Network


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

Hotspot


Dive into the research topics where Grace Chan is active.

Publication


Featured researches published by Grace Chan.


Circulation | 2005

Myeloid Differentiation Factor-88 Plays a Crucial Role in the Pathogenesis of Coxsackievirus B3–Induced Myocarditis and Influences Type I Interferon Production

Koichi Fuse; Grace Chan; Youan Liu; Patrick Gudgeon; Mansoor Husain; Manyin Chen; Wen-Chen Yeh; Shizuo Akira; Peter Liu

Background— Myeloid differentiation factor (MyD)-88 is a key adaptor protein that plays a major role in the innate immune pathway. How MyD88 may regulate host response in inflammatory heart disease is unknown. Methods and Results— We found that the cardiac protein level of MyD88 was significantly increased in the hearts of wild-type mice after exposure to Coxsackievirus B3 (CVB3). MyD88−/− mice showed a dramatic higher survival rate (86%) in contrast to the low survival (35%) in the MyD88+/+ mice after CVB3 infection (P<0.0001). Pathological examination showed a significant decrease of cardiac and pancreatic inflammation in the MyD88−/− mice. Viral concentrations in the hearts were significantly decreased in the MyD88−/− mice. Cardiac mRNA levels for interleukin (IL)-1&bgr;, tumor necrosis factor (TNF)-&agr;, interferon (IFN)-&ggr;, and IL-18 were significantly decreased in the MyD88−/− mice. Similarly, serum levels of T-helper 1 cytokines were significantly decreased in the MyD88−/− mice. In contrast, cardiac protein levels of the activated interferon regulatory factor (IRF)-3 and IFN-&bgr; were significantly increased in the MyD88−/− mice but not other usual upstream signals to IRF-3. The cardiac expression of coxsackie-adenoviral receptor and p56lck were also significantly decreased. Conclusions— MyD88 appears to be a key contributor to cardiac inflammation, mediating cytokine production and T-helper-1/2 cytokine balance, increasing coxsackie-adenoviral receptor and p56lck expression and viral titers after CVB3 exposure. Absence of MyD88 confers host protection possibly through novel direct activation of IRF-3 and IFN-&bgr;.


Science Translational Medicine | 2012

Targeting Cancer with a Lupus Autoantibody

James E. Hansen; Grace Chan; Yanfeng Liu; Denise C. Hegan; Shibani Dalal; Eloise Dray; Youngho Kwon; Yuanyuan Xu; Xiaohua Xu; Elizabeth Peterson-Roth; Erik J. Geiger; Yilun Liu; Joseph Gera; Joann B. Sweasy; Patrick Sung; Sara Rockwell; Robert N. Nishimura; Richard H. Weisbart; Peter M. Glazer

A cell-penetrating lupus anti-DNA antibody inhibits DNA repair, sensitizes cancer cells to DNA-damaging therapy in vitro and in vivo, and is synthetically lethal to BRCA2-deficient human cancer cells. Taming the Big Bad Wolf Just like the wolves for which lupus is named, the antibodies involved in its pathogenesis can attack almost any part of a patient, causing widespread damage. Now, Hansen et al. show that these biological wolves can sometimes be tamed and their ferociousness put to use in treating another deadly disease. Lupus is an autoimmune disease associated with antibodies that target host DNA, wreaking havoc on patients’ cells throughout the body. Recently, cancer researchers have tried to co-opt some of these antibodies, particularly those that can penetrate human cells, for use as vehicles for therapeutic agents. While using lupus antibodies to deliver proteins to protect normal cells from therapeutic ionizing radiation delivered to a tumor, researchers discovered that one antibody, 3E10, could itself sensitize cancer cells to radiation treatment. The authors then characterized this observed effect in malignant cells and determined its mechanism. They found that 3E10 bound single-stranded DNA and interfered with its repair, making the cells more susceptible to DNA-damaging agents such as doxorubicin and radiation. In addition, 3E10 alone was toxic to cancer cells with deficient DNA repair pathways, such as those that harbor BRCA2 mutations. Further research is necessary to identify other pathways that make tumor cells susceptible to 3E10 and to analyze the pharmacokinetics and other characteristics of this treatment. However, 3E10 was already shown to be safe in a previous phase 1 trial in lupus patients and should now be able to transition into clinical trials for cancer patients as well. Although researchers have not yet discovered a cure for lupus, the big bad wolf’s offspring may potentially tame another life-threatening illness. Systemic lupus erythematosus (SLE) is distinct among autoimmune diseases because of its association with circulating autoantibodies reactive against host DNA. The precise role that anti-DNA antibodies play in SLE pathophysiology remains to be elucidated, and potential applications of lupus autoantibodies in cancer therapy have not previously been explored. We report the unexpected finding that a cell-penetrating lupus autoantibody, 3E10, has potential as a targeted therapy for DNA repair–deficient malignancies. We find that 3E10 preferentially binds DNA single-strand tails, inhibits key steps in DNA single-strand and double-strand break repair, and sensitizes cultured tumor cells and human tumor xenografts to DNA-damaging therapy, including doxorubicin and radiation. Moreover, we demonstrate that 3E10 alone is synthetically lethal to BRCA2-deficient human cancer cells and selectively sensitizes such cells to low-dose doxorubicin. Our results establish an approach to cancer therapy that we expect will be particularly applicable to BRCA2-related malignancies such as breast, ovarian, and prostate cancers. In addition, our findings raise the possibility that lupus autoantibodies may be partly responsible for the intrinsic deficiencies in DNA repair and the unexpectedly low rates of breast, ovarian, and prostate cancers observed in SLE patients. In summary, this study provides the basis for the potential use of a lupus anti-DNA antibody in cancer therapy and identifies lupus autoantibodies as a potentially rich source of therapeutic agents.


Cancer Research | 2007

Antibody-Mediated p53 Protein Therapy Prevents Liver Metastasis In vivo

James E. Hansen; Laurice K. Fischer; Grace Chan; Sophia S. Chang; Scott Baldwin; Robert Aragon; Jacqueline J. Carter; Michael B. Lilly; Robert N. Nishimura; Richard H. Weisbart; Mark E. Reeves

To evaluate the clinical efficacy of monoclonal antibody (mAb) 3E10 Fv antibody-mediated p53 protein therapy, an Fv-p53 fusion protein produced in Pichia pastoris was tested on CT26.CL25 colon cancer cells in vitro and in vivo in a mouse model of colon cancer metastasis to the liver. In vitro experiments showed killing of CT26.CL25 cells by Fv-p53 but not Fv or p53 alone, and immunohistochemical staining confirmed that Fv was required for transport of p53 into cells. Prevention of liver metastasis in vivo was tested by splenic injection of 100 nmol/L Fv-p53 10 min and 1 week after injection of CT26.CL25 cancer cells into the portal vein of BALB/c mice. Mice were sacrificed 1 week after the second injection of Fv-p53 and assigned a quantitative metastasis score. Control mice had an average metastasis score of 3.3 +/- 1.3, whereas mice treated with Fv-p53 had an average metastasis score of 0.8 +/- 0.4 (P = 0.004). These results indicate that Fv-p53 treatment had a profound effect on liver metastasis and represent the first demonstration of effective full-length p53 protein therapy in vivo. mAb 3E10 Fv has significant clinical potential as a mediator of intracellular and intranuclear delivery of p53 for prevention and treatment of cancer metastasis.


Molecular Cancer Therapeutics | 2012

A Cell-Penetrating Bispecific Antibody for Therapeutic Regulation of Intracellular Targets

Richard H. Weisbart; Joseph Gera; Grace Chan; James E. Hansen; Erica Li; Cheri Cloninger; Arnold J. Levine; Robert N. Nishimura

The therapeutic use of antibodies is restricted by the limited access of antibodies to intracellular compartments. To overcome this limitation, we developed a cell-penetrating monoclonal antibody, mAb 3E10, as an intracellular delivery vehicle for the intracellular and intranuclear delivery of antibodies constructed as bispecific single-chain Fv fragments. Because MDM2 is an important target in cancer therapy, we selected monoclonal antibody (mAb) 3G5 for intracellular transport. mAb 3G5 binds MDM2 and blocks binding of MDM2 to p53. Here, we show that the resulting 3E10-3G5 bispecific antibody retains cell-penetrating and MDM2-binding activity, increases tumor p53 levels, and inhibits growth of MDM2-addicted tumors. The use of cell-penetrating bispecific antibodies in targeted molecular therapy will significantly broaden the spectrum of accessible intracellular targets and may have a profound impact in cancer therapy. Mol Cancer Ther; 11(10); 2169–73. ©2012 AACR.


Brain Research | 2006

Antibody-mediated Hsp70 protein therapy

James E. Hansen; William Sohn; Charles O. Kim; Sophia S. Chang; Natalie C. Huang; Donaldson G. Santos; Grace Chan; Richard H. Weisbart; Robert N. Nishimura

Intracellular Hsp70 provides cytoprotection against a variety of stressful stimuli, and an effective means of increasing intracellular Hsp70 levels could prove beneficial in the prevention and treatment of a variety of human diseases. A novel protein transduction domain consisting of the single chain Fv fragment of an anti-DNA antibody known to penetrate into living cells and tissues, mAb 3E10, has recently been used to deliver functional proteins to cells. The ability of the single chain Fv fragment to deliver Hsp70 into living cells was tested by generating an Fv-Hsp70 fusion protein. Fv-Hsp70 was produced as a secreted protein in both COS-7 cells and the methylotropic yeast strain Pichia pastoris and was shown capable of penetrating into COS-7 cells and primary rat cortical neurons. Pre-treatment with Fv-Hsp70 protected both COS-7 cells and primary rat cortical neurons against subsequent exposure to hydrogen peroxide. These results provide the first evidence that the Fv fragment of mAb 3E10 is capable of delivering proteins to neurons and indicate its potential in the development of Hsp70 protein therapy.


Molecular Immunology | 2003

Cell type specific targeted intracellular delivery into muscle of a monoclonal antibody that binds myosin IIb

Richard H. Weisbart; Fusheng Yang; Grace Chan; Rika Wakelin; Kevin Ferreri; Debra J Zack; Brooke C. Harrison; Leslie A. Leinwand; Greg M. Cole

Methods for cell type specific targeted intracellular delivery of proteins in vivo remain limited. A murine monoclonal anti-dsDNA antibody, mAb 3E10, was selectively transported into skeletal muscle cells in vivo. The antibody bound a 200 kDa protein only found in lysates of skeletal muscle by Western blotting. The 200 kDa protein was purified from muscle lysate by antibody affinity chromatography and identified as the skeletal muscle specific heavy chain of myosin IIb by electrospray mass spectrometry. Antibody binding specificity for myosin IIb was demonstrated in Western blots by binding myosin in skeletal muscle lysates from mice null for myosin IId but not in mice null for myosin IIb. Myosin IIb is implicated in the specific targeting of mAb 3E10 to skeletal muscle.


Journal of Drug Targeting | 2005

An intracellular delivery vehicle for protein transduction of micro-dystrophin

Richard H. Weisbart; James E. Hansen; Robert N. Nishimura; Grace Chan; Rika Wakelin; Sophia S. Chang; Larry Baresi; Jeffrey S. Chamberlain

The Fv fragment of an antibody that selectively targets and penetrates skeletal muscle in vivo was produced as a fusion protein with a micro-dystrophin for use as a delivery vehicle to transport micro-dystrophin into muscle cells. Fv-micro-dystrophin was produced as a secreted protein by transient transfection of Fv-micro-dystrophin cDNA in COS-7 cells and as a non-secreted protein by permanent transfection in Pichia pastoris. Isolated Fv-micro-dystrophin was shown to be full-length by Western blot analysis. Fv-micro-dystrophin penetrated multiple cell lines in vitro, and it localized to the plasma membrane of a cell line with membrane beta-dystroglycan. In the absence of membrane beta-dystroglycan, it localized to the cytoplasm. Antibody-mediated transduction of micro-dystrophin into muscle cells is a potential therapy for dystrophin-deficient muscular dystrophies.


Scientific Reports | 2015

DNA-dependent targeting of cell nuclei by a lupus autoantibody.

Richard H. Weisbart; Grace Chan; Gwen Jordaan; Philip W. Noble; Yanfeng Liu; Peter M. Glazer; Robert N. Nishimura; James E. Hansen

A nuclear-penetrating lupus anti-DNA autoantibody, 3E10, has been found to inhibit DNA repair and selectively kill certain cancer cells that are highly vulnerable to DNA damage. In addition, a 3E10 single chain variable fragment (scFv) has been developed for use as a delivery vehicle to carry therapeutic cargo proteins into cell nuclei. A greater understanding of the mechanism by which 3E10 penetrates cell nuclei is needed to help determine the scope of its potential therapeutic applications. Here we show that the presence of extracellular DNA significantly enhances the nuclear uptake of 3E10 scFv. In addition, we find that 3E10 scFv preferentially localizes into tumor cell nuclei in vivo, likely due to increased DNA in the local environment released from ischemic and necrotic regions of tumor. These data provide insight into the mechanism of nuclear penetration by 3E10 and demonstrate the potential for use of 3E10 in therapeutic approaches to diseases ranging from malignancy to ischemic conditions such as stroke.


Journal of Biological Chemistry | 2010

BRAF drives synovial fibroblast transformation in rheumatoid arthritis.

Richard H. Weisbart; Grace Chan; Emil Heinze; Rachel Mory; Robert N. Nishimura; Keith K. Colburn

Synovial fibroblasts destroy articular cartilage and bone in rheumatoid arthritis, but the mechanism of fibroblast transformation remains elusive. Because gain-of-function mutations of BRAF can transform fibroblasts, we examined BRAF in rheumatoid synovial fibroblasts. The strong gain-of-function mutation, V600R, of BRAF found in melanomas and other cancers was identified in first passage synovial fibroblasts from two of nine rheumatoid arthritis patients and confirmed by restriction site mapping. BRAF-specific siRNA inhibited proliferation of synovial fibroblasts with V600R mutations. A BRAF aberrant splice variant with an intact kinase domain and partial loss of the N-terminal autoinhibitory domain was identified in fibroblasts from an additional patient, and fibroblast proliferation was inhibited by BRAF-specific siRNA. Our finding is the first to establish mechanisms for fibroblast transformation responsible for destruction of articular cartilage and bone in rheumatoid arthritis and establishes a new target for therapeutic intervention.


Cancer Research | 2015

Optimizing a Lupus Autoantibody for Targeted Cancer Therapy

Philip W. Noble; Grace Chan; Melissa R. Young; Richard H. Weisbart; James E. Hansen

The specificity of binding by antibodies to target antigens is a compelling advantage to antibody-based cancer therapy, but most antibodies cannot penetrate cells to affect intracellular processes. Select lupus autoantibodies penetrate into cell nuclei, and the potential for application of these antibodies in cancer therapy is an emerging concept. Here, we show that a divalent lupus anti-DNA autoantibody fragment with enhancing mutations that increase its ability to penetrate cell nuclei and bind DNA causes accumulation of DNA double-strand breaks in and is highly and selectively toxic to cancer cells and tumors with defective homology-directed repair of DNA double-strand breaks. These findings provide proof of principle for the use of optimized lupus autoantibodies in targeted cancer therapy.

Collaboration


Dive into the Grace Chan's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rika Wakelin

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joseph Gera

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge