Network


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

Hotspot


Dive into the research topics where Yong Peng is active.

Publication


Featured researches published by Yong Peng.


Leukemia | 2014

CS1-specific chimeric antigen receptor (CAR)-engineered natural killer cells enhance in vitro and in vivo antitumor activity against human multiple myeloma

Jianhong Chu; Youcai Deng; Don M. Benson; Shun He; Tiffany Hughes; Jianying Zhang; Yong Peng; Hsiaoyin Mao; Ling Yi; Kalpana Ghoshal; Xiaoming He; Steven M. Devine; Xiaoliu Zhang; Michael A. Caligiuri; Craig C. Hofmeister; Jianhua Yu

Multiple myeloma (MM) is an incurable hematological malignancy. Chimeric antigen receptor (CAR)-expressing T cells have been demonstrated successfully in the clinic to treat B-lymphoid malignancies. However, the potential utility of antigen-specific CAR-engineered natural-killer (NK) cells to treat MM has not been explored. In this study, we determined whether CS1, a surface protein that is highly expressed on MM cells, can be targeted by CAR NK cells to treat MM. We successfully generated a viral construct of a CS1-specific CAR and expressed it in human NK cells. In vitro, CS1-CAR NK cells displayed enhanced MM cytolysis and interferon-γ (IFN-γ) production, and showed a specific CS1-dependent recognition of MM cells. Ex vivo, CS1-CAR NK cells also showed similarly enhanced activities when responding to primary MM tumor cells. More importantly, in an aggressive orthotopic MM xenograft mouse model, adoptive transfer of NK-92 cells expressing CS1-CAR efficiently suppressed the growth of human IM9 MM cells and also significantly prolonged mouse survival. Thus, CS1 represents a viable target for CAR-expressing immune cells, and autologous or allogeneic transplantation of CS1-specific CAR NK cells may be a promising strategy to treat MM.


Signal Transduction and Targeted Therapy | 2016

The role of MicroRNAs in human cancer

Yong Peng; Carlo M. Croce

MicroRNAs (miRNAs) are endogenous, small non-coding RNAs that function in regulation of gene expression. Compelling evidences have demonstrated that miRNA expression is dysregulated in human cancer through various mechanisms, including amplification or deletion of miRNA genes, abnormal transcriptional control of miRNAs, dysregulated epigenetic changes and defects in the miRNA biogenesis machinery. MiRNAs may function as either oncogenes or tumor suppressors under certain conditions. The dysregulated miRNAs have been shown to affect the hallmarks of cancer, including sustaining proliferative signaling, evading growth suppressors, resisting cell death, activating invasion and metastasis, and inducing angiogenesis. An increasing number of studies have identified miRNAs as potential biomarkers for human cancer diagnosis, prognosis and therapeutic targets or tools, which needs further investigation and validation. In this review, we focus on how miRNAs regulate the development of human tumors by acting as tumor suppressors or oncogenes.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Long-range interaction and correlation between MYC enhancer and oncogenic long noncoding RNA CARLo-5

Taewan Kim; Ri Cui; Young Jun Jeon; Ji Hoon Lee; Ju Hee Lee; Hosung Sim; Jong Kook Park; Paolo Fadda; Esmerina Tili; Hiroshi Nakanishi; Man Il Huh; Sung Hak Kim; Ju Hwan Cho; Bong Hwan Sung; Yong Peng; Tae Jin Lee; Zhenghua Luo; Hui Lung Sun; Huijun Wei; Hansjuerg Alder; Jeong Su Oh; Kang Sup Shim; Sang Bong Ko; Carlo M. Croce

Significance Many cancer-associated variants have been found in the 8q24.21 region harboring enhancer activity. However, the functional mechanism of the variants is not clear due to the lack of protein-coding genes in the region and no significant correlation with the nearest oncogene MYC. We identified long noncoding RNAs (lncRNAs) named cancer-associated region long noncoding RNAs (CARLos) in the 8q24.21 region. Interestingly, we found that the cancer-associated variant rs6983267 regulating the enhancer activity is significantly associated with the expression of one of the lncRNAs CARLo-5 and that CARLo-5 has an oncogenic function. By showing direct interaction between the enhancer region and active regulatory region of the CARLo-5 promoter, we provide a regulatory mechanism of cancer susceptibility caused by the cancer-associated variants. The mechanism by which the 8q24 MYC enhancer region, including cancer-associated variant rs6983267, increases cancer risk is unknown due to the lack of protein-coding genes at 8q24.21. Here we report the identification of long noncoding RNAs named cancer-associated region long noncoding RNAs (CARLos) in the 8q24 region. The expression of one of the long noncoding RNAs, CARLo-5, is significantly correlated with the rs6983267 allele associated with increased cancer susceptibility. We also found the MYC enhancer region physically interacts with the active regulatory region of the CARLo-5 promoter, suggesting long-range interaction of MYC enhancer with the CARLo-5 promoter regulates CARLo-5 expression. Finally, we demonstrate that CARLo-5 has a function in cell-cycle regulation and tumor development. Overall, our data provide a key of the mystery of the 8q24 gene desert.


Molecular Pharmaceutics | 2009

Transferrin Receptor-Targeted Lipid Nanoparticles for Delivery of an Antisense Oligodeoxyribonucleotide against Bcl-2

Xiaojuan Yang; Chee Guan Koh; Shujun Liu; Xiaogang Pan; Ramasamy Santhanam; Bo Yu; Yong Peng; Jiuxia Pang; Sharon Golan; Yeshayahu Talmon; Yan Jin; Natarajan Muthusamy; John C. Byrd; Kenneth K. Chan; L. James Lee; Guido Marcucci; Robert J. Lee

Antisense oligonucleotide G3139-mediated down-regulation of Bcl-2 is a potential strategy for overcoming chemoresistance in leukemia. However, the limited efficacy shown in recent clinical trials calls attention to the need for further development of novel and more efficient delivery systems. In order to address this issue, transferrin receptor (TfR)-targeted, protamine-containing lipid nanoparticles (Tf-LNs) were synthesized as delivery vehicles for G3139. The LNs were produced by an ethanol dilution method, and lipid-conjugated Tf ligand was then incorporated by a postinsertion method. The resulting Tf-LNs had a mean particle diameter of approximately 90 nm and G3139 loading efficiency of 90.4%. Antisense delivery efficiency of Tf-LNs was evaluated in K562, MV4-11, and Raji leukemia cell lines. The results showed that Tf-LNs were more effective than nontargeted LNs and free G3139 (p < 0.05) in decreasing Bcl-2 expression (by up to 62% at the mRNA level in K562 cells) and in inducing caspase-dependent apoptosis. In addition, Bcl-2 down-regulation and apoptosis induced by Tf-LN G3139 were shown to be blocked by excess free Tf and thus were TfR-dependent. Cell lines with higher TfR expression also showed greater Bcl-2 down-regulation. Furthermore, up-regulation of TfR expression in leukemia cells by iron chelator deferoxamine resulted in a further increase in antisense effect (up to 79% Bcl-2 reduction in K562 at the mRNA level) and in caspase-dependent apoptosis (by approximately 3-fold) by Tf-LN. Tf-LN-mediated delivery combined with TfR up-regulation by deferoxamine appears to be a potentially promising strategy for enhancing the delivery efficiency and therapeutic efficacy of antisense oligonucleotides.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Insulin growth factor signaling is regulated by microRNA-486, an underexpressed microRNA in lung cancer

Yong Peng; Yuntao Dai; Charles L. Hitchcock; Xiaojuan Yang; Edmund S. Kassis; Lunxu Liu; Zhenghua Luo; Hui Lung Sun; Ri Cui; Huijun Wei; Taewan Kim; Tae Jin Lee; Young Jun Jeon; Gerard J. Nuovo; Stefano Volinia; Qianchuan He; Jianhua Yu; Patrick Nana-Sinkam; Carlo M. Croce

MicroRNAs (miRNAs) are small 19- to 24-nt noncoding RNAs that have the capacity to regulate fundamental biological processes essential for cancer initiation and progression. In cancer, miRNAs may function as oncogenes or tumor suppressors. Here, we conducted global profiling for miRNAs in a cohort of stage 1 nonsmall cell lung cancers (n = 81) and determined that miR-486 was the most down-regulated miRNA in tumors compared with adjacent uninvolved lung tissues, suggesting that miR-486 loss may be important in lung cancer development. We report that miR-486 directly targets components of insulin growth factor (IGF) signaling including insulin-like growth factor 1 (IGF1), IGF1 receptor (IGF1R), and phosphoinositide-3-kinase, regulatory subunit 1 (alpha) (PIK3R1, or p85a) and functions as a potent tumor suppressor of lung cancer both in vitro and in vivo. Our findings support the role for miR-486 loss in lung cancer and suggest a potential biological link to p53.


PLOS ONE | 2013

Curcumin Down-Regulates DNA Methyltransferase 1 and Plays an Anti-Leukemic Role in Acute Myeloid Leukemia

Jianhua Yu; Yong Peng; Lai-Chu Wu; Zhiliang Xie; Youcai Deng; Tiffany Hughes; Shun He; Xiao Kui Mo; Ming Chiu; Qi-En Wang; Xiaoming He; Shujun Liu; Michael R. Grever; Kenneth K. Chan; Zhongfa Liu

Bioactive components from dietary supplements such as curcumin may represent attractive agents for cancer prevention or treatment. DNA methylation plays a critical role in acute myeloid leukemia (AML) development, and presents an excellent target for treatment of this disease. However, it remains largely unknown how curcumin, a component of the popular Indian spice turmeric, plays a role in DNA hypomethylation to reactivate silenced tumor suppressor genes and to present a potential treatment option for AML. Here we show that curcumin down-regulates DNMT1 expression in AML cell lines, both in vitro and in vivo, and in primary AML cells ex vivo. Mechanistically, curcumin reduced the expression of positive regulators of DNMT1, p65 and Sp1, which correlated with a reduction in binding of these transcription factors to the DNMT1 promoter in AML cell lines. This curcumin-mediated down-regulation of DNMT1 expression was concomitant with p15INK4B tumor suppressor gene reactivation, hypomethylation of the p15INK4B promoter, G1 cell cycle arrest, and induction of tumor cell apoptosis in vitro. In mice implanted with the human AML MV4–11 cell line, administration of curcumin resulted in remarkable suppression of AML tumor growth. Collectively, our data indicate that curcumin shows promise as a potential treatment for AML, and our findings provide a basis for future studies to test the clinical efficacy of curcumin – whether used as a single agent or as an adjuvant – for AML treatment.


Clinical Cancer Research | 2014

Genetic Modification of T Cells Redirected toward CS1 Enhances Eradication of Myeloma Cells

Jianhong Chu; Shun He; Youcai Deng; Jianying Zhang; Yong Peng; Tiffany Hughes; Ling Yi; Chang-Hyuk Kwon; Qi-En Wang; Steven M. Devine; Xiaoming He; Xue-Feng Bai; Craig C. Hofmeister; Jianhua Yu

Purpose: Our goal is to test whether CS1 could be targeted by chimeric antigen receptor (CAR) T cells to treat multiple myeloma (MM). Experimental Design: We generated a retroviral construct of a CS1-specific CAR and engineered primary human T cells expressing the CAR. We then tested the capacity of CS1–CAR T cells to eradicate human MM tumor cells in vitro, ex vivo, and in vivo using orthotopic MM xenograft mouse models. Results: In vitro, compared with mock-transduced T cells, upon recognizing CS1-positive MM cells, CS1–CAR-transduced T cells secreted more IFN-γ as well as interleukin (IL)-2, expressed higher levels of the activation marker CD69, showed higher capacity for degranulation, and displayed enhanced cytotoxicity. Ectopically forced expression of CS1 in MM cells with low CS1 expression enhanced recognition and killing by CAR T cells. Ex vivo, CS1–CAR T cells also showed similarly enhanced activities when responding to primary MM cells. More importantly, in orthotopic MM xenograft mouse models, adoptive transfer of human primary T cells expressing CS1–CAR efficiently suppressed the growth of human MM.1S and IM9 myeloma cells and significantly prolonged mouse survival. Conclusions: CS1 is a promising antigen that can be targeted by CAR-expressing T cells for treatment of MM. Clin Cancer Res; 20(15); 3989–4000. ©2014 AACR.


Proceedings of the National Academy of Sciences of the United States of America | 2015

MicroRNA-224 promotes tumor progression in nonsmall cell lung cancer

Ri Cui; Wei Meng; Hui Lung Sun; Taewan Kim; Zhenqing Ye; Matteo Fassan; Young Jun Jeon; Bin Li; Caterina Vicentini; Yong Peng; Tae Jin Lee; Zhenghua Luo; Lan Liu; Dongyuan Xu; Esmerina Tili; Victor X. Jin; Justin Middleton; Arnab Chakravarti; Tim Lautenschlaeger; Carlo M. Croce

Significance Aberrant microRNA (miRNA) expression is involved in tumorigenesis, and miR-224 was observed to be up-regulated in certain tumor types. However, the role of miR-224 in the pathogenesis of lung cancer remains poorly understood. Here, we comprehensively analyzed and revealed mechanisms of miR-224 up-regulation and its oncogenic role in nonsmall cell lung cancer (NSCLC). We showed that miR-224 promotes cellular migratory, invasive, and proliferative capacity and tumor growth both in vitro and in vivo. Furthermore, we identified TNFα-induced protein 1 and SMAD4 as targets of miR-224. In addition, up-regulated miR-224 expression in NSCLC is partially controlled by its promoter region’s hypomethylation and activated ERK signaling. Our finding suggests that targeting miR-224 might be a promising therapeutic strategy in the treatment of NSCLC. Lung cancer is the leading cause of cancer-related deaths worldwide. Despite advancements and improvements in surgical and medical treatments, the survival rate of lung cancer patients remains frustratingly poor. Local control for early-stage nonsmall cell lung cancer (NSCLC) has dramatically improved over the last decades for both operable and inoperable patients. However, the molecular mechanisms of NSCLC invasion leading to regional and distant disease spread remain poorly understood. Here, we identify microRNA-224 (miR-224) to be significantly up-regulated in NSCLC tissues, particularly in resected NSCLC metastasis. Increased miR-224 expression promotes cell migration, invasion, and proliferation by directly targeting the tumor suppressors TNFα-induced protein 1 (TNFAIP1) and SMAD4. In concordance with in vitro studies, mouse xenograft studies validated that miR-224 functions as a potent oncogenic miRNA in NSCLC in vivo. Moreover, we found promoter hypomethylation and activated ERK signaling to be involved in the regulation of miR-224 expression in NSCLC. Up-regulated miR-224, thus, facilitates tumor progression by shifting the equilibrium of the partially antagonist functions of SMAD4 and TNFAIP1 toward enhanced invasion and growth in NSCLC. Our findings indicate that targeting miR-224 could be effective in the treatment of certain lung cancer patients.


Journal of the National Cancer Institute | 2015

Role of MYC-Regulated Long Noncoding RNAs in Cell Cycle Regulation and Tumorigenesis

Taewan Kim; Young Jun Jeon; Ri Cui; Ji Hoon Lee; Yong Peng; Sung Hak Kim; Esmerina Tili; Hansjuerg Alder; Carlo M. Croce

BACKGROUNDnThe functions of long noncoding RNAs (lncRNAs) have been identified in several cancers, but the roles of lncRNAs in colorectal cancer (CRC) are less well understood. The transcription factor MYC is known to regulate lncRNAs and has been implicated in cancer cell proliferation and tumorigenesis.nnnMETHODSnCRC cells and tissues were profiled to identify lncRNAs differentially expressed in CRC, from which we further selected MYC-regulated lncRNAs. We used luciferase promoter assay, ChIP, RNA pull-down assay, deletion mapping assay, LC-MS/MS and RNA immunoprecipitation to determine the mechanisms of MYC regulation of lncRNAs. Moreover, soft agar assay and in vivo xenograft experiments (four athymic nude mice per group) provided evidence of MYC-regulated lncRNAs in cancer cell transformation and tumorigenesis. The Kaplan-Meier method was used for survival analyses. All statistical tests were two-sided.nnnRESULTSnWe identified lncRNAs differentially expressed in CRC (P < .05, greater than two-fold) and verified four lncRNAs upregulated and two downregulated in CRC cells and tissues. We further identified MYC-regulated lncRNAs, named MYCLos. The MYC-regulated MYCLos may function in cell proliferation and cell cycle by regulating MYC target genes such as CDKN1A (p21) and CDKN2B (p15), suggesting new regulatory mechanisms of MYC-repressed target genes through lncRNAs. RNA binding proteins including HuR and hnRNPK are involved in the function of MYCLos by interacting with MYCLo-1 and MYCLo-2, respectively. Knockdown experiments also showed that MYCLo-2, differentially expressed not only in CRC but also in prostate cancer, has a role in cancer transformation and tumorigenesis.nnnCONCLUSIONSnOur results provide novel regulatory mechanisms in MYC function through lncRNAs and new potential lncRNA targets of CRC.


Blood | 2013

MicroRNAs activate natural killer cells through Toll-like receptor signaling.

Shun He; Jianhong Chu; Lai-Chu Wu; Hsiaoyin Mao; Yong Peng; Christopher Alvarez-Breckenridge; Tiffany Hughes; Min Wei; Jianying Zhang; Shunzong Yuan; Sumeet Sandhu; Sumithira Vasu; Don M. Benson; Craig C. Hofmeister; Xiaoming He; Kalpana Ghoshal; Steven M. Devine; Michael A. Caligiuri; Jianhua Yu

MicroRNAs (miRNAs) bind to complementary sequences of target mRNAs, resulting in translational repression or target degradation and thus gene silencing. miRNAs are abundant in circulating blood, yet it is not known whether, as a class of regulatory molecules, they interact with human natural killer (NK) cells. Here we found that the treatment of human NK cells with several mature miRNAs in the presence of a low concentration of interleukin-12 induced CD69 expression, interferon-γ production, and degranulation marker CD107a expression. In vivo, infusion of several miRNAs alone in murine peripheral blood also resulted in comparable NK-cell activation, but not T-cell activation. Furthermore, miRNA administration significantly protected mice from tumor development in an NK cell-dependent manner. Mechanistically, we found that miRNA stimulation led to downstream activation of nuclear factor κB (NF-κB), an effect that was blunted by a block in Toll-like receptor 1(TLR1) signaling and attenuated in lymphoma patients. Knockdown of TLR1 resulted in less activation by miRNAs. Collectively, we show that miRNAs have a capacity to selectively activate innate immune effector cells that is, at least in part, via the TLR1-NF-κB signaling pathway. This may be important in the normal host defense against infection and/or malignant transformation.

Collaboration


Dive into the Yong Peng's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge