAT1 and HOXA11-AS, but also reveals new players such as DNM1P35 and DLX6-AS1with potential functional implications. Survival analysis further provides insights into the clinical significance of identified lncRNAs.
- Published
- 2020
- Full Text
- View/download PDF
17. DGM-CM6: A New Model to Predict Distant Recurrence Risk in Operable Endocrine-Responsive Breast Cancer
- Author
-
Lei Lei, Xiao-Jia Wang, Yin-Yuan Mo, Skye Hung-Chun Cheng, and Yunyun Zhou
- Subjects
clinical-genomic model ,breast cancer ,distant recurrence ,prognosis ,endocrine-responsive ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
To investigate the prognostic value of DGM-CM6 (Distant Genetic Model-Clinical variable Model 6) for endocrine-responsive breast cancer (ERBC) patients, we analyzed 752 operable breast cancer patients treated in a Taiwan cancer center from 2005 to 2014. Among them, 490 ERBC patients (identified by the PAM50 or immunohistochemistry method) were classified by DGM-CM6 into low- and high-risk groups (cutoff
- Published
- 2020
- Full Text
- View/download PDF
18. Linc-RoR promotes MAPK/ERK signaling and confers estrogen-independent growth of breast cancer
- Author
-
Wan-xin Peng, Jian-guo Huang, Liu Yang, Ai-hua Gong, and Yin-Yuan Mo
- Subjects
Breast cancer ,Estrogen-independent growth ,Linc-RoR ,ERK ,DUSP7 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background The conversion from estrogen-dependent to estrogen-independent state of ER+ breast cancer cells is the key step to promote resistance to endocrine therapies. Although the crucial role of MAPK/ERK signaling pathway in estrogen-independent breast cancer cell growth is well established, the underlying mechanism is not fully understood. Methods In this study, we profiled lncRNA expression against a focused group of lncRNAs selected from lncRNA database. CRISPR/Cas9 was employed to knockout (KO) linc-RoR in MCF-7 cells, while rescue experiments were carried out to re-express linc-RoR in KO cells. Colony formation and MTT assays were used to examine the role of linc-RoR in estrogen-independent growth and tamoxifen resistance. Western blot and qRT-PCR were used to determine the change of protein and lncRNA levels, respectively. The expression of DUSP7 in clinical specimens was downloaded from Oncomine ( www.oncomine.org ) and the dataset from Kaplan-Meier Plotter ( http://kmplot.com ) was used to analyze the clinical outcomes in relation to DUSP7. Results We identified that linc-RoR functions as an onco-lncRNA to promote estrogen-independent growth of ER+ breast cancer. Under estrogen deprivation, linc-RoR causes the upregulation of phosphorylated MAPK/ERK pathway which in turn activates ER signaling. Knockout of linc-RoR abrogates estrogen deprivation-induced ERK activation as well as ER phosphorylation, whereas re-expression of linc-RoR restores all above phenotypes. Moreover, we show that the ERK-specific phosphatase Dual Specificity Phosphatase 7 (DUSP7), also known as MKP-X, is involved in linc-RoR KO-induced repression of MAPK/ERK signaling. Interestingly, linc-RoR KO increases the protein stability of DUSP7, resulting in repression of ERK phosphorylation. Clinical data analysis reveal that DUSP7 expression is lower in ER+ breast cancer samples than that in ER- breast cancer. Moreover, downregulation of DUSP7 expression is associated with poor patient survival. Conclusion Taken together, these results suggest that linc-RoR promotes estrogen-independent growth and activation of MAPK/ERK pathway of breast cancer cells by regulating the ERK-specific phosphatase DUSP7. Thus, this study might help not only in establishing a role for linc-RoR in estrogen-independent and tamoxifen resistance of ER+ breast cancer, but also suggesting a link between linc-RoR and MAPK/ERK pathway.
- Published
- 2017
- Full Text
- View/download PDF
19. LncRNA AK023948 is a positive regulator of AKT
- Author
-
Pratirodh Koirala, Jianguo Huang, Tsui-Ting Ho, Fangting Wu, Xianfeng Ding, and Yin-Yuan Mo
- Subjects
Science - Abstract
The function of many human long non-coding RNAs (lncRNAs) is still undetermined. Here, the authors setup a gain of function CRISPR-based screen and identify a lncRNA that positively regulates AKT activity by interacting with the RNA helicase DHX9 resulting in stabilization of PI3K regulatory subunit p85.
- Published
- 2017
- Full Text
- View/download PDF
20. Data in support of transcriptional regulation and function of Fas-antisense long noncoding RNA during human erythropoiesis
- Author
-
Olga Villamizar, Christopher B. Chambers, Yin-Yuan Mo, Donald S. Torry, Reese Hofstrand, Janice M. Riberdy, Derek A. Persons, and Andrew Wilber
- Subjects
Computer applications to medicine. Medical informatics ,R858-859.7 ,Science (General) ,Q1-390 - Abstract
This paper describes data related to a research article titled, “Fas-antisense long noncoding RNA is differentially expressed during maturation of human erythrocytes and confers resistance to Fas-mediated cell death” [1]. Long noncoding RNAs (lncRNAs) are increasingly appreciated for their capacity to regulate many steps of gene expression. While recent studies suggest that many lncRNAs are functional, the scope of their actions throughout human biology is largely undefined including human red blood cell development (erythropoiesis). Here we include expression data for 82 lncRNAs during early, intermediate and late stages of human erythropoiesis using a commercial qPCR Array. From these data, we identified lncRNA Fas-antisense 1 (Fas-AS1 or Saf) described in the research article. Also included are 5′ untranslated sequences (UTR) for lncRNA Saf with transcription factor target sequences identified. Quantitative RT-PCR data demonstrate relative levels of critical erythroid transcription factors, GATA-1 and KLF1, in K562 human erythroleukemia cells and maturing erythroblasts derived from human CD34+ cells. End point and quantitative RT-PCR data for cDNA prepared using random hexamers versus oligo(dT)18 revealed that lncRNA Saf is not effectively polyadenylated. Finally, we include flow cytometry histograms demonstrating Fas levels on maturing erythroblasts derived from human CD34+ cells transduced using mock conditions or with lentivirus particles encoding for Saf. Keywords: Erythropoiesis, Long noncoding RNA, Saf, Apoptosis, Fas
- Published
- 2016
- Full Text
- View/download PDF
21. LncRNA HOTAIR Enhances the Androgen-Receptor-Mediated Transcriptional Program and Drives Castration-Resistant Prostate Cancer
- Author
-
Ali Zhang, Jonathan C. Zhao, Jung Kim, Ka-wing Fong, Yeqing Angela Yang, Debabrata Chakravarti, Yin-Yuan Mo, and Jindan Yu
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Understanding the mechanisms of androgen receptor (AR) activation in the milieu of low androgen is critical to effective treatment of castration-resistant prostate cancer (CRPC). Here, we report HOTAIR as an androgen-repressed lncRNA, and, as such, it is markedly upregulated following androgen deprivation therapies and in CRPC. We further demonstrate a distinct mode of lncRNA-mediated gene regulation, wherein HOTAIR binds to the AR protein to block its interaction with the E3 ubiquitin ligase MDM2, thereby preventing AR ubiquitination and protein degradation. Consequently, HOTAIR expression is sufficient to induce androgen-independent AR activation and drive the AR-mediated transcriptional program in the absence of androgen. Functionally, HOTAIR overexpression increases, whereas HOTAIR knockdown decreases, prostate cancer cell growth and invasion. Taken together, our results provide compelling evidence of lncRNAs as drivers of androgen-independent AR activity and CRPC progression, and they support the potential of lncRNAs as therapeutic targets.
- Published
- 2015
- Full Text
- View/download PDF
22. Mesenchymal Stem/Stromal Cells under Stress Increase Osteosarcoma Migration and Apoptosis Resistance via Extracellular Vesicle Mediated Communication.
- Author
-
Krishna C Vallabhaneni, Meeves-Yoni Hassler, Anu Abraham, Jason Whitt, Yin-Yuan Mo, Azeddine Atfi, and Radhika Pochampally
- Subjects
Medicine ,Science - Abstract
Studies have shown that mesenchymal stem/stromal cells (MSCs) from bone marrow are involved in the growth and metastasis of solid tumors but the mechanism remains unclear in osteosarcoma (OS). Previous studies have raised the possibility that OS cells may receive support from associated MSCs in the nutrient deprived core of the tumors through the release of supportive macromolecules and growth factors either in vesicular or non-vesicular forms. In the present study, we used stressed mesenchymal stem cells (SD-MSCs), control MSCs and OS cells to examine the hypothesis that tumor-associated MSCs in nutrient deprived core provide pro-proliferative, anti-apoptotic, and metastatic support to nearby tumor cells. Assays to study of the effects of SD-MSC conditioned media revealed that OS cells maintained proliferation when compared to OS cells grown under serum-starved conditions alone. Furthermore, OS cells in MSCs and SD-MSC conditioned media were significantly resistant to apoptosis and an increased wound healing rate was observed in cells exposed to either conditioned media or EVs from MSCs and SD-MSCs. RT-PCR assays of OS cells incubated with extracellular vesicles (EVs) from SD-MSCs revealed microRNAs that could potentially target metabolism and metastasis associated genes as predicted by in silico algorithms, including monocarboxylate transporters, bone morphogenic receptor type 2, fibroblast growth factor 7, matrix metalloproteinase-1, and focal adhesion kinase-1. Changes in the expression levels of focal adhesion kinase, STK11 were confirmed by quantitative PCR assays. Together, these data indicate a tumor supportive role of MSCs in osteosarcoma growth that is strongly associated with the miRNA content of the EVs released from MSCs under conditions that mimic the nutrient deprived core of solid tumors.
- Published
- 2016
- Full Text
- View/download PDF
23. Alternative approach to generate shRNA from cDNA
- Author
-
Anh Dinh and Yin-Yuan Mo
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Short hairpin RNA (shRNA) synthesized from vector-based expression is as effective as short interfering RNA (siRNA) synthesized in vitro for suppressing the expression of their corresponding genes. Recently, three groups independently reported a new technology to construct an shRNA library from cDNA, providing great hope for genome-wide functional screens in many biological systems. In the present study, we report an alternative approach to generate shRNA from cDNA. A major improvement was to use a nicking enzyme to open up the double-stranded DNA so that the loop region remains single-stranded while the rest of the DNA fragment is double-stranded at an elevated temperature (e.g., 72°C). The single-stranded DNA was then converted into double-stranded DNA by Taq DNA polymerase using the existing strand in the double-stranded region as a primer. Thus, the extended product carried a palindromic structure of 19 bp separated by a loop. Finally, the DNA fragment was cloned into a vector that carries an H1 promoter at the upstream region and ends with 5Ts, a terminator for the Pol III polymerase, at the downstream region. To prove the principle, we constructed shRNA from green fluorescent protein (GFP) cDNA and successfully suppressed GFP expression. Consequently, this simplified approach provides a better alternative to generate shRNA libraries from cDNA. Such shRNA libraries can be used to identify potential siRNA target sequences and study gene functions by a variety of selection methods.
- Published
- 2005
- Full Text
- View/download PDF
24. Overexpression of Human DNA Topoisomerase IIα by Fusion to Enhanced Green Fluorescent Protein
- Author
-
Yin-Yuan Mo, Keith A. Ameiss, and William T. Beck
- Subjects
Biology (General) ,QH301-705.5 - Abstract
DNA topoisomerase (topo) IIα is a major target for many anticancer agents. However, progress towards understanding how these agents interact with this enzyme in human cells and how resistance to these agents arises is greatly impeded by difficulties in expressing this gene. Here, we report on achieving a high level of expression of a full-length human topo IIα gene in human cells. We started with the topo IIα cDNA driven by a strong cytomegalovirus promoter and transiently transfected HeLa cells. Although topo IIα mRNA was consistently detected in transfected cells, no exogenous topo IIα protein was detected. By contrast, when the same cDNA was fused to an enhanced green fluorescent protein (EGFP), we detected a high level of expression at both mRNA and protein levels. The exogenous topo IIα was localized to cell nuclei as expected, indicating that the fusion protein is properly folded. Furthermore, overexpression of the EGFP-topo IIα fusion protein increased the sensitivity of the transfected cells to teniposide, suggesting that it functions as the endogenous counterpart. Thus, in addition to being used as a gene tag, the GFP fusion approach may be generally applicable for expressing genes, such as topo IIα, that are difficult to express by conventional methods.
- Published
- 1998
- Full Text
- View/download PDF
25. Targeting DNA-PKcs and ATM with miR-101 sensitizes tumors to radiation.
- Author
-
Dan Yan, Wooi Loon Ng, Xiangming Zhang, Ping Wang, Zhaobin Zhang, Yin-Yuan Mo, Hui Mao, Chunhai Hao, Jeffrey J Olson, Walter J Curran, and Ya Wang
- Subjects
Medicine ,Science - Abstract
BackgroundRadiotherapy kills tumor-cells by inducing DNA double strand breaks (DSBs). However, the efficient repair of tumors frequently prevents successful treatment. Therefore, identifying new practical sensitizers is an essential step towards successful radiotherapy. In this study, we tested the new hypothesis: identifying the miRNAs to target DNA DSB repair genes could be a new way for sensitizing tumors to ionizing radiation.Principal findingsHERE, WE CHOSE TWO GENES: DNA-PKcs (an essential factor for non-homologous end-joining repair) and ATM (an important checkpoint regulator for promoting homologous recombination repair) as the targets to search their regulating miRNAs. By combining the database search and the bench work, we picked out miR-101. We identified that miR-101 could efficiently target DNA-PKcs and ATM via binding to the 3'- UTR of DNA-PKcs or ATM mRNA. Up-regulating miR-101 efficiently reduced the protein levels of DNA-PKcs and ATM in these tumor cells and most importantly, sensitized the tumor cells to radiation in vitro and in vivo.ConclusionsThese data demonstrate for the first time that miRNAs could be used to target DNA repair genes and thus sensitize tumors to radiation. These results provide a new way for improving tumor radiotherapy.
- Published
- 2010
- Full Text
- View/download PDF
26. Novel gene selection method for breast cancer intrinsic subtypes from two large cohort study.
- Author
-
Silu Zhang, Yin-yuan Mo, Torumoy Ghoshal, Dawn Wilkins, Yixin Chen 0002, and Yunyun Zhou
- Published
- 2017
- Full Text
- View/download PDF
27. Data from Nimbolide, a Limonoid Triterpene, Inhibits Growth of Human Colorectal Cancer Xenografts by Suppressing the Proinflammatory Microenvironment
- Author
-
Bharat B. Aggarwal, Yin-Yuan Mo, Mangalam S. Nair, Dhanya R. Sethumadhavan, Sahdeo Prasad, and Subash C. Gupta
- Abstract
Purpose: Extensive research over the past decade has revealed that the proinflammatory microenvironment plays a critical role in the development of colorectal cancer. Whether nimbolide, a limonoid triterpene, can inhibit the growth of colorectal cancer was investigated in the present study.Experimental Design: The effect of nimbolide on proliferation of colorectal cancer cell lines was examined by MTT assay, apoptosis by caspase activation and poly-ADP ribose polymerase cleavage, NF-κB activation by DNA-binding assay, and protein expression by Western blotting. The effect of nimbolide on the tumor growth in vivo was examined in colorectal cancer xenografts in a nude mouse model.Results: Nimbolide inhibited proliferation, induced apoptosis, and suppressed NF-κB activation and NF-κB–regulated tumorigenic proteins in colorectal cancer cells. The suppression of NF-κB activation by nimbolide was caused by sequential inhibition of IκB kinase (IKK) activation, IκBα phosphorylation, and p65 nuclear translocation. Furthermore, the effect of nimbolide on IKK activity was found to be direct. In vivo, nimbolide (at 5 and 20 mg/kg body weight), injected intraperitoneally after tumor inoculation, significantly decreased the volume of colorectal cancer xenografts. The limonoid-treated xenografts exhibited significant downregulation in the expression of proteins involved in tumor cell survival (Bcl-2, Bcl-xL, c-IAP-1, survivin, and Mcl-1), proliferation (c-Myc and cyclin D1), invasion (MMP-9, ICAM-1), metastasis (CXCR4), and angiogenesis (VEGF). The limonoid was found to be bioavailable in the blood plasma and tumor tissues of treated mice.Conclusions: Our studies provide evidence that nimbolide can suppress the growth of human colorectal cancer through modulation of the proinflammatory microenvironment. Clin Cancer Res; 19(16); 4465–76. ©2013 AACR.
- Published
- 2023
28. Supplementary Figure 1 from Nimbolide, a Limonoid Triterpene, Inhibits Growth of Human Colorectal Cancer Xenografts by Suppressing the Proinflammatory Microenvironment
- Author
-
Bharat B. Aggarwal, Yin-Yuan Mo, Mangalam S. Nair, Dhanya R. Sethumadhavan, Sahdeo Prasad, and Subash C. Gupta
- Abstract
PDF file - 130K, Nimbolide is bioavailable in the plasma and colorectal tumor tissues of nude mice. A, HPLC chromatogram of pure nimbolide. B, HPLC chromatogram of nimbolide in plasma. C, HPLC chromatogram of nimbolide in tumor tissues.
- Published
- 2023
29. Figure S1-S7 from Loss of XIST in Breast Cancer Activates MSN-c-Met and Reprograms Microglia via Exosomal miRNA to Promote Brain Metastasis
- Author
-
Kounosuke Watabe, Ralph B D'Agostino, Linda J. Metheny-Barlow, Hui-Wen Lo, Boris C. Pasche, Waldemar Debinski, Jimmy Ruiz, Michael D. Chan, Abhishek Tyagi, Pierre-Alexandre Vidi, Ravi Singh, Guangxu Jin, Stephanie Sanders, Jiamei Feng, Yin-Yuan Mo, Sambad Sharma, Kerui Wu, Shih-Ying Wu, Yin Liu, and Fei Xing
- Abstract
The supplementary infomation containing Figure legends, seven supplemental figures and one supplemental table. Figure S1 XIST expression is preferentially correlated with brain metastasis in breast cancer. Figure S2 Knockout of XIST preferentially promotes brain metastases. Figure S3 Knockout of XIST enhances tumor growth and distant metastases. Figure S4 Knockdown of XIST promotes EMT and stemness. Figure S5 c-Met is regulated by X-chromosome gene, MSN. Figure S6 miR-503 promotes the conversion of M1 to M2 phenotype of microglia. Figure S7 Fludarabine selectively suppresses the growth of XISTlow cells. Table S1 reagent and resource.
- Published
- 2023
30. Supplementary Data from Epigenetic and Posttranscriptional Modulation of SOS1 Can Promote Breast Cancer Metastasis through Obesity-Activated c-Met Signaling in African-American Women
- Author
-
Kounosuke Watabe, Yin-Yuan Mo, Chinni Yalamanchili, Amar G. Chittiboyina, Lance D. Miller, Jacob Cleary, Yuezhu Wang, Ravindra Deshpande, Yin Liu, Sambad Sharma, Kerui Wu, Abhishek Tyagi, Shih-Ying Wu, Dan Zhao, and Fei Xing
- Abstract
supplementary figure legend
- Published
- 2023
31. Data from Epigenetic and Posttranscriptional Modulation of SOS1 Can Promote Breast Cancer Metastasis through Obesity-Activated c-Met Signaling in African-American Women
- Author
-
Kounosuke Watabe, Yin-Yuan Mo, Chinni Yalamanchili, Amar G. Chittiboyina, Lance D. Miller, Jacob Cleary, Yuezhu Wang, Ravindra Deshpande, Yin Liu, Sambad Sharma, Kerui Wu, Abhishek Tyagi, Shih-Ying Wu, Dan Zhao, and Fei Xing
- Abstract
Ethnicity is considered to be one of the major risk factors in certain subtypes of breast cancer. However, the mechanism of this racial disparity remains poorly understood. Here, we demonstrate that SOS1, a key regulator of Ras pathway, is highly expressed in African-American (AA) patients with breast cancer compared with Caucasian-American patients. Because of the higher obesity rate in AA women, increased levels of SOS1 facilitated signal transduction of the c-Met pathway, which was highly activated in AA patients with breast cancer via hepatocyte growth factor secreted from adipocytes. Elevated expression of SOS1 also enhanced cancer stemness through upregulation of PTTG1 and promoted M2 polarization of macrophages by CCL2 in metastatic sites. SOS1 was epigenetically regulated by a super-enhancer identified by H3K27ac in AA patients. Knockout of the super-enhancer by CRISPR in AA cell lines significantly reduced SOS1 expression. Furthermore, SOS1 was posttranscriptionally regulated by miR-483 whose expression is reduced in AA patients through histone trimethylation (H3K27me3) on its promoter. The natural compound, taxifolin, suppressed signaling transduction of SOS1 by blocking the interaction between SOS1 and Grb2, suggesting a potential utility of this compound as a therapeutic agent for AA patients with breast cancer.Significance:These findings elucidate the signaling network of SOS1-mediated metastasis in African-American patients, from the epigenetic upregulation of SOS1 to the identification of taxifolin as a potential therapeutic strategy against SOS1-driven tumor progression.
- Published
- 2023
32. Data from Loss of XIST in Breast Cancer Activates MSN-c-Met and Reprograms Microglia via Exosomal miRNA to Promote Brain Metastasis
- Author
-
Kounosuke Watabe, Ralph B D'Agostino, Linda J. Metheny-Barlow, Hui-Wen Lo, Boris C. Pasche, Waldemar Debinski, Jimmy Ruiz, Michael D. Chan, Abhishek Tyagi, Pierre-Alexandre Vidi, Ravi Singh, Guangxu Jin, Stephanie Sanders, Jiamei Feng, Yin-Yuan Mo, Sambad Sharma, Kerui Wu, Shih-Ying Wu, Yin Liu, and Fei Xing
- Abstract
Up to 30% of patients with metastatic breast cancer eventually develop brain metastasis, yet the pathologic mechanism behind this development remains poorly understood. Here, we profiled long noncoding RNAs in brain metastatic tumors from patients with breast cancer and found that the X-inactive–specific transcript (XIST) was significantly downregulated in these tissues. XIST expression levels inversely correlated with brain metastasis, but not with bone metastasis in patients. Silencing of XIST preferentially promoted brain metastatic growth of XISThigh cells in our xenograft models. Moreover, knockout of XIST in mice mammary glands accelerated primary tumor growth as well as metastases in the brain. Decreased expression of XIST stimulated epithelial–mesenchymal transition and activated c-Met via MSN-mediated protein stabilization, which resulted in the promotion of stemness in the tumor cells. Loss of XIST also augmented secretion of exosomal miRNA-503, which triggered M1–M2 polarization of microglia. This M1–M2 conversion upregulated immune suppressive cytokines in microglia that suppressed T-cell proliferation. Furthermore, we screened an FDA-approved drug library and identified fludarabine as a synthetic lethal drug for XISTlow breast tumor cells and found that fludarabine blocked brain metastasis in our animal model. Our results indicate that XIST plays a critical role in brain metastasis in breast cancer by affecting both tumor cells and the tumor microenvironment and that the XIST-mediated pathway may serve as an effective target for treating brain metastasis.Significance: These findings describe mechanisms of how loss of the lncRNA XIST promotes brain metastasis in breast cancer and identify fludarabine as a potential therapeutic agent that specifically eliminates XISTlow tumor cells in the brain. Cancer Res; 78(15); 4316–30. ©2018 AACR.
- Published
- 2023
33. Supplementary Legends 1-6 from MicroRNA-145 Suppresses Cell Invasion and Metastasis by Directly Targeting Mucin 1
- Author
-
Yin-Yuan Mo and Mohit Sachdeva
- Abstract
Supplementary Legends 1-6 from MicroRNA-145 Suppresses Cell Invasion and Metastasis by Directly Targeting Mucin 1
- Published
- 2023
34. Supplementary Figures 1-6, Table 1 from MicroRNA-145 Suppresses Cell Invasion and Metastasis by Directly Targeting Mucin 1
- Author
-
Yin-Yuan Mo and Mohit Sachdeva
- Abstract
Supplementary Figures 1-6, Table 1 from MicroRNA-145 Suppresses Cell Invasion and Metastasis by Directly Targeting Mucin 1
- Published
- 2023
35. RETRACTED: IncRNA RMST Enhances DNMT3 Expression through Interaction with HuR
- Author
-
Wan-Xin Peng, Pratirodh Koirala, Wei Zhang, Chao Ni, Zheng Wang, Liu Yang, and Yin-Yuan Mo
- Subjects
Pharmacology ,Drug Discovery ,Genetics ,Molecular Medicine ,Molecular Biology - Published
- 2023
36. Stabilization of UCA1 by N6-methyladenosine RNA methylation modification promotes colorectal cancer progression
- Author
-
Yin-Yuan Mo, Zheng Hu, Lili Duan, Rong-Zhang He, Weihao Luo, Jia Li, Jing Jiang, Ming Lei, Dixian Luo, Xinglin Hu, and Wanxin Peng
- Subjects
Cancer Research ,Colorectal cancer ,Immunoprecipitation ,RNA methylation ,Biology ,chemistry.chemical_compound ,m6A modification ,Downregulation and upregulation ,In vivo ,Genetics ,medicine ,Viability assay ,UCA1 ,RC254-282 ,IGF2BP2 ,QH573-671 ,Cancer ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,CRC ,Oncology ,chemistry ,Cancer research ,N6-Methyladenosine ,Primary Research ,Cytology - Abstract
BackgroundUCA1 is frequently upregulated in a variety of cancers, including CRC, and it can play an oncogenic role by various mechanisms. However, how UCA1 is regulated in cancer is largely unknown. In this study, we aimed to determine whether RNA methylation at N6-methyladenosine (m6A) can impact UCA1 expression in colorectal cancer (CRC).MethodsqRT-PCR was performed to detect the level of UCA1 and IGF2BP2 in CRC samples. CRISPR/Cas9 was employed to knockout (KO) UCA1, METTL3 and WTAP in DLD-1 and HCT-116 cells, while rescue experiments were carried out to re-express METTL3 and WTAP in KO cells. Immunoprecipitation using m6A antibody was performed to determine the m6A modification of UCA1. In vivo pulldown assays using S1m tagging combined with site-direct mutagenesis was carried out to confirm the recognition of m6A-modified UCA1 by IGF2BP2. Cell viability was measured by MTT and colony formation assays. The expression of UCA1 and IGF2BP2 in TCGA CRC database was obtained from GEPIA (http://gepia.cancer-pku.cn).ResultsOur results revealed that IGF2BP2 serves as a reader for m6A modified UCA1 and that adenosine at 1038 of UCA1 is critical to the recognition by IGF2BP2. Importantly, we showed that m6A writers, METTL3 and WTAP positively regulate UCA1 expression. Mechanically, IGF2BP2 increases the stability of m6A-modified UCA1. Clinically, IGF2BP2 is upregulated in CRC tissues compared with normal tissues.ConclusionThese results suggest that m6A modification is an important factor contributing to upregulation of UCA1 in CRC tissues.
- Published
- 2021
37. Retraction Notice to: lncRNA RMST Enhances DNMT3 Expression through Interaction with HuR
- Author
-
Wan-Xin Peng, Pratirodh Koirala, Wei Zhang, Chao Ni, Zheng Wang, Liu Yang, and Yin-Yuan Mo
- Subjects
Pharmacology ,Drug Discovery ,Genetics ,Molecular Medicine ,Molecular Biology - Published
- 2023
38. Epigenetic and Posttranscriptional Modulation of SOS1 Can Promote Breast Cancer Metastasis through Obesity-Activated c-Met Signaling in African-American Women
- Author
-
Shih-Ying Wu, Kounosuke Watabe, C Yalamanchili, Dan Zhao, Ravindra Pramod Deshpande, Lance D. Miller, Jacob Cleary, Yin Liu, Yin-Yuan Mo, Kerui Wu, Sambad Sharma, Amar G. Chittiboyina, Fei Xing, Abhishek Tyagi, and Yuezhu Wang
- Subjects
0301 basic medicine ,Cancer Research ,Lung Neoplasms ,C-Met ,Mice, Nude ,Apoptosis ,Breast Neoplasms ,CCL2 ,Article ,Epigenesis, Genetic ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Breast cancer ,Downregulation and upregulation ,Biomarkers, Tumor ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,Obesity ,Epigenetics ,Cell Proliferation ,GRB2 Adaptor Protein ,Mice, Inbred BALB C ,biology ,business.industry ,Anti-Inflammatory Agents, Non-Steroidal ,Cancer ,Proto-Oncogene Proteins c-met ,Prognosis ,medicine.disease ,Xenograft Model Antitumor Assays ,Black or African American ,Gene Expression Regulation, Neoplastic ,Mice, Inbred C57BL ,030104 developmental biology ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Female ,Quercetin ,GRB2 ,Signal transduction ,SOS1 Protein ,business - Abstract
Ethnicity is considered to be one of the major risk factors in certain subtypes of breast cancer. However, the mechanism of this racial disparity remains poorly understood. Here, we demonstrate that SOS1, a key regulator of Ras pathway, is highly expressed in African-American (AA) patients with breast cancer compared with Caucasian-American patients. Because of the higher obesity rate in AA women, increased levels of SOS1 facilitated signal transduction of the c-Met pathway, which was highly activated in AA patients with breast cancer via hepatocyte growth factor secreted from adipocytes. Elevated expression of SOS1 also enhanced cancer stemness through upregulation of PTTG1 and promoted M2 polarization of macrophages by CCL2 in metastatic sites. SOS1 was epigenetically regulated by a super-enhancer identified by H3K27ac in AA patients. Knockout of the super-enhancer by CRISPR in AA cell lines significantly reduced SOS1 expression. Furthermore, SOS1 was posttranscriptionally regulated by miR-483 whose expression is reduced in AA patients through histone trimethylation (H3K27me3) on its promoter. The natural compound, taxifolin, suppressed signaling transduction of SOS1 by blocking the interaction between SOS1 and Grb2, suggesting a potential utility of this compound as a therapeutic agent for AA patients with breast cancer. Significance: These findings elucidate the signaling network of SOS1-mediated metastasis in African-American patients, from the epigenetic upregulation of SOS1 to the identification of taxifolin as a potential therapeutic strategy against SOS1-driven tumor progression.
- Published
- 2021
39. Abstract P6-10-14: A new model to predict 10-year distant recurrence risk for operable endocrine-responsive breast cancer population
- Author
-
Yunyun Zhou, Xiaojia Wang, Lei Lei, Yin-Yuan Mo, and Skye Hung-Chun Cheng
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,education.field_of_study ,business.industry ,Distant recurrence ,Population ,Cancer ,medicine.disease ,Clinical trial ,Breast cancer ,Internal medicine ,medicine ,Biomarker (medicine) ,Endocrine system ,business ,education - Abstract
Background Few currently available biomarker panels can predict the risk of long-term distant recurrence (DR) in endocrine-responsive breast cancer (ERBC) patients based on lymph-node (LN) status. A new model, DGM-CM6 (Distant Genetic Model-Clinical variable Model 6), has been developed for the prediction of DR risk in our previous studies. To interrogate the prognostic value of our DGM-CM6 model for ERBC patients stratified by LN status, we retrospectively studied 752 operable breast cancer patients treated in a cancer center from 2005 to 2014. Patients and Methods Of 752 tumors, a total of 499 ERBC patients was identified from both IHC method (n=490) and PAM50 method (n=404). ERBC patients were classified by high and low-risk groups using the cutoff ( Results Significant differences were observed between DGM-CM6 low- and high-risk patients with 10-year DRFS in LN- (ERBC identified by IHC: P = 0.011; by PAM50: P< 0.0001) and LN+ (ERBC identified by IHC: P = 0.015; by PAM50: P = 0.019) by log-rank test. Compared to risk prediction by intrinsic subtype, DGM-CM6 performed better in LN- patients and equally good in LN+ patients. Further multivariate analysis confirmed the independent strength of DGM-CM6 model in the prediction of high- vs. low- risk groups in DRFS (P < 0.0001, HR: 6.76; 95% CI, 1.8-25.42) and OS (P = 0.01, HR: 6.06; 95% CI:1.55-23.47), respectively. Conclusion DGM-CM6 model can be used to predict low- and high-risk of 10-year distant recurrence in both LN- and LN+ ERBC patients. This model needs a large scale of clinical trials to validate its clinical utility. Table 1. Multivariate cox regression analyses for the prognosis of predicted risk groups after adjusted for other clinical variables in the distant recurrence (DR, p=0.005) and overall survival (OS, p=0.01) respectively. Results shows that there is no interaction between chemotherapy and risk groups (DR: p=0.163; OS: p=0.195). Abbreviations: DR, distant recurrence; OS: overall survival.GroupsDROSHR [95% CI]pvHR [95% CI]pvRisk: high vs.low6.76 [1.8;25.42]0.005 ***6.06 [1.55;23.74]0.01 ***Age: 50yrs0.86 [0.51;1.44]0.5630.68 [0.38;1.19]0.175LN: Pos. vs. Neg.1.64 [0.88;3.05]0.1161.6 [0.79;3.22]0.189Stage: II vs. I1.35 [0.76;2.4]0.31.45 [0.76;2.77]0.264Stage: III vs. I2.08 [0.59;7.3]0.2531.59 [0.34;7.46]0.557Grade: 2 vs. 12.14 [0.77;5.97]0.1461.81 [0.63;5.25]0.272Grade: 3 vs. 11.2 [0.4;3.62]0.7481.21 [0.38;3.86]0.752PAM50: Normal vs. LumA0.6 [0.13;2.64]0.4960.7 [0.16;3.16]0.645PAM50: LumB vs. LumA1.58 [0.84;2.98]0.1591.29 [0.65;2.59]0.469PAM50: Her2 vs. LumA0.81 [0.25;2.68]0.7311.01 [0.29;3.47]0.986RT: Yes vs. No0.88 [0.47;1.66]0.7010.71 [0.36;1.41]0.326CT: Yes vs.No0.36 [0.11;1.12]0.0770.3 [0.09;1]0.05 *Interaction: CT vs. Risk0.36 [0.09;1.51]0.1630.36 [0.08;1.68]0.195 Citation Format: Lei Lei, Skye Hung-Chun Cheng, Xiao-Jia Wang, Yin-Yuan Mo, Yun-Yun Zhou. A new model to predict 10-year distant recurrence risk for operable endocrine-responsive breast cancer population [abstract]. In: Proceedings of the 2019 San Antonio Breast Cancer Symposium; 2019 Dec 10-14; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2020;80(4 Suppl):Abstract nr P6-10-14.
- Published
- 2020
40. RETRACTED: lncRNA RMST Enhances DNMT3 Expression through Interaction with HuR
- Author
-
Wei Zhang, Liu Yang, Pratirodh Koirala, Chao Ni, Zheng Wang, Wanxin Peng, and Yin-Yuan Mo
- Subjects
Pharmacology ,Untranslated region ,0303 health sciences ,Cas9 ,DNMT3B ,Biology ,Cell biology ,03 medical and health sciences ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Drug Discovery ,RMST ,DNA methylation ,Genetics ,Molecular Medicine ,CRISPR ,Ectopic expression ,Molecular Biology ,Gene ,030304 developmental biology - Abstract
Large bodies of studies have shown that the CRISPR/Cas9-based library screening is a very powerful tool for the identification of gene functions. However, most of these studies have focused on protein-coding genes, and, furthermore, very few studies have used gene reporters for screening. In the present study, we generated DNA methyltransferase 3B (DNMT3B) reporter and screened a CRISPR/Cas9 synergistic activation mediator (SAM) library against a focused group of lncRNAs. With this screening approach, we identified Rhabdomyosarcoma 2-Associated Transcript (RMST) as a positive regulator for DNMT3B. This was confirmed by activation of the endogenous RMST by SAM or ectopic expression of RMST. Moreover, RMST knockout (KO) suppresses DNMT3, while rescue with RMST in the KO cells restores the DNMT3 level. Finally, RMST KO suppresses global DNA methylation, leading to the upregulation of methylation-regulated genes. Mechanistically, RMST promotes the interaction between the RNA-binding protein HuR and DNMT3B 3′ UTR, increasing the DNMT3B stability. Together, these results not only provide the feasibility of a reporter system for CRISPR library screening but also demonstrate the previously uncharacterized factor RMST as an important player in the modulation of DNA methylation.
- Published
- 2020
41. IGF2BP2 regulates DANCR by serving as an N6-methyladenosine reader
- Author
-
Xinchun Zhou, Yin-Yuan Mo, Xiaoge Hu, Liu Yang, Jiahong Jiang, Wanxin Peng, Dongsheng Huang, and Huaixiang Zhou
- Subjects
Adenosine ,Molecular biology ,medicine.medical_treatment ,Article ,chemistry.chemical_compound ,Mice ,Downregulation and upregulation ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Cancer ,Cell Proliferation ,Mice, Knockout ,Gene knockdown ,Mice, Inbred BALB C ,Cell growth ,Chemistry ,Growth factor ,RNA ,RNA-Binding Proteins ,Cell Biology ,medicine.disease ,Cell biology ,Pancreatic Neoplasms ,Ectopic expression ,RNA, Long Noncoding ,N6-Methyladenosine - Abstract
The major function of Insulin-like growth factor 2 mRNA-binding protein 2 (IGF2BP2) is to regulate cell metabolism. However, emerging evidence indicates that IGF2BP2 plays a role in cancer, but the underlying mechanism is largely unknown. Here we showed that upregulation of IGF2BP2 is associated with poor outcomes of pancreatic cancer patients and suppression of IGF2BP2 inhibits cell proliferation. We further showed that IGF2BP2 regulates lncRNA DANCR. Ectopic expression IGF2BP2 enhances, whereas knockdown (KD) or knockout (KO) of IGF2BP2 suppresses DANCR expression. Moreover, in vivo RNA precipitation and reciprocal RNA immunoprecipitation revealed that IGF2BP2 interacts with DANCR. DANCR promotes cell proliferation and stemness-like properties. Experiments with xenograft models revealed that while ectopic expression of DANCR promotes, DANCR KO suppresses tumor growth. Mechanistically, DANCR is modified at N6-methyladenosine (m6A) and mutagenesis assay identified that adenosine at 664 of DANCR is critical to the interaction between IGF2BP2 and DANCR where IGF2BP2 serves a reader for m6A modified DANCR and stabilizes DANCR RNA. Together, these results suggest that DANCR is a novel target for IGF2BP2 through m6A modification, and IGF2BP2 and DANCR work together to promote cancer stemness-like properties and pancreatic cancer pathogenesis.
- Published
- 2019
42. The association of prostatic lipids with progression, racial disparity and discovery of biomarkers in prostate cancer
- Author
-
Yin-Yuan Mo, Hao Mei, Zhenbang Chen, Wanxin Peng, Jinghe Mao, Patrick B. Kyle, Xinchun Zhou, and Timothy Craig Allen
- Subjects
Cancer Research ,Population ,Phospholipid ,urologic and male genital diseases ,Pathogenesis ,chemistry.chemical_compound ,Prostate cancer ,Lipidomics ,Caucasian Americans (CA) ,medicine ,Racial disparity ,Animal model ,Triglyceride (TG) ,education ,Pathological ,RC254-282 ,Original Research ,Prostate cancer (PCa) ,education.field_of_study ,business.industry ,African American (AA) ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,Oncology ,chemistry ,Cancer research ,Cholesteryl ester ,Population study ,lipids (amino acids, peptides, and proteins) ,Cholesteryl esters (CE) ,business ,Biomarkers - Abstract
Highlights • This study performed lipid profiling on human PCa and BPT tissues matched with patient's age and race, pathological grades and clinical stages. • The human prostatic lipid profiles were widely associated with the pathogenesis, progression and racial disparity of PCa. • Neutral lipids had greater impact on pathogenesis, progression and racial disparity of PCa as compared to phospholipids. Cholesteryl ester is the only lipid class significantly higher in PCa than in BPT in all population and stratified AA and CA populations. • A panel of prostatic lipid parameters in each study population were identified as diagnostic and prognostic biomarkers with high sensitivity, specificity and accuracy simultaneously. • Lipid profiling on mouse prostatic tissue from mouse PCa model further confirmed the roles of prostatic lipids on the pathogenesis, progression and discovery of diagnostic and prognostic biomarkers of PCa. In addition, this animal model was excellent to investigate prognostic lipid biomarkers in differentiation of indolent from aggressive PCa., Background It remains under-investigated whether prostatic lipid profiles are associated with pathogenesis, progression, racial disparity, and discovery of biomarkers in prostate cancer (PCa). Methods The electrospray ionization-tandem mass spectrometry was applied to quantitate prostatic lipids in human and mouse PCa and non-cancer prostatic tissues. Biostatistics and bioinformatics were used to compare the concentrations of prostatic lipids at levels of total lipid, group, class and individual species between PCa and benign prostatic tissues, between races, and among pathological conditions of PCa. Results Prostatic concentrations of total lipids as well as neutral lipids were significantly higher in PCa than in benign prostatic tissues in all population and Caucasian American population, but not in African American population. The prostatic phospholipid were not statistically different between PCa and benign prostatic tissues in all study populations. Cholesteryl ester is the only lipid class significantly higher in PCa than in benign prostatic tissues in all study populations. A panel of prostatic lipid parameters in each study population was identified as diagnostic and prognostic biomarkers with >60% of sensitivity, specificity and accuracy simultaneously. Lipid profiling on mouse prostatic tissues further confirmed correlation of prostatic lipid profiles to the pathogenesis and progression of PCa. In addition, a few prostatic lipids in mouse can serve as prognostic biomarkers in differentiation of indolent from aggressive PCa. Conclusion The prostatic lipids are widely associated with the pathogenesis, progression and racial disparity of PCa. A panel of prostatic lipids can serve as diagnostic, prognostic and race-specific biomarkers for PCa.
- Published
- 2021
43. Lnc-DC promotes estrogen independent growth and tamoxifen resistance in breast cancer
- Author
-
Liu Yang, Wanxin Peng, Yin-Yuan Mo, Ziqiang Zhang, Pratirodh Koirala, Huaixiang Zhou, and Jiahong Jiang
- Subjects
Cancer Research ,medicine.drug_class ,medicine.medical_treatment ,Immunology ,Mice, Nude ,Estrogen receptor ,Breast Neoplasms ,Article ,Non-coding RNAs ,Mice ,Cellular and Molecular Neuroscience ,Breast cancer ,Downregulation and upregulation ,medicine ,Animals ,Humans ,skin and connective tissue diseases ,Autocrine signalling ,QH573-671 ,business.industry ,Estrogens ,Cell Biology ,medicine.disease ,Tamoxifen ,Cytokine ,Drug Resistance, Neoplasm ,Estrogen ,Selective estrogen receptor modulator ,Cancer research ,Female ,RNA, Long Noncoding ,Cytology ,business ,hormones, hormone substitutes, and hormone antagonists ,medicine.drug - Abstract
Selective estrogen receptor modulators (SERMs) such as tamoxifen have proven to be effective in the treatment of estrogen receptor (ER) positive breast cancer. However, a major obstacle for such endocrine therapy is estrogen independent growth, leading to resistance, and the underlying mechanism is not fully understood. The purpose of this study was to determine whether long non-coding RNAs (lncRNAs) are involved in regulation of estrogen independent growth and tamoxifen resistance in ER positive breast cancer. Using a CRISPR/Cas9-based SAM (synergistic activation mediator) library against a focus group of lncRNAs, we identify Lnc-DC as a candidate lncRNA. Further analysis suggests that Lnc-DC is able to reduce tamoxifen-induced apoptosis by upregulation of anti-apoptotic genes such as Bcl2 and Bcl-xL. Furthermore, Lnc-DC activates STAT3 by phosphorylation (pSTAT3Y705), and the activated STAT3 subsequently induces expression of cytokines which in turn activate STAT3, forming an autocrine loop. Clinically, upregulation of Lnc-DC is associated with poor prognosis. In particular, analysis of a tamoxifen-treated patient cohort indicates that Lnc-DC expression can predict the response to tamoxifen. Together, this study demonstrates a previously uncharacterized function of Lnc-DC/STAT3/cytokine axis in estrogen independent growth and tamoxifen resistance, and Lnc-DC may serve as a potential predictor for tamoxifen response.
- Published
- 2021
44. Exosomal miR-19a and IBSP cooperate to induce osteolytic bone metastasis of estrogen receptor-positive breast cancer
- Author
-
Abhishek Tyagi, Shih-Ying Wu, Sambad Sharma, Kerui Wu, Feng Lyu, Takahiro Kimura, Dan Zhao, Xinhong Pei, Hiroyuki Takahashi, Yusuke Shiozawa, Marco Gabril Ruiz, Shunsuke Tsuzuki, Fei Xing, Yin-Yuan Mo, Yin Liu, Ravi Singh, Ravindra Pramod Deshpande, Jiamei Feng, and Kounosuke Watabe
- Subjects
Science ,Mice, Nude ,Osteoclasts ,General Physics and Astronomy ,Estrogen receptor ,Bone Neoplasms ,Breast Neoplasms ,Exosomes ,Article ,General Biochemistry, Genetics and Molecular Biology ,Mice ,Breast cancer ,Downregulation and upregulation ,Osteoclast ,medicine ,Animals ,Humans ,Integrin-Binding Sialoprotein ,Neoplasm Metastasis ,Liquid biopsy ,Receptor ,Cancer ,Mice, Knockout ,Multidisciplinary ,business.industry ,Bone metastasis ,General Chemistry ,medicine.disease ,MicroRNAs ,medicine.anatomical_structure ,Receptors, Estrogen ,Cancer research ,Female ,Complication ,business - Abstract
Bone metastasis is an incurable complication of breast cancer. In advanced stages, patients with estrogen-positive tumors experience a significantly higher incidence of bone metastasis (>87%) compared to estrogen-negative patients (, Bone metastasis is a major complication of breast cancer (BC) and ER+ tumors have a higher incidence of bone metastasis than ER− tumors. Here, the authors report that miR‐19a in exosomes and the bone matrix protein, IBSP, are upregulated and secreted by bone tropic ER+ BC cells, where they cooperatively induce osteoclastogenesis and promote bone colonization.
- Published
- 2021
45. LncRNA IPW inhibits growth of ductal carcinoma in situ by downregulating ID2 through miR-29c
- Author
-
Ravindra Pramod Deshpande, Sambad Sharma, Yin Liu, Puspa Raj Pandey, Xinhong Pei, Kerui Wu, Shih-Ying Wu, Abhishek Tyagi, Dan Zhao, Yin-Yuan Mo, and Kounosuke Watabe
- Subjects
miR-29c ,DCIS ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Breast Neoplasms ,ID2 ,body regions ,Gene Expression Regulation, Neoplastic ,MicroRNAs ,Carcinoma, Intraductal, Noninfiltrating ,Cell Line, Tumor ,Humans ,Female ,Genes, Tumor Suppressor ,RNA, Long Noncoding ,skin and connective tissue diseases ,LncRNA IPW ,neoplasms ,RC254-282 ,Inhibitor of Differentiation Protein 2 ,Research Article ,Toyocamycin - Abstract
Background Ductal carcinoma in situ (DCIS) of breast is the noninvasive lesion that has propensity to progress to the malignant form. At present, it is still unknown which lesions can potentially progress to invasive forms. In this study, we aimed to identify key lncRNAs involved in DCIS growth. Methods We employ disease-related lncProfiler array to identify IPW in specimens of DCIS and matching control samples and validate the observations in three DCIS-non-tumorigenic cell lines. Further, we examine the mechanism of IPW action and the downstream signaling in in vitro and in vivo assays. Importantly, we screened a library containing 390 natural compounds to identify candidate compound selectively inhibiting IPW low DCIS cells. Results We identified lncRNA IPW as a novel tumor suppressor critical for inhibiting DCIS growth. Ectopic expression of IPW in DCIS cells strongly inhibited cell proliferation, colony formation and cell cycle progression while silencing IPW in primary breast cells promoted their growth. Additionally, orthotropic implantation of cells with ectopic expression of IPW exhibited decreased tumor growth in vivo. Mechanistically, IPW epigenetically enhanced miR-29c expression by promoting H3K4me3 enrichment in its promoter region. Furthermore, we identified that miR-29c negatively regulated a stemness promoting gene, ID2, and diminished self-renewal ability of DCIS cells. Importantly, we screened a library containing 390 natural compounds and identified toyocamycin as a compound that selectively inhibited the growth of DCIS with low expression of IPW, while it did not affect DCIS with high IPW expression. Toyocamycin also suppressed genes associated with self-renewal ability and inhibited DCIS growth in vivo. Conclusion Our findings revealed a critical role of the IPW-miR-29c-ID2 axis in DCIS formation and suggested potential clinical use of toyocamycin for the treatment of DCIS.
- Published
- 2021
46. LINC00346 promotes pancreatic cancer progression through the CTCF-mediated Myc transcription
- Author
-
Rong-Zhang He, Yin-Yuan Mo, Liu Yang, Ziqiang Zhang, and Wanxin Peng
- Subjects
0301 basic medicine ,CCCTC-Binding Factor ,Cancer Research ,Transcription, Genetic ,Biology ,medicine.disease_cause ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Transcription (biology) ,Pancreatic tumor ,Pancreatic cancer ,Biomarkers, Tumor ,Genetics ,medicine ,Transcriptional regulation ,Humans ,Neoplasm Invasiveness ,Neoplasm Metastasis ,Promoter Regions, Genetic ,Molecular Biology ,Cell Proliferation ,Prognosis ,medicine.disease ,Pancreatic Neoplasms ,030104 developmental biology ,CTCF ,030220 oncology & carcinogenesis ,Disease Progression ,Cancer research ,Adenocarcinoma ,RNA, Long Noncoding ,Carcinogenesis ,Carcinoma, Pancreatic Ductal ,Protein Binding - Abstract
Although multiple factors are known to contribute to pancreatic ductal adenocarcinoma (PDAC) progression, the role of long non-coding RNAs (lncRNAs) in PDAC remains largely unknown. In this study, we present data that long intergenic non-coding RNA 346 (LINC00346) functions as a promoting factor for PDAC development. We first show that LINC00346 is highly expressed in pancreatic tumor specimens as compared to normal pancreatic tissue based on interrogation of The Cancer Genome Atlas (TCGA) pancreatic adenocarcinoma dataset. Of significance, this upregulation of LINC00346 is associated with overall survival (OS) and disease-free survival (DFS), respectively. We further show that knockout (KO) of LINC00346 impairs pancreatic cancer cell proliferation, tumorigenesis, migration, and invasion ability. Importantly, these phenotypes can be restored by LINC00346 re-expression in KO cells (i.e., rescue experiment). RNA precipitation assays combined with mass spectrometry analysis indicate that LINC00346 interacts with CCCTC-binding factor (CTCF), a known transcriptional repressor of c-Myc. This interaction between LINC00346 and CTCF prevents the binding of CTCF to c-Myc promoter, relieving the CTCF-mediated repression of c-Myc. Thus, LINC00346 functions as a positive transcriptional regulator of c-Myc. Together, these results suggest that LINC00346 contributes to PDAC pathogenesis by activating c-Myc, and as such, LINC00346 may serve as a potential biomarker and therapeutic target for PDAC.
- Published
- 2019
47. Emerging roles of lncRNAs in the post-transcriptional regulation in cancer
- Author
-
Yin-Yuan Mo, Di-Xian Luo, and Rong-Zhang He
- Subjects
0301 basic medicine ,RNA Stability ,lcsh:QH426-470 ,RNA-binding protein ,Biology ,medicine.disease_cause ,Biochemistry ,Article ,03 medical and health sciences ,0302 clinical medicine ,Gene expression ,Protein stability ,medicine ,Molecular Biology ,Post-transcriptional regulation ,Gene ,Genetics (clinical) ,Messenger RNA ,lcsh:R5-920 ,Alternative splicing ,Cell Biology ,RNA stability ,LncRNA ,Cell biology ,lcsh:Genetics ,030104 developmental biology ,030220 oncology & carcinogenesis ,RNA binding proteins ,Carcinogenesis ,lcsh:Medicine (General) ,Posttranscriptional regulation - Abstract
Accumulating evidence indicates that long non-coding RNAs (lncRNAs) can play a pivotal role in regulation of diverse cellular processes. In particular, lncRNAs can serve as master gene regulators at transcriptional and posttranscriptional levels, leading to tumorigenesis. In this review, we discuss latest developments in lncRNA-meditated gene expression at the post-transcriptional level, including gene splicing, mRNA stability, protein stability and nuclear trafficking. Keywords: Alternative splicing, LncRNA, Posttranscriptional regulation, Protein stability, RNA binding proteins, RNA stability
- Published
- 2019
48. Gut microbiota regulate tumor metastasis via circRNA/miRNA networks
- Author
-
Ziqiang Zhang, Ruilin Liu, Zhongmin Qiu, Xu Li, Jun Xing, Feng Hua, Yin-Yuan Mo, Chunxue Bai, Qiang Chen, Jianguo Huang, Zhu Zhuxian, and Yuan-Lin Song
- Subjects
0301 basic medicine ,Microbiology (medical) ,circular rna (circrna) ,cancer stem cell ,Epithelial-Mesenchymal Transition ,microrna ,Gut flora ,Microbiology ,digestive system ,Metastasis ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cancer stem cell ,microRNA ,medicine ,Animals ,cancer ,metastasis ,Neoplasm Metastasis ,lcsh:RC799-869 ,biology ,gut microbiota ,Mechanism (biology) ,Gastroenterology ,Cancer ,SOX9 Transcription Factor ,RNA, Circular ,biology.organism_classification ,medicine.disease ,Interleukin-11 ,Anti-Bacterial Agents ,Gastrointestinal Microbiome ,Gene Expression Regulation, Neoplastic ,MicroRNAs ,030104 developmental biology ,Infectious Diseases ,Cancer research ,Neoplastic Stem Cells ,Dysbiosis ,030211 gastroenterology & hepatology ,lcsh:Diseases of the digestive system. Gastroenterology ,interleukin-11(il-11) ,Signal Transduction ,Research Article ,Research Paper - Abstract
Background Increasing evidence indicates that gut microbiota plays an important role in cancer progression. However, the underlying mechanism remains largely unknown. Here, we report that broad-spectrum antibiotics (ABX) treatment leads to enhanced metastasis by the alteration of gut microbiome composition. Methods Cancer LLC and B16-F10 cell metastasis mouse models, and microarray/RNA sequencing analysis were used to reveal the regulatory functions of microbiota-mediated circular RNA (circRNA)/microRNA (miRNA) networks that may contribute to cancer metastasis. Results The specific pathogen-free (SPF) mice with ABX treatment demonstrated enhanced lung metastasis. Fecal microbiota transplantation (FMT) from SPF mice or Bifidobacterium into germ-free mice significantly suppressed lung metastasis. Mechanistically, gut microbiota impacts circRNA expression to regulate levels of corresponding miRNAs. Specifically, such modulations of gut microbiota inhibit mmu_circ_0000730 expression in an IL-11-dependent manner. Bioinformatics analysis combined with luciferase reporter assays revealed reciprocal repression between mmu_circ_0000730 and mmu-miR-466i-3p. We further showed that both mmu-miR-466i-3p and mmu-miR-466 f-3p suppresses a number of genes involved in epithelial-mesenchymal transition (EMT) and stemness of cancer stem cells such as SOX9. Conclusions These results provide evidence of a previously unrecognized regulatory role of non-coding RNAs in microbiota-mediated cancer metastasis, and thus, the microbiome may serve as a therapeutic target.
- Published
- 2020
49. DGM-CM6: A New Model to Predict Distant Recurrence Risk in Operable Endocrine-Responsive Breast Cancer
- Author
-
Skye Hung-Chun Cheng, Yunyun Zhou, Xiaojia Wang, Lei Lei, and Yin-Yuan Mo
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Multivariate analysis ,lcsh:RC254-282 ,03 medical and health sciences ,breast cancer ,0302 clinical medicine ,Breast cancer ,endocrine-responsive ,Internal medicine ,medicine ,Endocrine system ,Lymph node ,Original Research ,business.industry ,clinical-genomic model ,Distant recurrence ,Cancer ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,Clinical trial ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Immunohistochemistry ,distant recurrence ,prognosis ,business - Abstract
To investigate the prognostic value of DGM-CM6 (Distant Genetic Model-Clinical variable Model 6) for endocrine-responsive breast cancer (ERBC) patients, we analyzed 752 operable breast cancer patients treated in a Taiwan cancer center from 2005 to 2014. Among them, 490 ERBC patients (identified by the PAM50 or immunohistochemistry method) were classified by DGM-CM6 into low- and high-risk groups (cutoff
- Published
- 2020
50. A highly sensitive and selective signal-on strategy for microRNA quantification
- Author
-
Rui Xu, Yin-Yuan Mo, Huaisheng Zhang, Paul B. Tchounwou, Li Pan, Jingjin Zhao, Yi-Ming Liu, and Xiangtang Li
- Subjects
Analyte ,Calibration curve ,Cell Culture Techniques ,DNA, Single-Stranded ,02 engineering and technology ,01 natural sciences ,Biochemistry ,High-performance liquid chromatography ,Article ,Analytical Chemistry ,Limit of Detection ,Environmental Chemistry ,Animals ,Humans ,Solid phase extraction ,Bovine serum albumin ,Locked nucleic acid ,Spectroscopy ,Polymerase ,Chromatography, High Pressure Liquid ,Detection limit ,Chromatography ,biology ,Chemistry ,010401 analytical chemistry ,021001 nanoscience & nanotechnology ,0104 chemical sciences ,MicroRNAs ,Calibration ,biology.protein ,MCF-7 Cells ,Biological Assay ,Cattle ,0210 nano-technology ,Nucleic Acid Amplification Techniques - Abstract
MicroRNAs (miRNAs) are associated with physiological and pathological processes. They are recognized as biomarkers for diseases diagnosis and treatment evaluation. Herein we propose a simple and cost-effective HPLC method for quantitative assay of target miRNAs with femtomolar sensitivity, single-base discrimination selectivity and low background. The assay is based on an innovative signal-on strategy. In this strategy, polyadenylation of poly(A) polymerase extends an all ‘A’ sequence at the end of target miRNA, and the substantially increased number of adenine bases are labeled with 2-Chloroacetaldehyde (CAA) to open a signal-on mode and realize a signal amplification. The linearly amplified fluorescence signal is separated from other inference signals and quantified by high performance liquid chromatography with fluorescence detection (HPLC-FD). Combining with affinity magnetic solid phase extraction (MSPE), the method is well suited for analysis of complex biological samples such as serum and cell lysate with nearly zero background fluorescence. Taking miRNA-21 as the model analyte, this absolute quantification method has a limit of detection of 200 fM and a linear calibration curve (R2 = 0.999) in the range from 2.00 pM to 1.00 nM. Using locked nucleic acid (LNA) modified probes rather than ssDNA probes, the assay selectivity is improved. Moreover, analysis of bovine serum and cell lysate samples by using the method is demonstrated. Intracellular content of miRNA-21 is found to be 0.0150 amol/cell in MCF-7 cells with an assay repeatability of 4.0% (RSD, n = 3). The present HPLC quantification of miRNA offers an accurate, reliable, and cost-effective means for quantitative assay of miRNAs occurring in biological samples. Also importantly, it eliminates the need for total RNA isolation for the analysis. It may be useful for more effective diagnosis of diseases and therapeutic evaluation.
- Published
- 2019
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.