Network


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

Hotspot


Dive into the research topics where Yulong Chen is active.

Publication


Featured researches published by Yulong Chen.


Cancer Research | 2013

CXCR2 expression in tumor cells is a poor prognostic factor and promotes invasion and metastasis in lung adenocarcinoma

Pierre Saintigny; Erminia Massarelli; Steven H. Lin; Young Ho Ahn; Yulong Chen; Sangeeta Goswami; Baruch Erez; Michael S. O'Reilly; Diane Liu; J. Jack Lee; Li Zhang; Yuan Ping; Carmen Behrens; Luisa M.Solis Soto; John V. Heymach; Edward S. Kim; Roy S. Herbst; Scott M. Lippman; Ignacio I. Wistuba; Waun Ki Hong; Jonathan M. Kurie; Ja Seok Koo

CXCR2 in non-small cell lung cancer (NSCLC) has been studied mainly in stromal cells and is known to increase tumor inflammation and angiogenesis. Here, we examined the prognostic importance of CXCR2 in NSCLC and the role of CXCR2 and its ligands in lung cancer cells. The effect of CXCR2 expression on tumor cells was studied using stable knockdown clones derived from a murine KRAS/p53-mutant lung adenocarcinoma cell line with high metastatic potential and an orthotopic syngeneic mouse model and in vitro using a CXCR2 small-molecule antagonist (SB225002). CXCR2 protein expression was analyzed in tumor cells from 262 NSCLC. Gene expression profiles for CXCR2 and its ligands (CXCR2 axis) were analyzed in 52 human NSCLC cell lines and 442 human lung adenocarcinomas. Methylation of CXCR2 axis promoters was determined in 70 human NSCLC cell lines. Invasion and metastasis were decreased in CXCR2 knockdown clones in vitro and in vivo. SB225002 decreased invasion in vitro. In lung adenocarcinomas, CXCR2 expression in tumor cells was associated with smoking and poor prognosis. CXCR2 axis gene expression profiles in human NSCLC cell lines and lung adenocarcinomas defined a cluster driven by CXCL5 and associated with smoking, poor prognosis, and RAS pathway activation. Expression of CXCL5 was regulated by promoter methylation. The CXCR2 axis may be an important target in smoking-related lung adenocarcinoma.


Journal of Clinical Investigation | 2014

ZEB1 sensitizes lung adenocarcinoma to metastasis suppression by PI3K antagonism

Yanan Yang; Young Ho Ahn; Yulong Chen; Xiaochao Tan; Lixia Guo; Don L. Gibbons; Christin Ungewiss; David H. Peng; Xin Liu; Steven H. Lin; Nishan Thilaganathan; Ignacio I. Wistuba; Jaime Rodriguez-Canales; Georgia McLendon; Chad J. Creighton; Jonathan M. Kurie

Epithelial tumor cells that have undergone epithelial-to-mesenchymal transition (EMT) are typically prone to metastasis and drug resistance and contribute to a poor clinical outcome. The transcription factor ZEB1 is a known driver of EMT, and mediators of ZEB1 represent potential therapeutic targets for metastasis suppression. Here, we have shown that phosphatidylinositol 3-kinase-targeted (PI3K-targeted) therapy suppresses metastasis in a mouse model of Kras/Tp53-mutant lung adenocarcinoma that develops metastatic disease due to high expression of ZEB1. In lung adenocarcinoma cells from Kras/Tp53-mutant animals and human lung cancer cell lines, ZEB1 activated PI3K by derepressing miR-200 targets, including amphiregulin (AREG), betacellulin (BTC), and the transcription factor GATA6, which stimulated an EGFR/ERBB2 autocrine loop. Additionally, ZEB1-dependent derepression of the miR-200 and miR-183 target friend of GATA 2 (FOG2) enhanced GATA3-induced expression of the p110α catalytic subunit of PI3K. Knockdown of FOG2, p110α, and RHEB ameliorated invasive and metastatic propensities of tumor cells. Surprisingly, FOG2 was not required for mesenchymal differentiation, suggesting that mesenchymal differentiation and invasion are distinct and separable processes. Together, these results indicate that ZEB1 sensitizes lung adenocarcinoma cells to metastasis suppression by PI3K-targeted therapy and suggest that treatments to selectively modify the metastatic behavior of mesenchymal tumor cells are feasible and may be of clinical value.


Journal of Clinical Investigation | 2015

Lysyl hydroxylase 2 induces a collagen cross-link switch in tumor stroma

Yulong Chen; Masahiko Terajima; Yanan Yang; Li Sun; Young Ho Ahn; Daniela Pankova; Daniel S. Puperi; Takeshi Watanabe; Min P. Kim; Shanda H. Blackmon; Jaime Rodriguez; Hui Liu; Carmen Behrens; Ignacio I. Wistuba; Rosalba Minelli; Kenneth L. Scott; Johannah Sanchez-Adams; Farshid Guilak; Debananda Pati; Nishan Thilaganathan; Alan R. Burns; Chad J. Creighton; Elisabeth D. Martinez; Tomasz Zal; K. Jane Grande-Allen; Mitsuo Yamauchi; Jonathan M. Kurie

Epithelial tumor metastasis is preceded by an accumulation of collagen cross-links that heighten stromal stiffness and stimulate the invasive properties of tumor cells. However, the biochemical nature of collagen cross-links in cancer is still unclear. Here, we postulated that epithelial tumorigenesis is accompanied by changes in the biochemical type of collagen cross-links. Utilizing resected human lung cancer tissues and a p21CIP1/WAF1-deficient, K-rasG12D-expressing murine metastatic lung cancer model, we showed that, relative to normal lung tissues, tumor stroma contains higher levels of hydroxylysine aldehyde-derived collagen cross-links (HLCCs) and lower levels of lysine aldehyde-derived cross-links (LCCs), which are the predominant types of collagen cross-links in skeletal tissues and soft tissues, respectively. Gain- and loss-of-function studies in tumor cells showed that lysyl hydroxylase 2 (LH2), which hydroxylates telopeptidyl lysine residues on collagen, shifted the tumor stroma toward a high-HLCC, low-LCC state, increased tumor stiffness, and enhanced tumor cell invasion and metastasis. Together, our data indicate that LH2 enhances the metastatic properties of tumor cells and functions as a regulatory switch that controls the relative abundance of biochemically distinct types of collagen cross-links in the tumor stroma.


Molecular Cancer Research | 2016

Cancer-Associated Fibroblasts Induce a Collagen Cross-link Switch in Tumor Stroma

Daniela Pankova; Yulong Chen; Masahiko Terajima; Mark J. Schliekelman; Brandi N. Baird; Monica M. Fahrenholtz; Li Sun; Bartley J. Gill; Min P. Kim; Young Ho Ahn; Jonathon D. Roybal; Xin Liu; Edwin Roger Parra Cuentas; Jaime Rodriguez; Ignacio I. Wistuba; Chad J. Creighton; Don L. Gibbons; John Hicks; Mary E. Dickinson; Jennifer L. West; K. Jane Grande-Allen; Samir M. Hanash; Mitsuo Yamauchi; Jonathan M. Kurie

Intratumoral collagen cross-links heighten stromal stiffness and stimulate tumor cell invasion, but it is unclear how collagen cross-linking is regulated in epithelial tumors. To address this question, we used KrasLA1 mice, which develop lung adenocarcinomas from somatic activation of a KrasG12D allele. The lung tumors in KrasLA1 mice were highly fibrotic and contained cancer-associated fibroblasts (CAF) that produced collagen and generated stiffness in collagen gels. In xenograft tumors generated by injection of wild-type mice with lung adenocarcinoma cells alone or in combination with CAFs, the total concentration of collagen cross-links was the same in tumors generated with or without CAFs, but coinjected tumors had higher hydroxylysine aldehyde–derived collagen cross-links (HLCC) and lower lysine-aldehyde–derived collagen cross-links (LCCs). Therefore, we postulated that an LCC-to-HLCC switch induced by CAFs promotes the migratory and invasive properties of lung adenocarcinoma cells. To test this hypothesis, we created coculture models in which CAFs are positioned interstitially or peripherally in tumor cell aggregates, mimicking distinct spatial orientations of CAFs in human lung cancer. In both contexts, CAFs enhanced the invasive properties of tumor cells in three-dimensional (3D) collagen gels. Tumor cell aggregates that attached to CAF networks on a Matrigel surface dissociated and migrated on the networks. Lysyl hydroxylase 2 (PLOD2/LH2), which drives HLCC formation, was expressed in CAFs, and LH2 depletion abrogated the ability of CAFs to promote tumor cell invasion and migration. Implications: CAFs induce a collagen cross-link switch in tumor stroma to influence the invasive properties of tumor cells. Mol Cancer Res; 14(3); 287–95. ©2015 AACR.


PLOS ONE | 2013

Fibulin-2 is a driver of malignant progression in lung adenocarcinoma.

Brandi N. Baird; Mark J. Schliekelman; Young Ho Ahn; Yulong Chen; Jonathon D. Roybal; Bartley J. Gill; Dhruva K. Mishra; Baruch Erez; Michael S. O'Reilly; Yanan Yang; Mayuri Patel; Xin Liu; Nishan Thilaganathan; Irina V. Larina; Mary E. Dickinson; Jennifer L. West; Don L. Gibbons; Diane D. Liu; Min P. Kim; John Hicks; Ignacio I. Wistuba; Samir M. Hanash; Jonathan M. Kurie

The extracellular matrix of epithelial tumors undergoes structural remodeling during periods of uncontrolled growth, creating regional heterogeneity and torsional stress. How matrix integrity is maintained in the face of dynamic biophysical forces is largely undefined. Here we investigated the role of fibulin-2, a matrix glycoprotein that functions biomechanically as an inter-molecular clasp and thereby facilitates supra-molecular assembly. Fibulin-2 was abundant in the extracellular matrix of human lung adenocarcinomas and was highly expressed in tumor cell lines derived from mice that develop metastatic lung adenocarcinoma from co-expression of mutant K-ras and p53. Loss-of-function experiments in tumor cells revealed that fibulin-2 was required for tumor cells to grow and metastasize in syngeneic mice, a surprising finding given that other intra-tumoral cell types are known to secrete fibulin-2. However, tumor cells grew and metastasized equally well in Fbln2-null and -wild-type littermates, implying that malignant progression was dependent specifically upon tumor cell-derived fibulin-2, which could not be offset by other cellular sources of fibulin-2. Fibulin-2 deficiency impaired the ability of tumor cells to migrate and invade in Boyden chambers, to create a stiff extracellular matrix in mice, to cross-link secreted collagen, and to adhere to collagen. We conclude that fibulin-2 is a driver of malignant progression in lung adenocarcinoma and plays an unexpected role in collagen cross-linking and tumor cell adherence to collagen.


Journal of Biological Chemistry | 2016

Cyclophilin-B Modulates Collagen Cross-linking by Differentially Affecting Lysine Hydroxylation in the Helical and Telopeptidyl Domains of Tendon Type I Collagen

Masahiko Terajima; Yuki Taga; Yulong Chen; Wayne A. Cabral; Guo Hou-Fu; Sirivimol Srisawasdi; Masako Nagasawa; Noriko Sumida; Shunji Hattori; Jonathan M. Kurie; Joan C. Marini; Mitsuo Yamauchi

Covalent intermolecular cross-linking provides collagen fibrils with stability. The cross-linking chemistry is tissue-specific and determined primarily by the state of lysine hydroxylation at specific sites. A recent study on cyclophilin B (CypB) null mice, a model of recessive osteogenesis imperfecta, demonstrated that lysine hydroxylation at the helical cross-linking site of bone type I collagen was diminished in these animals (Cabral, W. A., Perdivara, I., Weis, M., Terajima, M., Blissett, A. R., Chang, W., Perosky, J. E., Makareeva, E. N., Mertz, E. L., Leikin, S., Tomer, K. B., Kozloff, K. M., Eyre, D. R., Yamauchi, M., and Marini, J. C. (2014) PLoS Genet. 10, e1004465). However, the extent of decrease appears to be tissue- and molecular site-specific, the mechanism of which is unknown. Here we report that although CypB deficiency resulted in lower lysine hydroxylation in the helical cross-linking sites, it was increased in the telopeptide cross-linking sites in tendon type I collagen. This resulted in a decrease in the lysine aldehyde-derived cross-links but generation of hydroxylysine aldehyde-derived cross-links. The latter were absent from the wild type and heterozygous mice. Glycosylation of hydroxylysine residues was moderately increased in the CypB null tendon. We found that CypB interacted with all lysyl hydroxylase isoforms (isoforms 1–3) and a putative lysyl hydroxylase-2 chaperone, 65-kDa FK506-binding protein. Tendon collagen in CypB null mice showed severe size and organizational abnormalities. The data indicate that CypB modulates collagen cross-linking by differentially affecting lysine hydroxylation in a site-specific manner, possibly via its interaction with lysyl hydroxylases and associated molecules. This study underscores the critical importance of collagen post-translational modifications in connective tissue formation.


Journal of Biological Chemistry | 2016

Lysyl Hydroxylase 2 Is Secreted By Tumor Cells and Can Modify Collagen in the Extracellular Space

Yulong Chen; Houfu Guo; Masahiko Terajima; Priyam Banerjee; Xin Liu; Jiang Yu; Amin Momin; Hiroyuki Katayama; Samir M. Hanash; Alan R. Burns; Gregg B. Fields; Mitsuo Yamauchi; Jonathan M. Kurie

Lysyl hydroxylase 2 (LH2) catalyzes the hydroxylation of lysine residues in the telopeptides of fibrillar collagens, which leads to the formation of stable collagen cross-links. Recently we reported that LH2 enhances the metastatic propensity of lung cancer by increasing the amount of stable hydroxylysine aldehyde-derived collagen cross-links (HLCCs), which generate a stiffer tumor stroma (Chen, Y., et al. (2015) J. Clin. Invest. 125, 125, 1147–1162). It is generally accepted that LH2 modifies procollagen α chains on the endoplasmic reticulum before the formation of triple helical procollagen molecules. Herein, we report that LH2 is also secreted and modifies collagen in the extracellular space. Analyses of lung cancer cell lines demonstrated that LH2 is present in the cell lysates and the conditioned media in a dimeric, active form in both compartments. LH2 co-localized with collagen fibrils in the extracellular space in human lung cancer specimens and in orthotopic lung tumors generated by injection of a LH2-expressing human lung cancer cell line into nude mice. LH2 depletion in MC3T3 osteoblastic cells impaired the formation of HLCCs, resulting in an increase in the unmodified lysine aldehyde-derived collagen cross-link (LCC), and the addition of recombinant LH2 to the media of LH2-deficient MC3T3 cells was sufficient to rescue HLCC formation in the extracellular matrix. The finding that LH2 modifies collagen in the extracellular space challenges the current view that LH2 functions solely on the endoplasmic reticulum and could also have important implications for cancer biology.


Quantum Well and Superlattice Physics | 1987

Temperature Dependent Optical Studies Of GaAs/AlGaAs Single Quantum Wells

Yulong Chen; Emil S. Koteles; Johnson Lee; J. Y. Chi; B. Elman

We report on detailed photoluminescence (PL) and photoluminescence excitation (PLE) studies of GaAs/A1GaAs single quantum wells (SQW) with differing well widths measured over temperatures ranging from 5K to 300K. A new trapping phenomenon was observed at low temperatures, which affects the linewidth and PL intensities (radiation lifetimes) of free excitons in the SQWs and which is related to the formation of bound excitons. At higher temperatures, excitonic linewidths were primarily broadened by optical phonon scattering. The temperature dependencies of the excitonic energies of SQWs were similar to that of bulk material and were independent of the quantum well width.


Proceedings of the National Academy of Sciences of the United States of America | 2018

Polo-like kinase 4 inhibition produces polyploidy and apoptotic death of lung cancers

Masanori Kawakami; Lisa Maria Mustachio; Lin Zheng; Yulong Chen; Jaime Rodriguez-Canales; Barbara Mino; Jonathan M. Kurie; Jason Roszik; Pamela Villalobos; Kelsie L. Thu; Jennifer Silvester; David W. Cescon; Ignacio I. Wistuba; Tak W. Mak; Xi Liu; Ethan Dmitrovsky

Significance Despite current treatments, lung cancers remain a major public health problem. Innovative ways are needed to treat or prevent these cancers. Centrosomes are critical for fidelity of mitosis. Abnormal centrosome numbers can cause aberrant mitosis and cell death. Polo-like kinase 4 (PLK4) regulates centriole duplication, and its deregulation alters centrosome number and mitosis. The potent PLK4 inhibitor CFI-400945 is reported here to exert marked antineoplastic effects against lung cancers. CDK2 inhibition also deregulates mitosis and was found to cooperate with PLK4 antagonism. CFI-400945 is now undergoing phase I clinical trial testing (NCT01954316). Taken together, targeting PLK4 for inhibition holds promise in lung cancer therapy either as a single agent or when combined with an agent that deregulates mitosis. Polo-like kinase 4 (PLK4) is a serine/threonine kinase regulating centriole duplication. CFI-400945 is a highly selective PLK4 inhibitor that deregulates centriole duplication, causing mitotic defects and death of aneuploid cancers. Prior work was substantially extended by showing CFI-400945 causes polyploidy, growth inhibition, and apoptotic death of murine and human lung cancer cells, despite expression of mutated KRAS or p53. Analysis of DNA content by propidium iodide (PI) staining revealed cells with >4N DNA content (polyploidy) markedly increased after CFI-400945 treatment. Centrosome numbers and mitotic spindles were scored. CFI-400945 treatment produced supernumerary centrosomes and mitotic defects in lung cancer cells. In vivo antineoplastic activity of CFI-400945 was established in mice with syngeneic lung cancer xenografts. Lung tumor growth was significantly inhibited at well-tolerated dosages. Phosphohistone H3 staining of resected lung cancers following CFI-400945 treatment confirmed the presence of aberrant mitosis. PLK4 expression profiles in human lung cancers were explored using The Cancer Genome Atlas (TCGA) and RNA in situ hybridization (RNA ISH) of microarrays containing normal and malignant lung tissues. PLK4 expression was significantly higher in the malignant versus normal lung and conferred an unfavorable survival (P < 0.05). Intriguingly, cyclin dependent kinase 2 (CDK2) antagonism cooperated with PLK4 inhibition. Taken together, PLK4 inhibition alone or as part of a combination regimen is a promising way to combat lung cancer.


Scientific Reports | 2017

FKBP65-dependent peptidyl-prolyl isomerase activity potentiates the lysyl hydroxylase 2-driven collagen cross-link switch

Yulong Chen; Masahiko Terajima; Priyam Banerjee; Houfu Guo; Xin Liu; Jiang Yu; Mitsuo Yamauchi; Jonathan M. Kurie

Bruck Syndrome is a connective tissue disease associated with inactivating mutations in lysyl hydroxylase 2 (LH2/PLOD2) or FK506 binding protein 65 (FKBP65/FKBP10). However, the functional relationship between LH2 and FKBP65 remains unclear. Here, we postulated that peptidyl prolyl isomerase (PPIase) activity of FKBP65 positively modulates LH2 enzymatic activity and is critical for the formation of hydroxylysine-aldehyde derived intermolecular collagen cross-links (HLCCs). To test this hypothesis, we analyzed collagen cross-links in Fkbp10-null and –wild-type murine embryonic fibroblasts. Although LH2 protein levels did not change, FKBP65 deficiency significantly diminished HLCCs and increased the non-hydroxylated lysine-aldehyde–derived collagen cross-links (LCCs), a pattern consistent with loss of LH2 enzymatic activity. The HLCC-to-LCC ratio was rescued in FKBP65-deficient murine embryonic fibroblasts by reconstitution with wild-type but not mutant FKBP65 that lacks intact PPIase domains. Findings from co-immunoprecipitation, protein-fragment complementation, and co-immunofluorescence assays showed that LH2 and FKBP65 are part of a common protein complex. We conclude that FKBP65 regulates LH2-mediated collagen cross-linking. Because LH2 promotes fibrosis and cancer metastasis, our findings suggest that pharmacologic strategies to target FKBP65 and LH2 may have complementary therapeutic activities.

Collaboration


Dive into the Yulong Chen's collaboration.

Top Co-Authors

Avatar

Jonathan M. Kurie

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Mitsuo Yamauchi

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Ignacio I. Wistuba

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Masahiko Terajima

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Xin Liu

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chad J. Creighton

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hou Fu Guo

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge