Network


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

Hotspot


Dive into the research topics where Baojun Chang is active.

Publication


Featured researches published by Baojun Chang.


Journal of Clinical Investigation | 2012

Endothelial epsin deficiency decreases tumor growth by enhancing VEGF signaling.

Satish Pasula; Xiaofeng Cai; Yunzhou Dong; Mirko Messa; John McManus; Baojun Chang; Xiaolei Liu; Hua Zhu; Robert Silasi Mansat; Seon Joo Yoon; Scott Hahn; Jacob Keeling; Debra Saunders; Genevieve Ko; John Knight; Gail Newton; Francis W. Luscinskas; Xiaohong Sun; Rheal A. Towner; Florea Lupu; Lijun Xia; Ottavio Cremona; Pietro De Camilli; Wang Min; Hong Chen

Epsins are a family of ubiquitin-binding, endocytic clathrin adaptors. Mice lacking both epsins 1 and 2 (Epn1/2) die at embryonic day 10 and exhibit an abnormal vascular phenotype. To examine the angiogenic role of endothelial epsins, we generated mice with constitutive or inducible deletion of Epn1/2 in vascular endothelium. These mice exhibited no abnormal phenotypes under normal conditions, suggesting that lack of endothelial epsins 1 and 2 did not affect normal blood vessels. In tumors, however, loss of epsins 1 and 2 resulted in disorganized vasculature, significantly increased vascular permeability, and markedly retarded tumor growth. Mechanistically, we show that VEGF promoted binding of epsin to ubiquitinated VEGFR2. Loss of epsins 1 and 2 specifically impaired endocytosis and degradation of VEGFR2, which resulted in excessive VEGF signaling that compromised tumor vascular function by exacerbating nonproductive leaky angiogenesis. This suggests that tumor vasculature requires a balance in VEGF signaling to provide sufficient productive angiogenesis for tumor development and that endothelial epsins 1 and 2 negatively regulate the output of VEGF signaling. Promotion of excessive VEGF signaling within tumors via a block of epsin 1 and 2 function may represent a strategy to prevent normal angiogenesis in cancer patients who are resistant to anti-VEGF therapies.


Science Signaling | 2014

Temporal and spatial regulation of epsin abundance and VEGFR3 signaling are required for lymphatic valve formation and function

Xiaolei Liu; Satish Pasula; Hoogeun Song; Kandice L. Tessneer; Yunzhou Dong; Scott Hahn; Tadayuki Yago; Megan L. Brophy; Baojun Chang; Xiaofeng Cai; Hao Wu; John McManus; Hirotake Ichise; Constantin Georgescu; Jonathan D. Wren; Courtney T. Griffin; Lijun Xia; R. Sathish Srinivasan; Hong Chen

Proteins involved in endocytosis promote the internalization and degradation of VEGFR3, ensuring valve formation in lymphatic vessels. Ensuring a One-Way Flow of Lymph Lymphatic vessels return lymph, which consists of fluid and mostly immune cells that go into the tissues from the blood, back to the circulation. Valves in lymphatic vessels ensure that lymph flows in one direction into veins and prevent the accumulation of lymph in tissues, a condition called edema. The receptor VEGFR3 is required for lymphatic vessel development. Liu et al. noted that the regions of developing lymphatic vessels that became valves had high VEGFR3 amounts but low amounts of epsin 1 and 2, proteins that are involved in endocytosis. They determined that epsin 1 and 2 suppressed VEGFR3 signaling in collecting lymphatic trunks by triggering endocytosis and degradation of VEGFR3. Furthermore, mice lacking epsin 1 and 2 in the endothelial cells that line lymphatic vessels had defective lymphatic valves and impaired drainage. Lymphatic valve formation was restored in these mice by deletion of a single allele of Vegfr3 or treatment with a VEGFR3 inhibitor. Lymphatic valves prevent the backflow of the lymph fluid and ensure proper lymphatic drainage throughout the body. Local accumulation of lymphatic fluid in tissues, a condition called lymphedema, is common in individuals with malformed lymphatic valves. The vascular endothelial growth factor receptor 3 (VEGFR3) is required for the development of lymphatic vascular system. The abundance of VEGFR3 in collecting lymphatic trunks is high before valve formation and, except at valve regions, decreases after valve formation. We found that in mesenteric lymphatics, the abundance of epsin 1 and 2, which are ubiquitin-binding adaptor proteins involved in endocytosis, was low at early stages of development. After lymphatic valve formation, the initiation of steady shear flow was associated with an increase in the abundance of epsin 1 and 2 in collecting lymphatic trunks, but not in valve regions. Epsin 1 and 2 bound to VEGFR3 and mediated the internalization and degradation of VEGFR3, resulting in termination of VEGFR3 signaling. Mice with lymphatic endothelial cell–specific deficiency of epsin 1 and 2 had dilated lymphatic capillaries, abnormally high VEGFR3 abundance in collecting lymphatics, immature lymphatic valves, and defective lymph drainage. Deletion of a single Vegfr3 allele or pharmacological suppression of VEGFR3 signaling restored normal lymphatic valve development and lymph drainage in epsin-deficient mice. Our findings establish a critical role for epsins in the temporal and spatial regulation of VEGFR3 abundance and signaling in collecting lymphatic trunks during lymphatic valve formation.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2014

Genetic Reduction of Vascular Endothelial Growth Factor Receptor 2 Rescues Aberrant Angiogenesis Caused by Epsin Deficiency

Kandice L. Tessneer; Satish Pasula; Xiaofeng Cai; Yunzhou Dong; John McManus; Xiaolei Liu; Lili Yu; Scott Hahn; Baojun Chang; Yiyuan Chen; Courtney T. Griffin; Lijun Xia; Ralf H. Adams; Hong Chen

Objective—We previously showed that endothelial epsin deficiency caused elevated vascular endothelial growth factor receptor 2 (VEGFR2) and enhanced VEGF signaling, resulting in aberrant tumor angiogenesis and reduced tumor growth in adult mice. However, direct evidence demonstrating that endothelial epsins regulate angiogenesis specifically through VEGFR2 downregulation is still lacking. In addition, whether the lack of epsins causes abnormal angiogenesis during embryonic development remains unclear. Approach and Results—A novel strain of endothelial epsin-deleted mice that are heterozygous for VEGFR2 (Epn1fl/fl; Epn2−/−; Flkfl/+; iCDH5 Cre mice) was created. Analysis of embryos at different developmental stages showed that deletion of epsins caused defective embryonic angiogenesis and retarded embryo development. In vitro angiogenesis assays using isolated primary endothelial cells (ECs) from Epn1fl/fl; Epn2−/−; iCDH5 Cre (EC-iDKO) and Epn1fl/fl; Epn2−/−; Flkfl/+; iCDH5 Cre (EC-iDKO-Flkfl/+) mice demonstrated that VEGFR2 reduction in epsin-depleted cells was sufficient to restore normal VEGF signaling, EC proliferation, EC migration, and EC network formation. These findings were complemented by in vivo wound healing, inflammatory angiogenesis, and tumor angiogenesis assays in which reduction of VEGFR2 was sufficient to rescue abnormal angiogenesis in endothelial epsin-deleted mice. Conclusions—Our results provide the first genetic demonstration that epsins function specifically to downregulate VEGFR2 by mediating activated VEGFR2 internalization and degradation and that genetic reduction of VEGFR2 level protects against excessive angiogenesis caused by epsin loss. Our findings indicate that epsins may be a potential therapeutic target in conditions in which tightly regulated angiogenesis is crucial, such as in diabetic wound healing and tumors.


Journal of Cancer Research Updates | 2013

Epsin Family of Endocytic Adaptor Proteins as Oncogenic Regulators of Cancer Progression.

Kandice L. Tessneer; Xiaofeng Cai; Satish Pasula; Yunzhou Dong; Xiaolei Liu; Baojun Chang; John McManus; Scott Hahn; Lili Yu; Hong Chen

Tumor angiogenesis, tumor cell proliferation, and tumor cell migration result from an accumulation of oncogenic mutations that alter protein expression and the regulation of various signaling cascades. Epsins, a small family of clathrin-mediated endocytic adaptor proteins, are reportedly upregulated in a variety of cancers. Importantly, loss of epsins protects against tumorigenesis, thus supporting an oncogenic role for epsins in cancer. Although a clear relationship between epsins and cancer has evolved, the importance of this relationship with regards to cancer progression and anti-cancer therapies remains unclear. In this review, we summarize epsins’ role as endocytic adaptors that modulate VEGF and Notch signaling through the regulated internalization of VEGFR2 and trans-endocytosis of Notch receptors. As both VEGF and Notch signaling have significant implications in angiogenesis, we focus on the newly identified role for epsins in tumor angiogenesis. In addition to epsins’ canonical role in receptor-mediated endocytosis, and the resulting downstream signaling regulation, we discuss the non-canonical role of epsins as regulators of small GTPases and the implications this has on tumor cell proliferation and invasion. Given epsins’ identified roles in tumor angiogenesis, tumor cell proliferation, and tumor cell invasion, we predict that the investigative links between epsins and cancer will provide new insights into the importance of endocytic adaptors and their potential use as future therapeutic targets.


Nature Communications | 2015

Epsin is required for Dishevelled stability and Wnt signalling activation in colon cancer development.

Baojun Chang; Kandice L. Tessneer; John McManus; Xiaolei Liu; Scott Hahn; Satish Pasula; Hao Wu; Hoogeun Song; Yiyuan Chen; Xiaofeng Cai; Yunzhou Dong; Megan L. Brophy; Ruby Rahman; Jian Xing Ma; Lijun Xia; Hong Chen

Uncontrolled canonical Wnt signaling supports colon epithelial tumor expansion and malignant transformation. Understanding the regulatory mechanisms involved is crucial for elucidating the pathogenesis of and will provide new therapeutic targets for colon cancer. Epsins are ubiquitin-binding adaptor proteins upregulated in several human cancers; however, epsins’ involvement in colon cancer is unknown. Here we show that loss of intestinal epithelial epsins protects against colon cancer by significantly reducing the stability of the crucial Wnt signaling effector, dishevelled (Dvl2), and impairing Wnt signaling. Consistently, epsins and Dvl2 are correspondingly upregulated in colon cancer. Mechanistically, epsin binds Dvl2 via its epsin N-terminal homology domain and ubiquitin-interacting motifs and prohibits Dvl2 polyubiquitination and degradation. Our findings reveal an unconventional role for epsins in stabilizing Dvl2 and potentiating Wnt signaling in colon cancer cells to ensure robust colon cancer progression. Epsins’ pro-carcinogenic role suggests they are potential therapeutic targets to combat colon cancer.


Circulation Research | 2016

Selective targeting of a novel Epsin-VEGFR2 interaction promotes VEGF-mediated angiogenesis

H. N. Ashiqur Rahman; Hao Wu; Yunzhou Dong; Satish Pasula; Aiyun Wen; Ye Sun; Megan L. Brophy; Kandice L. Tessneer; Xiaofeng Cai; John McManus; Baojun Chang; Sukyoung Kwak; Negar S. Rahman; Wenjia Xu; Conrad Fernandes; John Michael Mcdaniel; Lijun Xia; Lois E. H. Smith; R. Sathish Srinivasan; Hong Chen

RATIONALE We previously reported that vascular endothelial growth factor (VEGF)-induced binding of VEGF receptor 2 (VEGFR2) to epsins 1 and 2 triggers VEGFR2 degradation and attenuates VEGF signaling. The epsin ubiquitin interacting motif (UIM) was shown to be required for the interaction with VEGFR2. However, the molecular determinants that govern how epsin specifically interacts with and regulates VEGFR2 were unknown. OBJECTIVE The goals for the present study were as follows: (1) to identify critical molecular determinants that drive the specificity of the epsin and VEGFR2 interaction and (2) to ascertain whether such determinants were critical for physiological angiogenesis in vivo. METHODS AND RESULTS Structural modeling uncovered 2 novel binding surfaces within VEGFR2 that mediate specific interactions with epsin UIM. Three glutamic acid residues in epsin UIM were found to interact with residues in VEGFR2. Furthermore, we found that the VEGF-induced VEGFR2-epsin interaction promoted casitas B-lineage lymphoma-mediated ubiquitination of epsin, and uncovered a previously unappreciated ubiquitin-binding surface within VEGFR2. Mutational analysis revealed that the VEGFR2-epsin interaction is supported by VEGFR2 interacting specifically with the UIM and with ubiquitinated epsin. An epsin UIM peptide, but not a mutant UIM peptide, potentiated endothelial cell proliferation, migration and angiogenic properties in vitro, increased postnatal retinal angiogenesis, and enhanced VEGF-induced physiological angiogenesis and wound healing. CONCLUSIONS Distinct residues in the epsin UIM and VEGFR2 mediate specific interactions between epsin and VEGFR2, in addition to UIM recognition of ubiquitin moieties on VEGFR2. These novel interactions are critical for pathophysiological angiogenesis, suggesting that these sites could be selectively targeted by therapeutics to modulate angiogenesis.


International Scholarly Research Notices | 2013

Endocytic Adaptor Protein Epsin Is Elevated in Prostate Cancer and Required for Cancer Progression

Kandice L. Tessneer; Satish Pasula; Xiaofeng Cai; Yunzhou Dong; Xiaolei Liu; Lili Yu; Scott Hahn; John McManus; Yiyuan Chen; Baojun Chang; Hong Chen

Epsins have an important role in mediating clathrin-mediated endocytosis of ubiquitinated cell surface receptors. The potential role for epsins in tumorigenesis and cancer metastasis by regulating intracellular signaling pathways has largely not been explored. Epsins are reportedly upregulated in several types of cancer including human skin, lung, and canine mammary cancers. However, whether their expression is elevated in prostate cancer is unknown. In this study, we investigated the potential role of epsins in prostate tumorigenesis using the wild type or epsin-deficient human prostate cancer cells, LNCaP, in a human xenograft model, and the spontaneous TRAMP mouse model in wild type or epsin-deficient background. Here, we reported that the expression of epsins 1 and 2 is upregulated in both human and mouse prostate cancer cells and cancerous tissues. Consistent with upregulation of epsins in prostate tumors, we discovered that depletion of epsins impaired tumor growth in both the human LNCaP xenograft and the TRAMP mouse prostate. Furthermore, epsin depletion significantly prolonged survival in the TRAMP mouse model. In summary, our findings suggest that epsins may act as oncogenic proteins to promote prostate tumorigenesis and that depletion or inhibition of epsins may provide a novel therapeutic target for future prostate cancer therapies.


Journal of Clinical Investigation | 2015

Motif mimetic of epsin perturbs tumor growth and metastasis

Yunzhou Dong; Hao Wu; H. N. Ashiqur Rahman; Yanjun Liu; Satish Pasula; Kandice L. Tessneer; Xiaofeng Cai; Xiaolei Liu; Baojun Chang; John McManus; Scott Hahn; Jiali Dong; Megan L. Brophy; Lili Yu; Kai Song; Robert Silasi-Mansat; Debra Saunders; Charity Njoku; Hoogeun Song; Padmaja Mehta-D’souza; Rheal A. Towner; Florea Lupu; Rodger P. McEver; Lijun Xia; Derek Boerboom; R. Sathish Srinivasan; Hong Chen

Tumor angiogenesis is critical for cancer progression. In multiple murine models, endothelium-specific epsin deficiency abrogates tumor progression by shifting the balance of VEGFR2 signaling toward uncontrolled tumor angiogenesis, resulting in dysfunctional tumor vasculature. Here, we designed a tumor endothelium-targeting chimeric peptide (UPI) for the purpose of inhibiting endogenous tumor endothelial epsins by competitively binding activated VEGFR2. We determined that the UPI peptide specifically targets tumor endothelial VEGFR2 through an unconventional binding mechanism that is driven by unique residues present only in the epsin ubiquitin-interacting motif (UIM) and the VEGFR2 kinase domain. In murine models of neoangiogenesis, UPI peptide increased VEGF-driven angiogenesis and neovascularization but spared quiescent vascular beds. Further, in tumor-bearing mice, UPI peptide markedly impaired functional tumor angiogenesis, tumor growth, and metastasis, resulting in a notable increase in survival. Coadministration of UPI peptide with cytotoxic chemotherapeutics further sustained tumor inhibition. Equipped with localized tumor endothelium-specific targeting, our UPI peptide provides potential for an effective and alternative cancer therapy.


Journal of Clinical Investigation | 2018

Epsin deficiency promotes lymphangiogenesis through regulation of VEGFR3 degradation in diabetes

Hao Wu; H. N. Ashiqur Rahman; Yunzhou Dong; Xiaolei Liu; Yang Lee; Aiyun Wen; Kim H.T. To; Li Xiao; Amy E. Birsner; Lauren Bazinet; Scott Wong; Kai Song; Megan L. Brophy; M. Riaj Mahamud; Baojun Chang; Xiaofeng Cai; Satish Pasula; Sukyoung Kwak; Wenxia Yang; Joyce Bischoff; Jian Xu; Diane R. Bielenberg; J. Brandon Dixon; Robert J. D’Amato; R. Sathish Srinivasan; Hong Chen

Impaired lymphangiogenesis is a complication of chronic complex diseases, including diabetes. VEGF-C/VEGFR3 signaling promotes lymphangiogenesis, but how this pathway is affected in diabetes remains poorly understood. We previously demonstrated that loss of epsins 1 and 2 in lymphatic endothelial cells (LECs) prevented VEGF-C–induced VEGFR3 from endocytosis and degradation. Here, we report that diabetes attenuated VEGF-C–induced lymphangiogenesis in corneal micropocket and Matrigel plug assays in WT mice but not in mice with inducible lymphatic-specific deficiency of epsins 1 and 2 (LEC-iDKO). Consistently, LECs isolated from diabetic LEC-iDKO mice elevated in vitro proliferation, migration, and tube formation in response to VEGF-C over diabetic WT mice. Mechanistically, ROS produced in diabetes induced c-Src–dependent but VEGF-C–independent VEGFR3 phosphorylation, and upregulated epsins through the activation of transcription factor AP-1. Augmented epsins bound to and promoted degradation of newly synthesized VEGFR3 in the Golgi, resulting in reduced availability of VEGFR3 at the cell surface. Preclinically, the loss of lymphatic-specific epsins alleviated insufficient lymphangiogenesis and accelerated the resolution of tail edema in diabetic mice. Collectively, our studies indicate that inhibiting expression of epsins in diabetes protects VEGFR3 against degradation and ameliorates diabetes-triggered inhibition of lymphangiogenesis, thereby providing a novel potential therapeutic strategy to treat diabetic complications.


Cancer Research | 2013

Abstract 5128: Upregulation of epsin in breast cancer and critical role of epsin in promoting cancer growth and metastasis.

Xiaofeng Cai; Satish Pasula; Baojun Chang; Scott Hahn; John McManus; Hong Chen

Proceedings: AACR 104th Annual Meeting 2013; Apr 6-10, 2013; Washington, DC We have previously demonstrated that deficiency of epsins, a family of endocytic clathrin adaptor proteins, decreases tumor growth by enhancing VEGF signaling in endothelial cells and causing dysfunctional tumor angiogenesis. However, the role of epsin in cancer cells is unclear. Our preliminary data indicate that epsins are upregulated in various human cancers, including breast cancer, prostate cancer, ovary cancer, cervix cancer, bladder cancer, and esophagus cancer. Here, we determined the role of epsin in cancer cells in regulating breast cancer growth and metastasis. Increased epsin level was found in ER(+/-) and/or PR(+/-) human breast cancer cell lines including MDA-MB-231, BT-20, ZR751, MCF7 and T47D, and in mouse breast cancer cell lines, such as 168FARN and 4T1. The level of epsins was also considerably augmented in tumors derived from MMTV-PyMT spontaneous breast cancer mouse model at different progressive stages. We observed a correlative increase in epsin expression with cancer progression in MMTV-PyMT mice. We also generated stable epsin-deficient MDA-MB-231 and 4T1 cells. Epsin deficiency markedly inhibited cell proliferation and migration in vitro. Xenograft mouse models of epsin-deficient breast cancer cells revealed striking reduction in tumorigenesis and lung metastasis. Lung metastasis by tail-vein injection of epsin-deficient MDA-MB-231 cells was also decreased compared to controls. Mechanistically, we showed that epsin-deficiency induced morphological change from mesenchamal-like to epitheial-like in MDA-MB-231 cells, indicating a reversal of Epithelial-Mesenchymal Transition (EMT) to Mesenchymal-Epithelial-Transition (MET). Moreover, epsin-deficiency increased E-cadherin expression but decreased vimentin, and EMT inducing genes snail/slug expression in MDA-MB-231 cells. Epsin-deficiency also dramatically reduced cyclin D1 expression and Rb activation. Accordingly, elevated E-cadherin but attenuated vimentin, slug and cyclin D1 were observed in tumor tissues or lung tissues isolated from epsin-deficient MDA-MB-231 and 4T1 tumor models. Conversely, overexpression of epsin in MDA-MB-231 cells enhanced cyclin D1 and vimentin expression. Collectively, epsin promotes breast tumor growth and metastasis through coordinated regulation of cell cycle and EMT. Our study provides a basis for novel therapeutic targets for anti-cancer treatments. Citation Format: Xiaofeng Cai, Satish Pasula, Baojun Chang, Scott Hahn, John McManus, Hong Chen. Upregulation of epsin in breast cancer and critical role of epsin in promoting cancer growth and metastasis. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 5128. doi:10.1158/1538-7445.AM2013-5128

Collaboration


Dive into the Baojun Chang's collaboration.

Top Co-Authors

Avatar

Hong Chen

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Xiaofeng Cai

Oklahoma Medical Research Foundation

View shared research outputs
Top Co-Authors

Avatar

Satish Pasula

Oklahoma Medical Research Foundation

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John McManus

Oklahoma Medical Research Foundation

View shared research outputs
Top Co-Authors

Avatar

Scott Hahn

Oklahoma Medical Research Foundation

View shared research outputs
Top Co-Authors

Avatar

Kandice L. Tessneer

Oklahoma Medical Research Foundation

View shared research outputs
Top Co-Authors

Avatar

Xiaolei Liu

Oklahoma Medical Research Foundation

View shared research outputs
Top Co-Authors

Avatar

Hao Wu

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge