Network


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

Hotspot


Dive into the research topics where Anthony C. Wong is active.

Publication


Featured researches published by Anthony C. Wong.


Nature | 2011

Ebola virus entry requires the cholesterol transporter Niemann–Pick C1

Jan E. Carette; Matthijs Raaben; Anthony C. Wong; Andrew S. Herbert; Gregor Obernosterer; Nirupama Mulherkar; Ana I. Kuehne; Philip J. Kranzusch; April M. Griffin; Gordon Ruthel; Paola Dal Cin; John M. Dye; Sean P. J. Whelan; Kartik Chandran; Thijn R. Brummelkamp

Infections by the Ebola and Marburg filoviruses cause a rapidly fatal haemorrhagic fever in humans for which no approved antivirals are available. Filovirus entry is mediated by the viral spike glycoprotein (GP), which attaches viral particles to the cell surface, delivers them to endosomes and catalyses fusion between viral and endosomal membranes. Additional host factors in the endosomal compartment are probably required for viral membrane fusion; however, despite considerable efforts, these critical host factors have defied molecular identification. Here we describe a genome-wide haploid genetic screen in human cells to identify host factors required for Ebola virus entry. Our screen uncovered 67 mutations disrupting all six members of the homotypic fusion and vacuole protein-sorting (HOPS) multisubunit tethering complex, which is involved in the fusion of endosomes to lysosomes, and 39 independent mutations that disrupt the endo/lysosomal cholesterol transporter protein Niemann–Pick C1 (NPC1). Cells defective for the HOPS complex or NPC1 function, including primary fibroblasts derived from human Niemann–Pick type C1 disease patients, are resistant to infection by Ebola virus and Marburg virus, but remain fully susceptible to a suite of unrelated viruses. We show that membrane fusion mediated by filovirus glycoproteins and viral escape from the vesicular compartment require the NPC1 protein, independent of its known function in cholesterol transport. Our findings uncover unique features of the entry pathway used by filoviruses and indicate potential antiviral strategies to combat these deadly agents.


Journal of Experimental Medicine | 2003

Regulation and Function of the Interleukin 13 Receptor α 2 During a T Helper Cell Type 2–dominant Immune Response

Monica G. Chiaramonte; Margaret M. Mentink-Kane; Bruce A. Jacobson; Allen W. Cheever; Matthew J. Whitters; Mary E.P. Goad; Anthony C. Wong; Mary Collins; Debra D. Donaldson; Michael J. Grusby; Thomas A. Wynn

Highly polarized type 2 cytokine responses can be harmful and even lethal to the host if they are too vigorous or persist too long. Therefore, it is important to elucidate the mechanisms that down-regulate these reactions. Interleukin (IL)-13 has emerged as a central mediator of T helper cell (Th)2-dominant immune responses, exhibiting a diverse array of functional activities including regulation of airway hyperreactivity, resistance to nematode parasites, and tissue remodeling and fibrosis. Here, we show that IL-13 receptor (R)α2 is a critical down-regulatory factor of IL-13–mediated tissue fibrosis induced by the parasitic helminth Schistosoma mansoni. IL-13Rα2 expression was induced after the onset of the fibrotic response, IL-10, IL-13, and Stat6 dependent, and inhibited by the Th1-inducing adjuvant IL-12. Strikingly, schistosome-infected C57BL/6 and BALB/c IL-13Rα2–deficient mice showed a marked exacerbation in hepatic fibrosis, despite displaying no change in granuloma size, tissue eosinophilia, or mastocytosis. Fibrosis increased despite the fact that IL-13 levels decreased significantly in the liver and serum. Importantly, pathology was prevented when IL-13Rα2–deficient mice were treated with a soluble IL-13Rα2-Fc construct, formally demonstrating that their exacerbated fibrotic response was due to heightened IL-13 activity. Together, these studies illustrate the central role played by the IL-13Rα2 in the down-regulation of a chronic and pathogenic Th2-mediated immune response.


Cancer Research | 2010

HGF/c-Met Acts as an Alternative Angiogenic Pathway in Sunitinib- Resistant Tumors

Farbod Shojaei; Joseph H. Lee; Brett H. Simmons; Anthony C. Wong; Carlos O. Esparza; Pamela A. Plumlee; Junli Feng; Albert E. Stewart; Dana Hu-Lowe; James G. Christensen

Molecular and cellular mechanisms underlying resistance/low responsiveness to antiangiogenic compounds are under extensive investigations. Both populations of tumor and stroma (nontumor compartment) seem to contribute in inherent/acquired resistance to antiangiogenic therapy. Here, investigating in vivo efficacy of sunitinib in experimental models resulted in the identification of tumors that were resistant/sensitive to the therapy. Analysis of tumor protein lysates indicated a greater concentration of hepatocyte growth factor (HGF) in resistant tumors than in sensitive ones. In addition, using flow cytometry, c-Met expression was found to be significantly higher in endothelial cells than in tumor cells, suggesting that HGF might target the vascular endothelial cells in resistant tumors. Combination of sunitinib and a selective c-Met inhibitor significantly inhibited tumor growth compared with sunitinib or c-Met inhibitor alone in resistant tumors. Histology and in vitro analyses suggested that combination treatment mainly targeted the vasculature in the resistant tumors. Conversely, systemic injection of HGF in the sensitive tumor models conferred resistance to sunitinib through maintenance of tumor angiogenesis. In conclusion, our study indicates a role for HGF/c-Met pathway in development of resistance to antiangiogenic therapy and suggests a potential strategy to circumvent resistance to vascular endothelial growth factor receptor tyrosine kinase inhibitor in the clinic.


Nature Structural & Molecular Biology | 2011

A shared structural solution for neutralizing ebolaviruses

João M. Dias; Ana I. Kuehne; Dafna M. Abelson; Shridhar Bale; Anthony C. Wong; Peter Halfmann; Majidat Muhammad; Marnie L. Fusco; Samantha E. Zak; Eugene Kang; Yoshihiro Kawaoka; Kartik Chandran; John M. Dye; Erica Ollmann Saphire

Sudan virus (genus Ebolavirus) is lethal, yet no monoclonal antibody is known to neutralize it. We here describe antibody 16F6 that neutralizes Sudan virus and present its structure bound to the trimeric viral glycoprotein. Unexpectedly, the 16F6 epitope overlaps that of KZ52, the only other antibody against the GP1,2 core to be visualized to date. Furthermore, both antibodies against this crucial epitope bridging GP1–GP2 neutralize at a post-internalization step—probably fusion.


Journal of Virology | 2010

A Forward Genetic Strategy Reveals Destabilizing Mutations in the Ebolavirus Glycoprotein That Alter Its Protease Dependence during Cell Entry

Anthony C. Wong; Rohini G. Sandesara; Nirupama Mulherkar; Sean P. J. Whelan; Kartik Chandran

ABSTRACT Ebolavirus (EBOV) entry into cells requires proteolytic disassembly of the viral glycoprotein, GP. This proteolytic processing, unusually extensive for an enveloped virus entry protein, is mediated by cysteine cathepsins, a family of endosomal/lysosomal proteases. Previous work has shown that cleavage of GP by cathepsin B (CatB) is specifically required to generate a critical entry intermediate. The functions of this intermediate are not well understood. We used a forward genetic strategy to investigate this CatB-dependent step. Specifically, we generated a replication-competent recombinant vesicular stomatitis virus bearing EBOV GP as its sole entry glycoprotein and used it to select viral mutants resistant to a CatB inhibitor. We obtained mutations at six amino acid positions in GP that independently confer complete resistance. All of the mutations reside at or near the GP1-GP2 intersubunit interface in the membrane-proximal base of the prefusion GP trimer. This region forms a part of the “clamp” that holds the fusion subunit GP2 in its metastable prefusion conformation. Biochemical studies suggest that most of the mutations confer CatB independence not by altering specific cleavage sites in GP but rather by inducing conformational rearrangements in the prefusion GP trimer that dramatically enhance its susceptibility to proteolysis. The remaining mutants did not show the preceding behavior, indicating the existence of multiple mechanisms for acquiring CatB independence during entry. Altogether, our findings suggest that CatB cleavage is required to facilitate the triggering of viral membrane fusion by destabilizing the prefusion conformation of EBOV GP.


Clinical Cancer Research | 2012

[18F]FLT–PET Imaging Does Not Always “Light Up” Proliferating Tumor Cells

Cathy Zhang; Zhengming Yan; Wenlin Li; Kyle Kuszpit; Cory L. Painter; Qin Zhang; Patrick B. Lappin; Timothy Nichols; Maruja E. Lira; Timothy Affolter; Neeta Fahey; Carleen Cullinane; Mary E. Spilker; Kenneth R. Zasadny; Peter J. O'Brien; Dana Buckman; Anthony C. Wong; James G. Christensen

Purpose: [18F]FLT (3′-Fluoro-3′ deoxythymidine)–PET imaging was proposed as a tool for measuring in vivo tumor cell proliferation. The aim of this article was to validate the use of [18F]FLT–PET imaging for measuring xenograft proliferation and subsequent monitoring of targeted therapy. Experimental Design: In exponentially growing xenografts, factors that could impact the outcome of [18F]FLT–PET imaging, such as nucleoside transporters, thymidine kinase 1, the relative contribution of DNA salvage pathway, and the ratio of FLT to thymidine, were evaluated. The [18F]FLT tracer avidity was compared with other proliferation markers. Results: In a panel of proliferating xenografts, [18F]FLT or [3H]thymidine tracer avidity failed to reflect the tumor growth rate across different tumor types, despite the high expressions of Ki67 and TK1. When FLT was injected at the same dose level as used in the preclinical [18F]FLT–PET imaging, the plasma exposure ratio of FLT to thymidine was approximately 1:200. Thymidine levels in different tumor types seemed to be variable and exhibited an inverse relationship with the FLT tracer avidity. In contrast, high-dose administration of bromdeoxyuridine (BrdUrd; 50 mg/kg) yielded a plasma exposure of more than 4-fold higher than thymidine and leads to a strong correlation between the BrdUrd uptake and the tumor proliferation rate. In FLT tracer-avid models, [18F]FLT–PET imaging as a surrogate biomarker predicted the therapeutic response of CDK4/6 inhibitor PD-0332991. Conclusions: Tumor thymidine level is one of the factors that impact the correlation between [18F]FLT uptake and tumor cell proliferation. With careful validation, [18F]FLT–PET imaging can be used to monitor antiproliferative therapies in tracer-avid malignancies. Clin Cancer Res; 18(5); 1303–12. ©2011 AACR.


Clinical Cancer Research | 2010

PF-03732010: a fully human monoclonal antibody against P-cadherin with antitumor and antimetastatic activity.

Cathy Zhang; Zhengming Yan; Qin Zhang; Kyle Kuszpit; Kenneth R. Zasadny; Ming Qiu; Cory L. Painter; Anthony C. Wong; Eugenia Kraynov; Maria E. Arango; Pramod P. Mehta; Ian Popoff; Gerald Fries Casperson; Gerrit Los; Steve Bender; Kenna Anderes; James G. Christensen; Todd VanArsdale

Purpose: P-cadherin is a membrane glycoprotein that functionally mediates tumor cell adhesion, proliferation, and invasiveness. We characterized the biological properties of PF-03732010, a human monoclonal antibody against P-cadherin, in cell-based assays and tumor models. Experimental Design: The affinity, selectivity, and cellular inhibitory activity of PF-03732010 were tested in vitro. Multiple orthotopic and metastatic tumor models were used for assessing the antitumor and antimetastatic activities of PF-03732010. Treatment-associated pharmacodynamic changes were also investigated. Results: PF-03732010 selectively inhibits P-cadherin–mediated cell adhesion and aggregation in vitro. In the P-cadherin–overexpressing tumor models, including MDA-MB-231-CDH3, 4T1-CDH3, MDA-MB-435HAL-CDH3, HCT116, H1650, PC3M-CDH3, and DU145, PF-03732010 inhibited the growth of primary tumors and metastatic progression, as determined by bioluminescence imaging. Computed tomography imaging, H&E stain, and quantitative PCR analysis confirmed the antimetastatic activity of PF-03732010. In contrast, PF-03732010 did not show antitumor and antimetastatic efficacy in the counterpart tumor models exhibiting low P-cadherin expression. Mechanistic studies via immunofluorescence, immunohistochemical analyses, and 3′-[18F]fluoro-3′-deoxythymidine–positron emission tomography imaging revealed that PF-03732010 suppressed P-cadherin levels, caused degradation of membrane β-catenin, and concurrently suppressed cytoplasmic vimentin, resulting in diminished metastatic capacity. Changes in the levels of Ki67, caspase-3, and 3′-[18F]fluoro-3′-deoxythymidine tracer uptake also indicated antiproliferative activity and increased apoptosis in the tested xenografts. Conclusions: These findings suggest that interrupting the P-cadherin signaling pathway may be a novel therapeutic approach for cancer therapy. PF-03732010 is presently undergoing evaluation in Phase 1 clinical trials. Clin Cancer Res; 16(21); 5177–88. ©2010 AACR.


Viruses | 2012

Structural Basis for Differential Neutralization of Ebolaviruses

Shridhar Bale; João M. Dias; Marnie L. Fusco; Takao Hashiguchi; Anthony C. Wong; Tong Liu; Ana I. Keuhne; Sheng Li; Virgil L. Woods; Kartik Chandran; John M. Dye; Erica Ollmann Saphire

There are five antigenically distinct ebolaviruses that cause hemorrhagic fever in humans or non-human primates (Ebola virus, Sudan virus, Reston virus, Taï Forest virus, and Bundibugyo virus). The small handful of antibodies known to neutralize the ebolaviruses bind to the surface glycoprotein termed GP1,2. Curiously, some antibodies against them are known to neutralize in vitro but not protect in vivo, whereas other antibodies are known to protect animal models in vivo, but not neutralize in vitro. A detailed understanding of what constitutes a neutralizing and/or protective antibody response is critical for development of novel therapeutic strategies. Here, we show that paradoxically, a lower affinity antibody with restricted access to its epitope confers better neutralization than a higher affinity antibody against a similar epitope, suggesting that either subtle differences in epitope, or different characteristics of the GP1,2 molecules themselves, confer differential neutralization susceptibility. Here, we also report the crystal structure of trimeric, prefusion GP1,2 from the original 1976 Boniface variant of Sudan virus complexed with 16F6, the first antibody known to neutralize Sudan virus, and compare the structure to that of Sudan virus, variant Gulu. We discuss new structural details of the GP1-GP2 clamp, thermal motion of various regions in GP1,2 across the two viruses visualized, details of differential interaction of the crystallized neutralizing antibodies, and their relevance for virus neutralization.


Orthopedics | 2012

Pain management after total knee arthroplasty using a multimodal approach.

Morteza Meftah; Anthony C. Wong; Danyal H. Nawabi; Richard J Yun; Amar S. Ranawat; Chitranjan S. Ranawat

Improvements in pain management techniques over the past decade have had a significant impact on the outcomes of total knee arthroplasty. Of these techniques, multimodal approaches have shown potential. The purpose of this study was to compare the results of periarticular injection (PAI) to a combination of patient-controlled epidural analgesia and femoral nerve block (PCEA/FNB). Ninety patients undergoing primary unilateral total knee arthroplasty between June 2010 and March 2011 were randomized into 2 groups. The first group received the PCEA/FNB protocol, whereas the second group received the PAI. Mean patient age was 66.1 ± 8.7 years. All patients were operated on using a similar standard medial parapatellar approach, and all received preemptive analgesia and postoperative pain protocols. All patients were interviewed twice daily for the first 3 days postoperatively, once on day 7, and once in month 6. The 2 groups had similar readiness for discharge (PCEA/FNB group, 3.3 ± 1.2 days; PAI group, 3.2 ± 1.9 days). The results indicated no statistical difference between the 2 groups in 3 of 4 categories (rest in the morning, rest in the evening, and ambulation in the morning). Pain on ambulation was the only category that was statistically lower in the PCEA/FNB group than in the PAI group.Although the study demonstrates similar results between the 2 groups, PAI can play a major role in postoperative pain control in institutions that may not have appropriately trained individuals, equipment, and resources for PCEA/FNB. It also reduces many of the side effects and complications associated with regional anesthesia.


Clinical Cancer Research | 2009

PF-00477736 Mediates Checkpoint Kinase 1 Signaling Pathway and Potentiates Docetaxel-Induced Efficacy in Xenografts

Cathy Zhang; Zhengming Yan; Cory L. Painter; Qin Zhang; Enhong Chen; Maria E. Arango; Kyle Kuszpit; Kenneth R. Zasadny; Max Hallin; Jill Hallin; Anthony C. Wong; Dana Buckman; Guizhen Sun; Ming Qiu; Kenna Anderes; James G. Christensen

Purpose: Checkpoint kinase 1 (Chk1) plays a critical role in the activation of mitotic spindle checkpoint and DNA damage checkpoint. We examined the preclinical use of the Chk1 inhibitor PF-00477736 as a docetaxel-sensitizing agent. Specifically, we investigated the correlation between PF-00477736–mediated modulation of biomarkers and the sensitization of docetaxel efficacy. Experimental Design:In vitro and in vivo studies using COLO205 and other cell lines were done to assess PF-00477736–induced enhancement of docetaxel efficacy and effects on associated biomarkers. Results: PF-00477736 significantly enhanced the docetaxel-induced efficacy in tumor cells and xenografts. Docetaxel induced dose- and time-dependent increase in the levels of phosphorylated Chk1 (Ser345), phosphorylated histone H3 (Ser10), and γH2AX foci and promoted the cytoplasmic localization of phosphorylated Cdc25C (Ser216). PF-00477736 cotreatment suppressed docetaxel-induced changes in phosphorylated histone H3 and cytoplasmic phosphorylated Cdc25C (Ser216) levels and concurrently sensitized the docetaxel-induced apoptosis. Docetaxel alone or in combination with PF-00477736 induced significant antiproliferative activity in xenografts, shown via [18F]FLT-PET imaging. However, changes in [18F]FLT uptake did not reflect the potentiation of docetaxel efficacy. In contrast, bioluminescence imaging showed that PF-00477736 sensitized docetaxel-induced suppression of tumor survival. Conclusions: Docetaxel triggers mitotic spindle checkpoint activation at low concentrations and activates both the DNA damage checkpoint and the spindle checkpoint at high concentrations. In combination with docetaxel, PF-00477736 abrogates the mitotic checkpoint, as well as the DNA damage checkpoint, and results in sensitization to docetaxel. Chk1 inhibitor PF-00477736 offers a therapeutic potential for the enhancement of taxane therapy.

Collaboration


Dive into the Anthony C. Wong's collaboration.

Top Co-Authors

Avatar

Kartik Chandran

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John M. Dye

United States Army Medical Research Institute of Infectious Diseases

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge