Network


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

Hotspot


Dive into the research topics where Victoria Liu is active.

Publication


Featured researches published by Victoria Liu.


Journal of Immunology | 2007

Tumor Evasion of the Immune System by Converting CD4+CD25− T Cells into CD4+CD25+ T Regulatory Cells: Role of Tumor-Derived TGF-β

Victoria Liu; Larry Wong; Thomas L. Jang; Ali H. Shah; Irwin Park; Ximing J. Yang; Qiang Zhang; Scott Lonning; Beverly A. Teicher; Chung Lee

CD4+CD25+ T regulatory (Treg) cells were initially described for their ability to suppress autoimmune diseases in animal models. An emerging interest is the potential role of Treg cells in cancer development and progression because they have been shown to suppress antitumor immunity. In this study, CD4+CD25− T cells cultured in conditioned medium (CM) derived from tumor cells, RENCA or TRAMP-C2, possess similar characteristics as those of naturally occurring Treg cells, including expression of Foxp3, a crucial transcription factor of Treg cells, production of low levels of IL-2, high levels of IL-10 and TGF-β, and the ability to suppress CD4+CD25− T cell proliferation. Further investigation revealed a critical role of tumor-derived TGF-β in converting CD4+CD25− T cells into Treg cells because a neutralizing Ab against TGF-β, 1D11, completely abrogated the induction of Treg cells. CM from a nontumorigenic cell line, NRP-152, or irradiated tumor cells did not convert CD4+CD25− T cells to Treg cells because they produce low levels of TGF-β in CM. Finally, we observed a reduced tumor burden in animals receiving 1D11. The reduction in tumor burden correlated with a decrease in tumor-derived TGF-β. Treatment of 1D11 also reduced the conversion of CD4+ T cells into Treg cells and subsequent Treg cell-mediated suppression of antitumor immunity. In summary, we have demonstrated that tumor cells directly convert CD4+CD25− T cells to Treg cells through production of high levels of TGF-β, suggesting a possible mechanism through which tumor cells evade the immune system.


Cancer Research | 2005

Adoptive Transfer of Tumor-Reactive Transforming Growth Factor-β–Insensitive CD8+ T Cells: Eradication of Autologous Mouse Prostate Cancer

Qiang Zhang; Ximing J. Yang; Michael Pins; Borko Javonovic; Timothy M. Kuzel; Seong-Jin Kim; Luk Van Parijs; Norman M. Greenberg; Victoria Liu; Yinglu Guo; Chung Lee

Transforming growth factor (TGF)-beta is a potent immunosuppressant. Overproduction of TGF-beta by tumor cells may lead to tumor evasion from the host immune surveillance and tumor progression. The present study was conducted to develop a treatment strategy through adoptive transfer of tumor-reactive TGF-beta-insensitive CD8+ T cells. The mouse TRAMP-C2 prostate cancer cells produced large amounts of TGF-beta1 and were used as an experimental model. C57BL/6 mice were primed with irradiated TRAMP-C2 cells. CD8+ T cells were isolated from the spleen of primed animals, were expanded ex vivo, and were rendered TGF-beta insensitive by infecting with a retrovirus containing dominant-negative TGF-beta type II receptor. Results of in vitro cytotoxic assay revealed that these CD8+ T cells showed a specific and robust tumor-killing activity against TRAMP-C2 cells but were ineffective against an irrelevant tumor line, B16-F10. To determine the in vivo antitumor activity, recipient mice were challenged with a single injection of TRAMP-C2 cells for a period up to 21 days before adoptive transfer of CD8+ T cells was done. Pulmonary metastasis was either eliminated or significantly reduced in the group receiving adoptive transfer of tumor-reactive TGF-beta-insensitive CD8+ T cells. Results of immunofluorescent studies showed that only tumor-reactive TGF-beta-insensitive CD8+ T cells were able to infiltrate into the tumor and mediate apoptosis in tumor cells. Furthermore, transferred tumor-reactive TGF-beta-insensitive CD8+ T cells were able to persist in tumor-bearing hosts but declined in tumor-free animals. These results suggest that adoptive transfer of tumor-reactive TGF-beta-insensitive CD8+ T cells may warrant consideration for cancer therapy.


Journal of Immunology | 2008

Cutting Edge: TGF-β-Induced Expression of Foxp3 in T cells Is Mediated through Inactivation of ERK

Xunrong Luo; Qiang Zhang; Victoria Liu; Zhenbiao Xia; Kathryn L. Pothoven; Chung Lee

The peripheral induction of T regulatory cells can be accomplished by TGF-β through an epigenetic regulation leading to the expression of Foxp3. However, the exact mechanism of such a TGF-β-mediated action remains unclear. In the current study, we found that TGF-β treatment of CD4+CD25− T cells during T cell activation led to a transient inhibition of the phosphorylation of ERK followed by the induction of Foxp3 expression in these cells. Direct treatment with a specific ERK inhibitor, UO126, during CD4+CD25− T cell activation also induced Foxp3 expression and conferred a suppressive function to the induced Foxp3+ T cells. Furthermore, treatment of T cells with either TGF-β or UO126 significantly down-regulated the expression of DNMTs, a reaction normally elicited by demethylation agents, such as 5-Aza-2′-deoxycytidine. These results indicate that the epigenetic regulation of TGF-β-induced expression of Foxp3 may be mediated through the inactivation of ERK.


Cancer Research | 2007

Effect of Dominant Negative Transforming Growth Factor-β Receptor Type II on Cytotoxic Activity of RAW 264.7, a Murine Macrophage Cell Line

Geun Taek Lee; Jun Hyuk Hong; Cheol Kwak; Jaesung Woo; Victoria Liu; Chung Lee; Isaac Yi Kim

Transforming growth factor-beta (TGF-beta) is a potent suppressor of the immune system. In the present study, we investigated the effect of TGF-beta resistance on a murine macrophage cell line, RAW 264.7, by overexpressing a dominant negative TGF-beta receptor type II (TbetaRIIDN) construct. As expected, TbetaRIIDN-expressing RAW cells, designated as RAW-TbetaRIIDN, were resistant to TGF-beta signaling. When these cells were cocultured with the murine renal cell carcinoma cell line, Renca, a dramatic increase in apoptosis of Renca cells was observed. Simultaneously, elevated levels of inducible nitric oxide synthase (iNOS) and tumor necrosis factor-alpha (TNF-alpha) in association with IFN-gamma were detected in RAW-TbetaRIIDN cells. When the effects of TNF-alpha and iNOS were neutralized through the use of neutralizing antibody and N(G)-methyl-L-arginine, respectively, the enhanced cytotoxicity of TbetaRIIDN-RAW cells was partially reversed. Taken together, these results show that TGF-beta-resistant RAW 264.7 murine macrophage cells have increased cytotoxic activity that is in part mediated by iNOS and TNF-alpha.


Endocrinology | 2009

17β-Estradiol at Low Concentrations Acts through Distinct Pathways in Normal Versus Benign Prostatic Hyperplasia-Derived Prostate Stromal Cells

Irwin Park; Qiang Zhang; Victoria Liu; James M. Kozlowski; Ju Zhang; Chung Lee

The aim of this study was to identify differential responses to low concentrations of 17beta-estradiol (E2) in primary stromal cell cultures derived from either normal organ donors or benign prostatic hyperplasia or hypertrophy (BPH) specimens. Furthermore, we sought to identify the potential mechanism of E2 action in these cell types, through either a genomic or nongenomic mechanism. We initially treated stromal cells derived from five normal prostates or five BPH specimens with low concentrations of E2 (0.001-1.0 nM) and analyzed their growth response. To determine whether genomic or nongenomic pathways were involved, we performed studies using specific estrogen receptor antagonists to confirm transcriptional activity or MAPK inhibitors to confirm the involvement of rapid signaling. Results of these studies revealed a fundamental difference in the mechanism of the response to E2. In normal cells, we found that a nongenomic, rapid E2 signaling pathway is predominantly involved, mediated by G protein-coupled receptor-30 and the subsequent activation of ERK1/2. In BPH-derived prostate stromal cells, a genomic pathway is predominantly involved because the addition of ICI 182780 was sufficient to abrogate any estrogenic effects. In conclusion, prostate stromal cells respond to far lower concentrations of E2 than previously recognized or examined, and this response is mediated through two distinct mechanisms, depending on its origin. This may provide the basis for new insights into the causes of, and possible treatments for, BPH.


Urology | 2008

Treatment of Transforming Growth Factor-Beta-Insensitive Mouse Renca Tumor by Transforming Growth Factor-Beta Elimination

Kent T. Perry; Larry Wong; Victoria Liu; Irwin Park; Qiang Zhang; Varun Rejen; Xuemei Huang; Norm D. Smith; Borko Jovanovic; Scott Lonning; Beverly A. Teicher; Chung Lee

OBJECTIVES The mouse renal cell carcinoma line, Renca, is insensitive to transforming growth factor-beta (TGF-beta) in vitro. The present study was conducted to determine whether removal of TGF-beta from these tumor cells would inhibit tumor progression in vivo. METHODS TGF-beta elimination was accomplished either by administration of neutralizing TGF-beta antibody into mice receiving intravenous injection of Renca tumor cells or infection of TGF-beta antisense expression vector into these tumor cells before subcutaneous injection into recipient mice. RESULTS Although a low dose of TGF-beta antibody (5 mg/kg every 3 days) was without any effect, a high dose of TGF-beta antibody (50 mg/kg every 3 days), administered to recipient mice, resulted in a significant reduction in lung metastasis and was accompanied by increased apoptosis in the tumor cells. When the tumor cells were transfected with a TGF-beta1 antisense expressing vector, a significant reduction occurred in the tumor incidence, as well as the tumor burden. However, in nude mice, cells with reduced TGF-beta1 production grew almost as well as did the unmodified Renca cells, suggesting that the hosts immune system might play an antitumor role. CONCLUSIONS These results indicate that progression of Renca tumor can be inhibited by eliminating TGF-beta from the tumor cells. Our results also suggest that, although insensitive to TGF-beta under in vitro conditions, Renca tumors could be inhibited by TGF-beta removal through the systemic host environment.


Archive | 2007

Role of Transforming Growth Factor-β in Immunotherapy of Prostate Cancer

Chung Lee; Ali H. Shah; Victoria Liu; Irwin Park; Larry Wong; Xuemei Huang; Lijun Huang; Tim Fermin; Tom Jang; Som Kundu; Vivian Zhou; Qiang Zhang; Norm D. Smith

In this chapter, we introduce a novel concept for an effective treatment of advanced prostate cancer, using our understanding of transforming growth factor (TGF)-β signaling. TGF-β plays an important role in prostate cancer development and progression. Two characteristic features of TGF-β signaling in aggressive prostate cancer are a reduced sensitivity to TGF-β and an overproduction of TGF-β. A reduced sensitivity to TGF-β removes the inhibitory effect of TGF-β and provides a growth advantage for the cancer cells. An overproduction of TGF-β endows cancer cells the ability to metastasize, to enhance angiogenesis, and to evade host’s immune surveillance program, leading to tumor progression and metastasis. TGF-β is a potent tumor-induced immunosuppression. A therapeutic strategy rending host immune cells insensitive to TGF-β should offer an effective approach to eradicate advanced prostate cancer.


Archive | 2008

Adoptive Transfer of Tumor Reactive TGF-β Insensitive CD8+ T-cells for Cancer Therapy

Chung Lee; Ali H. Shah; Victoria Liu; Irwin Park; Larry Wong; Xuemei Huang; Lijun Huang; Vivian Zhou; Terry R. Medler; Shilajit Kundu; Qiang Zhang; Norm D. Smith

Tumor immunology is characterized by an insufficient immunosurveillance, as most tumors are able to evade host’s immune surveillance program. One of the most profound tumor-derived immune suppressive factors has been TGF-β. Based on this understanding, we have tested adoptive transfer of Indeed, our experimental findings have confirmed this impression. These findings have allowed us to introduce the concept of an antitumor immune response cycle. The postulated cycle consists of three major participants, which are the transferred cytotoxic T-cells, the autologous tumor cells, and the host’s own immune system. The present chapter provides the necessary background, the experimental findings, and the clinical implications of the proposed antitumor immune response cycle. Based on this postulated cycle, we will be able to devise a successful antitumor strategy for advanced cancers.


The Prostate | 2008

The toll-like receptor pathway: a novel mechanism of infection-induced carcinogenesis of prostate epithelial cells.

Shilajit Kundu; Chung Lee; Benjamin K. Billips; Geoffrey M. Habermacher; Qiang Zhang; Victoria Liu; Larry Wong; David J. Klumpp; Praveen Thumbikat


Cancer Research | 2002

Suppression of tumor metastasis by blockade of transforming growth factor β signaling in bone marrow cells through a retroviral-mediated gene therapy in mice

Ali H. Shah; William B. Tabayoyong; Shilajit Kundu; Seong-Jin Kim; Luk Van Parijs; Victoria Liu; Eugene Kwon; Norman M. Greenberg; Chung Lee

Collaboration


Dive into the Victoria Liu's collaboration.

Top Co-Authors

Avatar

Chung Lee

Northwestern University

View shared research outputs
Top Co-Authors

Avatar

Qiang Zhang

Northwestern University

View shared research outputs
Top Co-Authors

Avatar

Irwin Park

Northwestern University

View shared research outputs
Top Co-Authors

Avatar

Larry Wong

Northwestern University

View shared research outputs
Top Co-Authors

Avatar

Ali H. Shah

Northwestern University

View shared research outputs
Top Co-Authors

Avatar

Beverly A. Teicher

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Thomas L. Jang

University of Medicine and Dentistry of New Jersey

View shared research outputs
Researchain Logo
Decentralizing Knowledge