Network


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

Hotspot


Dive into the research topics where Qinlong Li is active.

Publication


Featured researches published by Qinlong Li.


Oncogene | 2015

Stromal fibroblast-derived miR-409 promotes epithelial-to-mesenchymal transition and prostate tumorigenesis.

Sajni Josson; Murali Gururajan; S Y Sung; Peizhen Hu; Chen Shao; Haiyen E. Zhau; Chunyan Liu; Jake Lichterman; Peng Duan; Qinlong Li; Andre Rogatko; Edwin M. Posadas; Christopher L. Haga; Leland W.K. Chung

Tumor–stromal interaction is a dynamic process that promotes tumor growth and metastasis via cell–cell interaction and extracellular vesicles. Recent studies demonstrate that stromal fibroblast-derived molecular signatures can be used to predict disease progression and drug resistance. To identify the epigenetic role of stromal noncoding RNAs in tumor–stromal interactions in the tumor microenvironment, we performed microRNA profiling of patient cancer-associated prostate stromal fibroblasts isolated by laser capture dissection microscopy and in bone-associated stromal models. We found specific upregulation of miR-409-3p and miR-409-5p located within the embryonically and developmentally regulated DLK1-DIO3 (delta-like 1 homolog-deiodinase, iodothyronine 3) cluster on human chromosome 14. The findings in cell lines were further validated in human prostate cancer tissues. Strikingly, ectopic expression of miR-409 in normal prostate fibroblasts conferred a cancer-associated stroma-like phenotype and led to the release of miR-409 via extracellular vesicles to promote tumor induction and epithelial-to-mesenchymal transition in vitro and in vivo. miR-409 promoted tumorigenesis through repression of tumor suppressor genes such as Ras suppressor 1 and stromal antigen 2. Thus, stromal fibroblasts derived miR-409-induced tumorigenesis, epithelial-to-mesenchymal transition and stemness of the epithelial cancer cells in vivo. Therefore, miR-409 appears to be an attractive therapeutic target to block the vicious cycle of tumor–stromal interactions that plagues prostate cancer patients.


Journal of Clinical Investigation | 2014

Monoamine oxidase A mediates prostate tumorigenesis and cancer metastasis

Jason Boyang Wu; Chen Shao; Qinlong Li; Peizhen Hu; Changhong Shi; Yang Li; Yi-Ting Chen; Fei Yin; Chun-Peng Liao; Bangyan L. Stiles; Haiyen E. Zhau; Jean C. Shih; Leland W.K. Chung

Tumors from patients with high-grade aggressive prostate cancer (PCa) exhibit increased expression of monoamine oxidase A (MAOA), a mitochondrial enzyme that degrades monoamine neurotransmitters and dietary amines. Despite the association between MAOA and aggressive PCa, it is unclear how MAOA promotes PCa progression. Here, we found that MAOA functions to induce epithelial-to-mesenchymal transition (EMT) and stabilize the transcription factor HIF1α, which mediates hypoxia through an elevation of ROS, thus enhancing growth, invasiveness, and metastasis of PCa cells. Knockdown and overexpression of MAOA in human PCa cell lines indicated that MAOA induces EMT through activation of VEGF and its coreceptor neuropilin-1. MAOA-dependent activation of neuropilin-1 promoted AKT/FOXO1/TWIST1 signaling, allowing FOXO1 binding at the TWIST1 promoter. Importantly, the MAOA-dependent HIF1α/VEGF-A/FOXO1/TWIST1 pathway was activated in high-grade PCa specimens, and knockdown of MAOA reduced or even eliminated prostate tumor growth and metastasis in PCa xenograft mouse models. Pharmacological inhibition of MAOA activity also reduced PCa xenograft growth in mice. Moreover, high MAOA expression in PCa tissues correlated with worse clinical outcomes in PCa patients. These findings collectively characterize the contribution of MAOA in PCa pathogenesis and suggest that MAOA has potential as a therapeutic target in PCa.


Biomaterials | 2015

Near-infrared fluorescence heptamethine carbocyanine dyes mediate imaging and targeted drug delivery for human brain tumor

Jason Boyang Wu; Changhong Shi; Gina Chia-Yi Chu; Qijin Xu; Yi Zhang; Qinlong Li; John S. Yu; Haiyen E. Zhau; Leland W.K. Chung

Brain tumors and brain metastases are among the deadliest malignancies of all human cancers, largely due to the cellular blood-brain and blood-tumor barriers that limit the delivery of imaging and therapeutic agents from the systemic circulation to tumors. Thus, improved strategies for brain tumor visualization and targeted treatment are critically needed. Here we identified and synthesized a group of near-infrared fluorescence (NIRF) heptamethine carbocyanine dyes and derivative NIRF dye-drug conjugates for effective imaging and therapeutic targeting of brain tumors of either primary or metastatic origin in mice, which is mechanistically mediated by tumor hypoxia and organic anion-transporting polypeptide genes. We also demonstrate that these dyes, when conjugated to chemotherapeutic agents such as gemcitabine, significantly restricted the growth of both intracranial glioma xenografts and prostate tumor brain metastases and prolonged survival in mice. These results show promise in the application of NIRF dyes as novel theranostic agents for the detection and treatment of brain tumors.


Oncotarget | 2016

SREBP-2 promotes stem cell-like properties and metastasis by transcriptional activation of c-Myc in prostate cancer.

Jason Boyang Wu; Qinlong Li; Katsumi Shigemura; Leland W.K. Chung; Wen-Chin Huang

Sterol regulatory element-binding protein-2 (SREBP-2) transcription factor mainly controls cholesterol biosynthesis and homeostasis in normal cells. The role of SREBP-2 in lethal prostate cancer (PCa) progression remains to be elucidated. Here, we showed that expression of SREBP-2 was elevated in advanced pathologic grade and metastatic PCa and significantly associated with poor clinical outcomes. Biofunctional analyses demonstrated that SREBP-2 induced PCa cell proliferation, invasion and migration. Furthermore, overexpression of SREBP-2 increased the PCa stem cell population, prostasphere-forming ability and tumor-initiating capability, whereas genetic silencing of SREBP-2 inhibited PCa cell growth, stemness, and xenograft tumor growth and metastasis. Clinical and mechanistic data showed that SREBP-2 was positively correlated with c-Myc and induced c-Myc activation by directly interacting with an SREBP-2-binding element in the 5′-flanking c-Myc promoter region to drive stemness and metastasis. Collectively, these clinical and experimental results reveal a novel role of SREBP-2 in the induction of a stem cell-like phenotype and PCa metastasis, which sheds light on translational potential by targeting SREBP-2 as a promising therapeutic approach in PCa.


Cancer Cell | 2017

MAOA-Dependent Activation of Shh-IL6-RANKL Signaling Network Promotes Prostate Cancer Metastasis by Engaging Tumor-Stromal Cell Interactions

Jason Boyang Wu; Lijuan Yin; Changhong Shi; Qinlong Li; Peng Duan; Jen-Ming Huang; Chunyan Liu; Fubo Wang; Michael Lewis; Yang Wang; Tzu-Ping Lin; Chin-Chen Pan; Edwin M. Posadas; Haiyen E. Zhau; Leland W.K. Chung

Metastasis is a predominant cause of death for prostate cancer (PCa) patients; however, the underlying mechanisms are poorly understood. We report that monoamine oxidase A (MAOA) is a clinically and functionally important mediator of PCa bone and visceral metastases, activating paracrine Shh signaling in tumor-stromal interactions. MAOA provides tumor cell growth advantages in the bone microenvironment by stimulating interleukin-6 (IL6) release from osteoblasts, and triggers skeletal colonization by activating osteoclastogenesis through osteoblast production of RANKL and IL6. MAOA inhibitor treatment effectively reduces metastasis and prolongs mouse survival by disengaging the Shh-IL6-RANKL signaling network in stromal cells in the tumor microenvironment. These findings provide a rationale for targeting MAOA and its associated molecules to treat PCa metastasis.


PLOS ONE | 2013

Convergent RANK- and c-Met-mediated signaling components predict survival of patients with prostate cancer: an interracial comparative study.

Peizhen Hu; Leland W.K. Chung; Dror Berel; Henry F. Frierson; Hua Yang; Chunyan Liu; Ruoxiang Wang; Qinlong Li; Andre Rogatko; Haiyen E. Zhau

We reported (PLoS One 6 (12):e28670, 2011) that the activation of c-Met signaling in RANKL-overexpressing bone metastatic LNCaP cell and xenograft models increased expression of RANK, RANKL, c-Met, and phosphorylated c-Met, and mediated downstream signaling. We confirmed the significance of the RANK-mediated signaling network in castration resistant clinical human prostate cancer (PC) tissues. In this report, we used a multispectral quantum dot labeling technique to label six RANK and c-Met convergent signaling pathway mediators simultaneously in formalin fixed paraffin embedded (FFPE) tissue specimens, quantify the intensity of each expression at the sub-cellular level, and investigated their potential utility as predictors of patient survival in Caucasian-American, African-American and Chinese men. We found that RANKL and neuropilin-1 (NRP-1) expression predicts survival of Caucasian-Americans with PC. A Gleason score ≥8 combined with nuclear p-c-Met expression predicts survival in African-American PC patients. Neuropilin-1, p-NF-κB p65 and VEGF are predictors for the overall survival of Chinese men with PC. These results collectively support interracial differences in cell signaling networks that can predict the survival of PC patients.


Asian Journal of Andrology | 2013

Interracial differences in prostate cancer progression among patients from the United States, China and Japan

Haiyen E. Zhau; Qinlong Li; Leland W.K. Chung

Although previous studies indicate interracial differences in prostate cancer epidemiology based on gene expression profiles among patients from the United States, China and Japan, evidence at the genetic and phenotypic levels that these differences exist and manifest along ethnic lines has been sparse. Recent studies, however, suggest that genetic differences, such as the lower incidence of Chinese prostate cancers harboring TMPRSS2-ERG translocations compared to patients from Western countries, should be carefully considered in the context of genotypic and phenotypic differences among interracial groups. New, more efficient technologies need to be developed to validate genetic, gene expression and/or phenotypic differences associated with prostate cancer tissue specimens obtained from interracial groups, to establish reliable clinical standards that take racial/ethnic data into account to improve the diagnosis, prognosis and treatment of patients with prostate cancer.


Oncotarget | 2016

Keratin 13 expression reprograms bone and brain metastases of human prostate cancer cells

Qinlong Li; Lijuan Yin; Lawrence W. Jones; Gina C.Y. Chu; Jason B-Y. Wu; Jen-Ming Huang; Quanlin Li; Sungyong You; Jayoung Kim; Yi-Tsung Lu; Stefan Mrdenovic; Ruoxiang Wang; Michael R. Freeman; Isla P. Garraway; Michael Lewis; Leland W.K. Chung; Haiyen E. Zhau

Lethal progression of prostate cancer metastasis can be improved by developing animal models that recapitulate the clinical conditions. We report here that cytokeratin 13 (KRT13), an intermediate filament protein, plays a directive role in prostate cancer bone, brain, and soft tissue metastases. KRT13 expression was elevated in bone, brain, and soft tissue metastatic prostate cancer cell lines and in primary and metastatic clinical prostate, lung, and breast cancer specimens. When KRT13 expression was determined at a single cell level in primary tumor tissues of 44 prostate cancer cases, KRT13 level predicted bone metastasis and the overall survival of prostate cancer patients. Genetically enforced KRT13 expression in human prostate cancer cell lines drove metastases toward mouse bone, brain and soft tissues through a RANKL-independent mechanism, as KRT13 altered the expression of genes associated with EMT, stemness, neuroendocrine/neuromimicry, osteomimicry, development, and extracellular matrices, but not receptor activator NF-κB ligand (RANKL) signaling networks in prostate cancer cells. Our results suggest new inhibitors targeting RANKL-independent pathways should be developed for the treatment of prostate cancer bone and soft tissue metastases.


The Prostate | 2015

Metastasis initiating cells in primary prostate cancer tissues from transurethral resection of the prostate (TURP) predicts castration‐resistant progression and survival of prostate cancer patients

Qinlong Li; Quanlin Li; Jill Nuccio; Chunyan Liu; Peng Duan; Ruoxiang Wang; Lawrence W. Jones; Leland W.K. Chung; Haiyen E. Zhau

We previously reported that the activation of RANK and c‐Met signaling components in both experimental mouse models and human prostate cancer (PC) specimens predicts bone metastatic potential and PC patient survival. This study addresses whether a population of metastasis‐initiating cells (MICs) known to express a stronger RANKL, phosphorylated c‐Met (p‐c‐Met), and neuropilin‐1 (NRP1) signaling network than bystander or dormant cells (BDCs) can be detected in PC tissues from patients subjected to transurethral resection of the prostate (TURP) for urinary obstruction prior to the diagnosis of PC with or without prior hormonal manipulation, and whether the relative abundance of MICs over BDCs could predict castration‐resistant progression and PC patient survival.


Molecular Cancer Research | 2018

R1 Regulates Prostate Tumor Growth and Progression By Transcriptional Suppression of the E3 Ligase HUWE1 to Stabilize c-Myc

Tzu-Ping Lin; Jingjing Li; Qinlong Li; Chunyan Liu; Ni Zeng; Jen-Ming Huang; Gina Chia-Yi Chu; Chi-Hung Lin; Haiyen E. Zhau; Leland W.K. Chung; Boyang Jason Wu; Jean C. Shih

Prostate cancer is a prevalent public health problem, especially because noncutaneous advanced malignant forms significantly affect the lifespan and quality of life of men worldwide. New therapeutic targets and approaches are urgently needed. The current study reports elevated expression of R1 (CDCA7L/RAM2/JPO2), a c-Myc–interacting protein and transcription factor, in human prostate cancer tissue specimens. In a clinical cohort, high R1 expression is associated with disease recurrence and decreased patient survival. Overexpression and knockdown of R1 in human prostate cancer cells indicate that R1 induces cell proliferation and colony formation. Moreover, silencing R1 dramatically reduces the growth of prostate tumor xenografts in mice. Mechanistically, R1 increases c-Myc protein stability by inhibiting ubiquitination and proteolysis through transcriptional suppression of HUWE1, a c-Myc–targeting E3 ligase, via direct interaction with a binding element in the promoter. Moreover, transcriptional repression is supported by a negative coexpression correlation between R1 and HUWE1 in a prostate cancer clinical dataset. Collectively, these findings, for the first time, characterize the contribution of R1 to prostate cancer pathogenesis. Implications: These findings provide evidence that R1 is a novel regulator of prostate tumor growth by stabilizing c-Myc protein, meriting further investigation of its therapeutic and prognostic potential.

Collaboration


Dive into the Qinlong Li's collaboration.

Top Co-Authors

Avatar

Leland W.K. Chung

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Haiyen E. Zhau

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jason Boyang Wu

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Changhong Shi

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Chunyan Liu

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Ruoxiang Wang

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Lijuan Yin

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Peizhen Hu

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Peng Duan

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Gina Chia-Yi Chu

Cedars-Sinai Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge