Network


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

Hotspot


Dive into the research topics where Mee Young Chang is active.

Publication


Featured researches published by Mee Young Chang.


Cancer Discovery | 2012

IDO is a nodal pathogenic driver of lung cancer and metastasis development

Courtney Smith; Mee Young Chang; Katherine H. Parker; James B. DuHadaway; Hollie Flick; Janette Boulden; Erika Sutanto-Ward; Alejandro Peralta Soler; Lisa Laury-Kleintop; Laura Mandik-Nayak; Richard Metz; Suzanne Ostrand-Rosenberg; George C. Prendergast; Alexander J. Muller

UNLABELLED Indoleamine 2,3-dioxygenase (IDO) enzyme inhibitors have entered clinical trials for cancer treatment based on preclinical studies, indicating that they can defeat immune escape and broadly enhance other therapeutic modalities. However, clear genetic evidence of the impact of IDO on tumorigenesis in physiologic models of primary or metastatic disease is lacking. Investigating the impact of Ido1 gene disruption in mouse models of oncogenic KRAS-induced lung carcinoma and breast carcinoma-derived pulmonary metastasis, we have found that IDO deficiency resulted in reduced lung tumor burden and improved survival in both models. Micro-computed tomographic (CT) imaging further revealed that the density of the underlying pulmonary blood vessels was significantly reduced in Ido1-nullizygous mice. During lung tumor and metastasis outgrowth, interleukin (IL)-6 induction was greatly attenuated in conjunction with the loss of IDO. Biologically, this resulted in a consequential impairment of protumorigenic myeloid-derived suppressor cells (MDSC), as restoration of IL-6 recovered both MDSC suppressor function and metastasis susceptibility in Ido1-nullizygous mice. Together, our findings define IDO as a prototypical integrative modifier that bridges inflammation, vascularization, and immune escape to license primary and metastatic tumor outgrowth. SIGNIFICANCE This study provides preclinical, genetic proof-of-concept that the immunoregulatory enzyme IDO contributes to autochthonous carcinoma progression and to the creation of a metastatic niche. IDO deficiency in vivo negatively impacted both vascularization and IL-6–dependent, MDSC-driven immune escape, establishing IDO as an overarching factor directing the establishment of a protumorigenic environment.


Cancer Research | 2007

Bin1 Ablation Increases Susceptibility to Cancer during Aging, Particularly Lung Cancer

Mee Young Chang; Janette Boulden; Jessica B. Katz; Liwei Wang; Thomas J. Meyer; Alejandro Peralta Soler; Alexander J. Muller; George C. Prendergast

Age is the major risk factor for cancer, but few genetic pathways that modify cancer incidence during aging have been described. Bin1 is a prototypic member of the BAR adapter gene family that functions in vesicle dynamics and nuclear processes. Bin1 limits oncogenesis and is often attenuated in human cancers, but its role in cancer suppression has yet to be evaluated fully in vivo. In the mouse, homozygous deletion of Bin1 causes developmental lethality, so to assess this role, we examined cancer incidence in mosaic null mice generated by a modified Cre-lox technology. During study of these animals, one notable phenotype was an extended period of female fecundity during aging, with mosaic null animals retaining reproductive capability until the age of 17.3 +/- 1.1 months. Through 1 year of age, cancer incidence was unaffected by Bin1 ablation; however, by 18 to 20 months of age, approximately 50% of mosaic mice presented with lung adenocarcinoma and approximately 10% with hepatocarcinoma. Aging mosaic mice also displayed a higher incidence of inflammation and/or premalignant lesions, especially in the heart and prostate. In mice where colon tumors were initiated by a ras-activating carcinogen, Bin1 ablation facilitated progression to more aggressive invasive status. In cases of human lung and colon cancers, immunohistochemical analyses evidenced frequent attenuation of Bin1 expression, paralleling observations in other solid tumors. Taken together, our findings highlight an important role for Bin1 as a negative modifier of inflammation and cancer susceptibility during aging.


Cancer Immunology, Immunotherapy | 2010

Non-hematopoietic expression of IDO is integrally required for inflammatory tumor promotion

Alexander J. Muller; James B. DuHadaway; Mee Young Chang; Arivudinambi Ramalingam; Erika Sutanto-Ward; Janette Boulden; Alejandro Peralta Soler; Laura Mandik-Nayak; Susan K. Gilmour; George C. Prendergast

Indoleamine 2,3-dioxygenase (IDO) is generally considered to be immunosuppressive but recent findings suggest this characterization oversimplifies its role in disease pathogenesis. Recently, we showed that IDO is essential for tumor outgrowth in the classical two-stage model of inflammatory skin carcinogenesis. Here, we report that IDO loss did not exacerbate classical inflammatory responses. Rather, IDO induction could be elicited by environmental signals and tumor promoters as an integral component of the inflammatory tissue microenvironment even in the absence of cancer. IDO loss had limited impact on tumor outgrowth in carcinogenesis models that lacked an explicit inflammatory tumor promoter. In the context of inflammatory carcinogenesis where IDO was critical to tumor development, the most important source of IDO was radiation-resistant non-hematopoietic cells, consistent with evidence that loss of the IDO regulatory tumor suppressor gene Bin1 in transformed skin cells facilitates IDO-mediated immune escape by a cell autonomous mechanism. Taken together, our results identify IDO as an integral component of ‘cancer-associated’ inflammation that tilts the immune system toward tumor support. More generally, they promote the concept that mediators of immune escape and cancer-associated inflammation may be genetically synonymous.


Cancer Research | 2007

Bin1 ablation in mammary gland delays tissue remodeling and drives cancer progression

Mee Young Chang; Janette Boulden; Erika Sutanto-Ward; James B. DuHadaway; Alejandro Peralta Soler; Alexander J. Muller; George C. Prendergast

Genes that modify oncogenesis may influence dormancy versus progression in cancer, thereby affecting clinical outcomes. The Bin1 gene encodes a nucleocytosolic adapter protein that interacts with and suppresses the cell transforming activity of Myc. Bin1 is often attenuated in breast cancer but its ability to negatively modify oncogenesis or progression in this context has not been gauged directly. In this study, we investigated the effects of mammary gland-specific deletion of Bin1 on initiation and progression of breast cancer in mice. Bin1 loss delayed the outgrowth and involution of the glandular ductal network during pregnancy but had no effect on tumor susceptibility. In contrast, in mice where tumors were initiated by the ras-activating carcinogen 7,12-dimethylbenz(a)anthracene, Bin1 loss strongly accentuated the formation of poorly differentiated tumors characterized by increased proliferation, survival, and motility. This effect was specific as Bin1 loss did not accentuate progression of tumors initiated by an overexpressed mouse mammary tumor virus-c-myc transgene, which on its own produced poorly differentiated and aggressive tumors. These findings suggest that Bin1 loss cooperates with ras activation to drive progression, establishing a role for Bin1 as a negative modifier of oncogenicity and progression in breast cancer.


Cancer Biology & Therapy | 2011

Cardiac and gastrointestinal liabilities caused by deficiency in the immune modulatory enzyme indoleamine 2,3-dioxygenase

Mee Young Chang; Courtney Smith; James B. DuHadaway; Pyle; Janette Boulden; Alejandro Peralta Soler; Alexander J. Muller; Lisa Laury-Kleintop; George C. Prendergast

Indoleamine 2,3-dioxygenase (IDO) modifies adaptive immunity, in part by determining the character of inflammatory responses in the tissue microenvironment. Small molecule inhibitors of IDO are being developed to treat cancer, chronic infections and other diseases, so the systemic effects of IDO disruption on inflammatory phenomena may influence the design and conduct of early phase clinical investigations of this new class of therapeutic agents. Here, we report cardiac and gastrointestinal phenotypes observed in IDO deficient mice that warrant consideration in planned assessments of the safety risks involved in clinical development of IDO inhibitors. Calcification of the cardiac endometrium proximal to the right ventricle was a sexually dimorphic strain-specific phenotype with ~30% penetrance in BALB/c mice lacking IDO. Administration of complete Freund’s adjuvant containing Toll-like receptor ligands known to induce IDO caused acute pancreatitis in IDO deficient mice, with implications for the design of planned combination studies of IDO inhibitors with cancer vaccines. In an established model of hyperlipidemia, IDO deficiency caused a dramatic elevation in levels of serum triglycerides. In the large intestine, IDO loss only slightly increased sensitivity to induction of acute colitis, but it markedly elevated tumor incidence, multiplicity and staging during inflammatory colon carcinogenesis. Together, our findings suggest potential cardiac and gastrointestinal risks of IDO inhibitors that should be monitored in patients as this new class of drugs enter early clinical development.


Digestive Diseases and Sciences | 2012

Bin1 Attenuation Suppresses Experimental Colitis by Enforcing Intestinal Barrier Function

Mee Young Chang; Janette Boulden; M. Carmen Valenzano; Alejandro Peralta Soler; Alexander J. Muller; James M. Mullin; George C. Prendergast

BackgroundInflammatory bowel disease (IBD) is associated with defects in intestinal barriers that rely upon cellular tight junctions. Thus, identifying genes that could be targeted to enforce tight junctions and improve barrier function may lead to new treatment strategies for IBD.AimsThis preclinical study aimed to evaluate an hypothesized role for the tumor suppressor gene Bin1 as a modifier of the severity of experimental colitis.MethodsWe ablated the Bin1 gene in a mosaic mouse model to evaluate its effects on experimental colitis and intestinal barrier function. Gross pathology, histology and inflammatory cytokine expression patterns were characterized and ex vivo physiology determinations were conducted to evaluate barrier function in intact colon tissue.ResultsBin1 attenuation limited experimental colitis in a sexually dimorphic manner with stronger protection in female subjects. Colitis suppression was associated with an increase in basal transepithelial electrical resistance (TER) and a decrease in paracellular transepithelial flux, compared to control wild-type animals. In contrast, Bin1 attenuation did not affect short circuit current, nor did it alter the epithelial barrier response to non-inflammatory permeability enhancers in the absence of inflammatory stimuli.ConclusionsBin1 is a genetic modifier of experimental colitis that controls the paracellular pathway of transcellular ion transport regulated by cellular tight junctions. Our findings offer a preclinical validation of Bin1 as a novel therapeutic target for IBD treatment.


Journal of Cellular Biochemistry | 2015

Cardiac-Specific Disruption of Bin1 in Mice Enables a Model of Stress- and Age-Associated Dilated Cardiomyopathy

Lisa Laury-Kleintop; Jennifer Mulgrew; Ido Heletz; Radu Alexandru Nedelcoviciu; Mee Young Chang; David M. Harris; Walter J. Koch; Michael D. Schneider; Alexander J. Muller; George C. Prendergast

Non‐compensated dilated cardiomyopathy (DCM) leading to death from heart failure is rising rapidly in developed countries due to aging demographics, and there is a need for informative preclinical models to guide the development of effective therapeutic strategies to prevent or delay disease onset. In this study, we describe a novel model of heart failure based on cardiac‐specific deletion of the prototypical mammalian BAR adapter‐encoding gene Bin1, a modifier of age‐associated disease. Bin1 deletion during embryonic development causes hypertrophic cardiomyopathy and neonatal lethality, but there is little information on how Bin1 affects cardiac function in adult animals. Here we report that cardiomyocyte‐specific loss of Bin1 causes age‐associated dilated cardiomyopathy (DCM) beginning by 8–10 months of age. Echocardiographic analysis showed that Bin1 loss caused a 45% reduction in ejection fraction during aging. Younger animals rapidly developed DCM if cardiac pressure overload was created by transverse aortic constriction. Heterozygotes exhibited an intermediate phenotype indicating Bin1 is haplo‐insufficient to sustain normal heart function. Bin1 loss increased left ventricle (LV) volume and diameter during aging, but it did not alter LV volume or diameter in hearts from heterozygous mice nor did it affect LV mass. Bin1 loss increased interstitial fibrosis and mislocalization of the voltage‐dependent calcium channel Cav1.2, and the lipid raft scaffold protein caveolin‐3, which normally complexes with Bin1 and Cav1.2 in cardiomyocyte membranes. Our findings show how cardiac deficiency in Bin1 function causes age‐ and stress‐associated heart failure, and they establish a new preclinical model of this terminal cardiac disease. J. Cell. Biochem. 116: 2541–2551, 2015.


Atlas of genetics and cytogenetics in oncology and haematology | 2011

BIN1 (bridging integrator 1)

Sunil Thomas; Mee Young Chang; George C. Prendergast

Review on BIN1 (bridging integrator 1), with data on DNA, on the protein encoded, and where the gene is implicated.


Atlas of genetics and cytogenetics in oncology and haematology | 2011

IDO2 (indoleamine 2,3 -dioxygenase 2)

Mee Young Chang; Richard Metz; Alexander J. Muller; George C. Prendergast

Review on IDO2 (indoleamine 2,3-dioxygenase 2), with data on DNA, on the protein encoded, and where the gene is implicated.


Archive | 2011

Indoleamine 2,3-Dioxygenase Amino Acid Metabolism and Tumour-Associated Macrophages: Regulation in Cancer-Associated Inflammation and Immune Escape

George C. Prendergast; Richard Metz; Mee Young Chang; Courtney Smith; Alexander J. Muller; Suzanne Ostrand-Rosenberg

Indoleamine 2,3-dioxygenases (IDO) enzymes that catabolize tryptophan are expressed in tumor, stromal, and immune cells in the tumor microenvironment, including in monocytic cells such as macrophages (TAMs). Recent mouse genetic studies define a crucial role for IDO in supporting inflammatory carcinogenesis. Key gaps remain in elucidating the immunoregulatory pathways that modify progression versus dormancy in early stage tumors, an important clinical concern. Within the tumor microenvironment, chronic inflammation and immune escape are critical contributors to progression. However, there is still limited knowledge about the molecular mechanisms which underlie cancer-associated inflammation and immune escape in cancer. Both the IDO1 and IDO2 enzymes that catabolize tryptophan have been implicated in immune escape (pathogenic immune tolerance). Contributions from different parts of the tumor microenvironment have yet to be fully defined, but an important implication of these recent studies is that pathways of immune escape and pathogenic inflammation in cancer may be genetically synonymous. IDO enzymes are generally regarded as being immunosuppressive, however, emerging evidence suggests that this view is too narrow and that IDO enzymes may function more broadly to modify or ‘flavor’ the nature of an inflammatory microenvironment, rendering it supportive to tumor progression. In this review, we summarize work on IDO in immune escape and pathogenic inflammation in cancer, discussing potential roles for IDO pathways and other amino acid catabolism pathways in TAMs and other immune cells that may contribute to shaping tissue microenvironments that nurture the development and progression of malignancy.

Collaboration


Dive into the Mee Young Chang's collaboration.

Top Co-Authors

Avatar

George C. Prendergast

Lankenau Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Alexander J. Muller

Lankenau Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Alejandro Peralta Soler

Lankenau Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

James B. DuHadaway

Lankenau Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Janette Boulden

Lankenau Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Courtney Smith

Lankenau Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Erika Sutanto-Ward

Lankenau Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar

Lisa Laury-Kleintop

Lankenau Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge