Network


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

Hotspot


Dive into the research topics where Chang-Lung Lee is active.

Publication


Featured researches published by Chang-Lung Lee.


Plant Physiology | 2004

Molecular Analyses of the Arabidopsis TUBBY-Like Protein Gene Family

Chia-Ping Lai; Chang-Lung Lee; Po-Hsuan Chen; Shu-Hsing Wu; Chien-Chih Yang; Jei-Fu Shaw

In mammals, TUBBY-like proteins play an important role in maintenance and function of neuronal cells during postdifferentiation and development. We have identified a TUBBY-like protein gene family with 11 members in Arabidopsis, named AtTLP1-11. Although seven of the AtTLP genes are located on chromosome I, no local tandem repeats or gene clusters are identified. Except for AtTLP4, reverse transcription-PCR analysis indicates that all these genes are expressed in various organs in 6-week-old Arabidopsis. AtTLP1, 2, 3, 6, 7, 9, 10, and 11 are expressed ubiquitously in all the organs tested, but the expression of AtTLP5 and 8 shows dramatic organ specificity. These 11 family members share 30% to 80% amino acid similarities across their conserved C-terminal tubby domains. Unlike the highly diverse N-terminal region of animal TUBBY-like proteins, all AtTLP members except AtTLP8 contain a conserved F-box domain (51–57 residues). The interaction between AtTLP9 and ASK1 (Arabidopsis Skp1-like 1) is confirmed via yeast (Saccharomyces cerevisiae) two-hybrid assays. Abscisic acid (ABA)-insensitive phenotypes are observed for two independent AtTLP9 mutant lines, whereas transgenic plants overexpressing AtTLP9 are hypersensitive to ABA. These results suggest that AtTLP9 may participate in the ABA signaling pathway.


Nature Medicine | 2014

A next-generation dual-recombinase system for time and host specific targeting of pancreatic cancer

Nina Schönhuber; Barbara Seidler; Kathleen Schuck; Christian Veltkamp; Christina Schachtler; Magdalena Zukowska; Stefan Eser; Thorsten B. Feyerabend; Mariel C. Paul; Philipp Eser; Sabine Klein; Andrew M. Lowy; Ruby Banerjee; Fangtang Yang; Chang-Lung Lee; Everett J. Moding; David G. Kirsch; Angelika Scheideler; Dario R. Alessi; Ignacio Varela; Allan Bradley; Alexander Kind; Angelika Schnieke; Hans Reimer Rodewald; Roland Rad; Roland M. Schmid; Günter Schneider; Dieter Saur

Genetically engineered mouse models (GEMMs) have dramatically improved our understanding of tumor evolution and therapeutic resistance. However, sequential genetic manipulation of gene expression and targeting of the host is almost impossible using conventional Cre-loxP–based models. We have developed an inducible dual-recombinase system by combining flippase-FRT (Flp-FRT) and Cre-loxP recombination technologies to improve GEMMs of pancreatic cancer. This enables investigation of multistep carcinogenesis, genetic manipulation of tumor subpopulations (such as cancer stem cells), selective targeting of the tumor microenvironment and genetic validation of therapeutic targets in autochthonous tumors on a genome-wide scale. As a proof of concept, we performed tumor cell–autonomous and nonautonomous targeting, recapitulated hallmarks of human multistep carcinogenesis, validated genetic therapy by 3-phosphoinositide-dependent protein kinase inactivation as well as cancer cell depletion and show that mast cells in the tumor microenvironment, which had been thought to be key oncogenic players, are dispensable for tumor formation.


Science Translational Medicine | 2016

A mouse-human phase 1 co-clinical trial of a protease-activated fluorescent probe for imaging cancer

Melodi Javid Whitley; Diana M. Cardona; Alexander L. Lazarides; Ivan Spasojevic; Jorge M. Ferrer; Joan Cahill; Chang-Lung Lee; Matija Snuderl; Dan G. Blazer; E. Shelley Hwang; Rachel A. Greenup; Paul J. Mosca; Jeffrey K. Mito; Kyle C. Cuneo; Nicole Larrier; Erin K. O’Reilly; Richard F. Riedel; William C. Eward; David B. Strasfeld; Dai Fukumura; Rakesh K. Jain; W. David Lee; Linda G. Griffith; Moungi G. Bawendi; David G. Kirsch; Brian E. Brigman

A first-in-human phase 1 clinical trial of the PEGylated protease-activated fluorescent probe, LUM015, enables tumor imaging at a safe and tolerable dose in humans. Protease probe tested in humans Cancer cells secrete more of the protease cathepsin than healthy cells, partly as a way to enzymatically remodel their surroundings for tumor growth and metastasis. Whitley et al. developed an imaging probe that could be activated in the presence of these cathepsins, thus allowing surgeons to distinguish tumor margins intraoperatively. Their probe, called LUM015, was able to signal the presence of cancer in vivo in a mouse sarcoma model, and in a so-called “co-clinical trial” in 15 patients, it was safe and cleaved as expected in different types of tumor tissues. With favorable biodistribution and pharmacokinetics also demonstrated, protease-activated probes are now poised for further adaptation to tumor resections, signaling the presence of residual cancer. Local recurrence is a common cause of treatment failure for patients with solid tumors. Intraoperative detection of microscopic residual cancer in the tumor bed could be used to decrease the risk of a positive surgical margin, reduce rates of reexcision, and tailor adjuvant therapy. We used a protease-activated fluorescent imaging probe, LUM015, to detect cancer in vivo in a mouse model of soft tissue sarcoma (STS) and ex vivo in a first-in-human phase 1 clinical trial. In mice, intravenous injection of LUM015 labeled tumor cells, and residual fluorescence within the tumor bed predicted local recurrence. In 15 patients with STS or breast cancer, intravenous injection of LUM015 before surgery was well tolerated. Imaging of resected human tissues showed that fluorescence from tumor was significantly higher than fluorescence from normal tissues. LUM015 biodistribution, pharmacokinetic profiles, and metabolism were similar in mouse and human subjects. Tissue concentrations of LUM015 and its metabolites, including fluorescently labeled lysine, demonstrated that LUM015 is selectively distributed to tumors where it is activated by proteases. Experiments in mice with a constitutively active PEGylated fluorescent imaging probe support a model where tumor-selective probe distribution is a determinant of increased fluorescence in cancer. These co-clinical studies suggest that the tumor specificity of protease-activated imaging probes, such as LUM015, is dependent on both biodistribution and enzyme activity. Our first-in-human data support future clinical trials of LUM015 and other protease-sensitive probes.


Journal of Clinical Investigation | 2014

MicroRNA-182 drives metastasis of primary sarcomas by targeting multiple genes

Mohit Sachdeva; Jeffrey K. Mito; Chang-Lung Lee; Minsi Zhang; Zhizhong Li; Rebecca D. Dodd; David Cason; Lixia Luo; Yan Ma; David Van Mater; Rebecca A. Gladdy; Dina Lev; Diana M. Cardona; David G. Kirsch

Metastasis causes most cancer deaths, but is incompletely understood. MicroRNAs can regulate metastasis, but it is not known whether a single miRNA can regulate metastasis in primary cancer models in vivo. We compared the expression of miRNAs in metastatic and nonmetastatic primary mouse sarcomas and found that microRNA-182 (miR-182) was markedly overexpressed in some tumors that metastasized to the lungs. By utilizing genetically engineered mice with either deletion of or overexpression of miR-182 in primary sarcomas, we discovered that deletion of miR-182 substantially decreased, while overexpression of miR-182 considerably increased, the rate of lung metastasis after amputation of the tumor-bearing limb. Additionally, deletion of miR-182 decreased circulating tumor cells (CTCs), while overexpression of miR-182 increased CTCs, suggesting that miR-182 regulates intravasation of cancer cells into the circulation. We identified 4 miR-182 targets that inhibit either the migration of tumor cells or the degradation of the extracellular matrix. Notably, restoration of any of these targets in isolation did not alter the metastatic potential of sarcoma cells injected orthotopically, but the simultaneous restoration of all 4 targets together substantially decreased the number of metastases. These results demonstrate that a single miRNA can regulate metastasis of primary tumors in vivo by coordinated regulation of multiple genes.


Cancer | 2012

Intraoperative detection and removal of microscopic residual sarcoma using wide-field imaging

Jeffrey K. Mito; Jorge Ferrer; Brian E. Brigman; Chang-Lung Lee; Rebecca D. Dodd; William C. Eward; Lisa F. Marshall; Kyle C. Cuneo; Jessica E. Carter; Shalini Ramasunder; Yongbaek Kim; W. David Lee; Linda G. Griffith; Moungi G. Bawendi; David G. Kirsch

The goal of limb‐sparing surgery for a soft tissue sarcoma of the extremity is to remove all malignant cells while preserving limb function. After initial surgery, microscopic residual disease in the tumor bed will cause a local recurrence in approximately 33% of patients with sarcoma. To help identify these patients, the authors developed an in vivo imaging system to investigate the suitability of molecular imaging for intraoperative visualization.


Science Signaling | 2012

p53 Functions in Endothelial Cells to Prevent Radiation-Induced Myocardial Injury in Mice

Chang-Lung Lee; Everett J. Moding; Kyle C. Cuneo; Yifan Li; Julie M. Sullivan; Lan Mao; Iman Washington; Laura B. Jeffords; Rafaela C. Rodrigues; Yan Ma; S Das; Christopher D. Kontos; Yongbaek Kim; Howard A. Rockman; David G. Kirsch

Without the transcription factor p53 in cardiac endothelial cells, the heart is more susceptible to radiation-induced damage. Keeping the Heart Safe from Radiation The cells that make up the muscular part of the heart, or the myocardium, are generally not actively dividing. Therefore, one would not expect radiation therapy aimed at eliminating rapidly dividing cancer cells to cause heart disease. However, radiation-related heart disease is a clinically important long-term side effect of radiation therapy. The transcription factor p53 is activated in response to radiation and other DNA-damaging stresses; whether it promotes or attenuates radiation-related heart disease is unclear. Lee et al. generated mice lacking p53 in endothelial cells, the cells that line blood vessels, and whole-heart irradiation in these mice resulted in damage to the vasculature of the heart, triggering ischemia (disrupted blood flow) in the myocardium and eventual heart failure. p53 inhibitors that protect normal cells from the effects of radiation therapy have been proposed as an approach to improve the therapeutic ratio of radiation therapy, but these data suggest that combining radiation therapy with p53 inhibitors may actually increase the risk of cardiac injury. Radiation therapy, which is used for the treatment of some cancers, can cause delayed heart damage. In the heart, p53 influences myocardial injury that occurs after multiple types of stress. Here, we demonstrated that p53 functioned in endothelial cells to protect mice from myocardial injury after whole-heart irradiation. Mice with an endothelial cell–specific deletion of p53 succumbed to heart failure after whole-heart irradiation as a result of myocardial necrosis, systolic dysfunction, and cardiac hypertrophy. Moreover, the onset of cardiac dysfunction was preceded by alterations in myocardial vascular permeability and density, which resulted in cardiac ischemia and myocardial hypoxia. Mechanistic studies with primary cardiac endothelial cells irradiated in vitro indicated that p53 signaling caused mitotic arrest and protected cardiac endothelial cells from cell death resulting from abnormal mitosis or mitotic catastrophe. Furthermore, mice lacking the cyclin-dependent kinase inhibitor p21, which is a transcriptional target of p53, were also sensitized to myocardial injury after whole-heart irradiation. Together, our results demonstrate that the p53-p21 axis functions to prevent radiation-induced myocardial injury in mice.


Journal of Clinical Investigation | 2014

Atm deletion with dual recombinase technology preferentially radiosensitizes tumor endothelium

Everett J. Moding; Chang-Lung Lee; Katherine D. Castle; Patrick Oh; Lan Mao; Shan Zha; Hooney Min; Yan Ma; S Das; David G. Kirsch

Cells isolated from patients with ataxia telangiectasia are exquisitely sensitive to ionizing radiation. Kinase inhibitors of ATM, the gene mutated in ataxia telangiectasia, can sensitize tumor cells to radiation therapy, but concern that inhibiting ATM in normal tissues will also increase normal tissue toxicity from radiation has limited their clinical application. Endothelial cell damage can contribute to the development of long-term side effects after radiation therapy, but the role of endothelial cell death in tumor response to radiation therapy remains controversial. Here, we developed dual recombinase technology using both FlpO and Cre recombinases to generate primary sarcomas in mice with endothelial cell-specific deletion of Atm to determine whether loss of Atm in endothelial cells sensitizes tumors and normal tissues to radiation. Although deletion of Atm in proliferating tumor endothelial cells enhanced the response of sarcomas to radiation, Atm deletion in quiescent endothelial cells of the heart did not sensitize mice to radiation-induced myocardial necrosis. Blocking cell cycle progression reversed the effect of Atm loss on tumor endothelial cell radiosensitivity. These results indicate that endothelial cells must progress through the cell cycle in order to be radiosensitized by Atm deletion.


Science Translational Medicine | 2015

Tumor cells, but not endothelial cells, mediate eradication of primary sarcomas by stereotactic body radiation therapy

Everett J. Moding; Katherine D. Castle; Bradford A. Perez; Patrick Oh; Hooney Min; Hannah Norris; Yan Ma; Diana M. Cardona; Chang-Lung Lee; David G. Kirsch

Tumor cells, rather than endothelial cells, are critical targets that regulate primary sarcoma eradication by radiation therapy. Not all cells are eradicated equally Personalized cancer therapies dominate the news. But radiation therapy continues to be an essential part of the treatment regimens of nearly half of all cancer patients—sometimes achieving complete tumor regression through the safe delivery of high doses of radiation. Previous research with transplanted tumor models in mice has suggest that radiation targets, not only the tumor cells themselves, but also components of the surrounding milieu, which comprises blood vessels and various cell types that influence tumor growth. Now Moding et al. challenge the earlier findings in studies conducted with primary sarcomas in mice that carried, in either tumor or endothelial cells, genetic mutations that modulate radiation sensitivity. The authors found that it was the tumor, rather than endothelial, cells that mediate primary sarcoma shrinkage by radiation therapy and that selective small-molecule inhibition of a DNA-damage response enzyme can enhance radiosensitization of some tumors. Cancer clinics currently use high-dose stereotactic body radiation therapy as a curative treatment for several kinds of cancers. However, the contribution of vascular endothelial cells to tumor response to radiation remains controversial. Using dual recombinase technology, we generated primary sarcomas in mice with targeted genetic mutations specifically in tumor cells or endothelial cells. We selectively mutated the proapoptotic gene Bax or the DNA damage response gene Atm to genetically manipulate the radiosensitivity of endothelial cells in primary soft tissue sarcomas. Bax deletion from endothelial cells did not affect radiation-induced cell death in tumor endothelial cells or sarcoma response to radiation therapy. Although Atm deletion increased endothelial cell death after radiation therapy, deletion of Atm from endothelial cells failed to enhance sarcoma eradication. In contrast, deletion of Atm from tumor cells increased sarcoma eradication by radiation therapy. These results demonstrate that tumor cells, rather than endothelial cells, are critical targets that regulate sarcoma eradication by radiation therapy. Treatment with BEZ235, a small-molecule protein kinase inhibitor, radiosensitized primary sarcomas more than the heart. These results suggest that inhibiting ATM kinase during radiation therapy is a viable strategy for radiosensitization of some tumors.


Translational cancer research | 2013

Role of p53 in regulating tissue response to radiation by mechanisms independent of apoptosis

Chang-Lung Lee; Jordan M. Blum; David G. Kirsch

Radiation exposure leads to diverse outcomes in vivo across different tissues and even within the same cell lineage. The diversity of radiation response in vivo is at least partially attributable to the status of the tumor suppressor p53, a master regulator of cellular response to stress, and activation of its transcriptional targets. In certain cells, such as hematopoietic progenitors and transit amplifying cells in the gastrointestinal epithelium, activation of p53 by radiation triggers the intrinsic pathway of apoptosis. However, in many other cells, activation of p53 by radiation does not result in apoptosis, which underscores the importance of understanding the role of p53 in regulating radiation response through alternative mechanisms. In this review, we summarize recent studies using genetically engineered mice to dissect the role of p53 in 1) cells where its activation is dissociated from the intrinsic pathway of apoptosis, such as hematopoietic stem cells and vascular endothelial cells and 2) tissues where activation of the intrinsic pathway of apoptosis does not promote the acute radiation syndrome, such as the gastrointestinal epithelium. We highlight findings showing that the apoptosis-independent response of p53 to radiation in vivo can contribute to death or survival in a cell-type dependent manner, which underscores the complexity by which p53 regulates the cellular and tissue response to radiation.


Disease Models & Mechanisms | 2012

Generation of primary tumors with Flp recombinase in FRT-flanked p53 mice

Chang-Lung Lee; Everett J. Moding; Xiaofang Huang; Yifan Li; Loretta Z. Woodlief; Rafaela C. Rodrigues; Yan Ma; David G. Kirsch

SUMMARY The site-specific recombinases Cre and Flp can mutate genes in a spatially and temporally restricted manner in mice. Conditional recombination of the tumor suppressor gene p53 using the Cre-loxP system has led to the development of multiple genetically engineered mouse models of human cancer. However, the use of Cre recombinase to initiate tumors in mouse models limits the utilization of Cre to genetically modify other genes in tumor stromal cells in these models. To overcome this limitation, we inserted FRT (flippase recognition target) sites flanking exons 2–6 of the endogenous p53 gene in mice to generate a p53FRT allele that can be deleted by Flp recombinase. We show that FlpO-mediated deletion of p53 in mouse embryonic fibroblasts impairs the p53-dependent response to genotoxic stress in vitro. In addition, using FSF-KrasG12D/+; p53FRT/FRT mice, we demonstrate that an adenovirus expressing FlpO recombinase can initiate primary lung cancers and sarcomas in mice. p53FRT mice will enable dual recombinase technology to study cancer biology because Cre is available to modify genes specifically in stromal cells to investigate their role in tumor development, progression and response to therapy.

Collaboration


Dive into the Chang-Lung Lee'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